epidermal-growth-factor and Cell-Transformation--Neoplastic

epidermal-growth-factor has been researched along with Cell-Transformation--Neoplastic* in 568 studies

Reviews

77 review(s) available for epidermal-growth-factor and Cell-Transformation--Neoplastic

ArticleYear
Human Colorectal Cancer from the Perspective of Mouse Models.
    Genes, 2019, 10-11, Volume: 10, Issue:10

    Colorectal cancer (CRC) is a heterogeneous disease that includes both hereditary and sporadic types of tumors. Tumor initiation and growth is driven by mutational or epigenetic changes that alter the function or expression of multiple genes. The genes predominantly encode components of various intracellular signaling cascades. In this review, we present mouse intestinal cancer models that include alterations in the Wnt, Hippo, p53, epidermal growth factor (EGF), and transforming growth factor β (TGFβ) pathways; models of impaired DNA mismatch repair and chemically induced tumorigenesis are included. Based on their molecular biology characteristics and mutational and epigenetic status, human colorectal carcinomas were divided into four so-called consensus molecular subtype (CMS) groups. It was shown subsequently that the CMS classification system could be applied to various cell lines derived from intestinal tumors and tumor-derived organoids. Although the CMS system facilitates characterization of human CRC, individual mouse models were not assigned to some of the CMS groups. Thus, we also indicate the possible assignment of described animal models to the CMS group. This might be helpful for selection of a suitable mouse strain to study a particular type of CRC.

    Topics: Animals; Carcinogenesis; Cell Transformation, Neoplastic; Colonic Neoplasms; Colorectal Neoplasms; Disease Models, Animal; DNA Mismatch Repair; Epidermal Growth Factor; Gene Expression Regulation, Neoplastic; Genes, p53; Hippo Signaling Pathway; Humans; Mice; Protein Serine-Threonine Kinases; Transforming Growth Factor beta; Wnt Signaling Pathway

2019
Epidermal growth factor signaling in transformed cells.
    International review of cell and molecular biology, 2015, Volume: 314

    Members of the epidermal growth factor receptor (EGFR/ErbB) family play a critical role in normal cell growth and development. However, many ErbB family members, especially EGFR, are aberrantly expressed or deregulated in tumors and are thought to play crucial roles in cancer development and metastatic progression. In this chapter, we provide an overview of key mechanisms contributing to aberrant EGFR/ErbB signaling in transformed cells, which results in many phenotypic changes associated with the earliest stages of tumor formation, including several hallmarks of epithelial-mesenchymal transition (EMT). These changes often occur through interaction with other major signaling pathways important to tumor progression, causing a multitude of transcriptional changes that ultimately impact cell morphology, proliferation, and adhesion, all of which are crucial for tumor progression. The resulting mesh of signaling networks will need to be taken into account as new regimens are designed for targeting EGFR for therapeutic intervention. As new insights are gained into the molecular mechanisms of cross talk between EGFR signaling and other signaling pathways, including their roles in therapeutic resistance to anti-EGFR therapies, a continual reassessment of clinical therapeutic regimes and strategies will be required. Understanding the consequences and complexity of EGF signaling and how it relates to tumor progression is critical for the development of clinical compounds and establishing clinical protocols for the treatment of cancer.

    Topics: Animals; Cell Proliferation; Cell Transformation, Neoplastic; Epidermal Growth Factor; ErbB Receptors; Humans; Neoplasms; Signal Transduction

2015
Rac signaling in breast cancer: a tale of GEFs and GAPs.
    Cellular signalling, 2012, Volume: 24, Issue:2

    Rac GTPases, small G-proteins widely implicated in tumorigenesis and metastasis, transduce signals from tyrosine-kinase, G-protein-coupled receptors (GPCRs), and integrins, and control a number of essential cellular functions including motility, adhesion, and proliferation. Deregulation of Rac signaling in cancer is generally a consequence of enhanced upstream inputs from tyrosine-kinase receptors, PI3K or Guanine nucleotide Exchange Factors (GEFs), or reduced Rac inactivation by GTPase Activating Proteins (GAPs). In breast cancer cells Rac1 is a downstream effector of ErbB receptors and mediates migratory responses by ErbB1/EGFR ligands such as EGF or TGFα and ErbB3 ligands such as heregulins. Recent advances in the field led to the identification of the Rac-GEF P-Rex1 as an essential mediator of Rac1 responses in breast cancer cells. P-Rex1 is activated by the PI3K product PIP3 and Gβγ subunits, and integrates signals from ErbB receptors and GPCRs. Most notably, P-Rex1 is highly overexpressed in human luminal breast tumors, particularly those expressing ErbB2 and estrogen receptor (ER). The P-Rex1/Rac signaling pathway may represent an attractive target for breast cancer therapy.

    Topics: Animals; Breast Neoplasms; Cell Adhesion; Cell Communication; Cell Movement; Cell Proliferation; Cell Transformation, Neoplastic; Epidermal Growth Factor; ErbB Receptors; Female; GTPase-Activating Proteins; Guanine Nucleotide Exchange Factors; Humans; Mice; Neuregulins; Phosphatidylinositol 3-Kinases; rac GTP-Binding Proteins; Receptors, G-Protein-Coupled; Signal Transduction; Transforming Growth Factor alpha

2012
Epithelial and mesenchymal phenotypic switchings modulate cell motility in metastasis.
    Frontiers in bioscience (Landmark edition), 2011, 01-01, Volume: 16, Issue:3

    The most ominous stage of cancer progression is metastasis, or the dissemination of carcinoma cells from the primary site into distant organs. Metastases are often resistant to current extirpative therapies and even the newest biological agents cure only a small subset of patients. Therefore a greater understanding of tumor biology that integrates properties intrinsic to carcinomas with tissue environmental modulators of behavior is needed. In no aspect of tumor progression is this more evident than the acquisition of cell motility that is critical for both escape from the primary tumor and colonization. In this overview, we discuss how this behavior is modified by carcinoma cell phenotypic plasticity that is evidenced by reversible switching between epithelial and mesenchymal phenotypes. The presence or absence of intercellular adhesions mediate these switches and dictate the receptivity towards signals from the extracellular milieu. These signals, which include soluble growth factors, cytokines, and extracellular matrix embedded with matrikines and matricryptines will be discussed in depth. Finally, we will describe a new mode of discerning the balance between epithelioid and mesenchymal movement.

    Topics: Cadherins; Cell Adhesion; Cell Movement; Cell Transformation, Neoplastic; Cytokines; Desmosomes; Epidermal Growth Factor; Epithelial-Mesenchymal Transition; Extracellular Matrix Proteins; Gap Junctions; Hepatocyte Growth Factor; Humans; Insulin-Like Growth Factor I; Integrins; Neoplasm Metastasis; Neoplasms; Phenotype; Signal Transduction; Tight Junctions; Transforming Growth Factor beta; Tumor Necrosis Factor-alpha

2011
Activated epidermal growth factor receptor in ovarian cancer.
    Cancer treatment and research, 2009, Volume: 149

    Topics: Carcinoma; Cell Line, Tumor; Cell Survival; Cell Transformation, Neoplastic; Drug Resistance, Neoplasm; Enzyme Activation; Epidermal Growth Factor; ErbB Receptors; Female; Gene Expression Regulation, Neoplastic; Genes, erbB-1; Humans; Ligands; Neoplasm Metastasis; Neoplasm Proteins; Ovarian Neoplasms; Prognosis

2009
Phosphoproteomics, oncogenic signaling and cancer research.
    Proteomics, 2008, Volume: 8, Issue:21

    The past 5 years have seen an explosion of phosphoproteomics methods development. In this review, using epidermal growth-factor signaling as a model, we will discuss how phosphoproteomics, along with bioinformatics and computational modeling, have impacted key aspects of oncogenic signaling such as in the temporal fine mapping of phosphorylation events, and the identification of novel tyrosine kinase substrates and phosphorylation sites. We submit that the next decade will see considerable exploitation of phosphoproteomics in cancer research. Such a phenomenon is already happening as exemplified by its use in promoting the understanding of the molecular etiology of cancer and target-directed therapeutics.

    Topics: Animals; Cell Transformation, Neoplastic; Computational Biology; Computer Simulation; Epidermal Growth Factor; ErbB Receptors; Humans; Neoplasms; Oncogenes; Phosphoproteins; Phosphorylation; Proteomics; Signal Transduction

2008
Ba/F3 cells and their use in kinase drug discovery.
    Current opinion in oncology, 2007, Volume: 19, Issue:1

    Due to their ability to function as dominant oncogenes, protein kinases have become favored targets in the quest for 'molecularly-targeted' cancer chemotherapeutics. The discovery of a large number of cancer-associated mutations in the kinome, and the progress in developing specific small-molecule kinase inhibitors has increased the need for accurate, reproducible, and efficient kinase activity-dependent cellular assay systems.. Ba/F3, a murine interleukin-3 dependent pro-B cell line is increasingly popular as a model system for assessing both the potency and downstream signaling of kinase oncogenes, and the ability of small-molecule kinase inhibitors to block kinase activity. Facilitated by their growth properties, Ba/F3 cells have recently been adapted to high-throughput assay formats for compound profiling. Further, several published approaches show promise in predicting resistance to small-molecule kinase inhibitors elicited by point mutations interfering with inhibitor binding.. Ba/F3 cells are an increasingly popular tool in kinase drug discovery. The ability to test the transforming capacity of newly identified kinase mutations, and to profile drug candidates and compound libraries in high-throughput fashion, combined with the use of Ba/F3 cells to predict clinical resistance will greatly facilitate developments in this field.

    Topics: Animals; Antineoplastic Agents; Cell Line; Cell Transformation, Neoplastic; Drug Design; Drug Resistance, Neoplasm; Epidermal Growth Factor; ErbB Receptors; Mice; Protein Kinase Inhibitors; Signal Transduction

2007
Interplay of distinct growth factors during epithelial mesenchymal transition of cancer progenitor cells and molecular targeting as novel cancer therapies.
    Annals of oncology : official journal of the European Society for Medical Oncology, 2007, Volume: 18, Issue:10

    In this review, we describe the critical functions assumed by the interplay of epidermal growth factor, hedgehog, Wnt/beta-catenin, tumor growth factor-beta and integrin signaling cascades in tumorigenic and migrating cancer progenitor cells and activated stromal cells during carcinogenesis. These growth factors provide an important role for the sustained growth and survival of tumorigenic cancer progenitor cells and their progeny by up-regulating numerous mitotic and antiapoptotic signaling cascades. Furthermore, these potent morphogens may cooperate for inducing the molecular events associated with the epithelial-mesenchymal program in cancer cells including the alterations in epithelial cell shape and motility through the dissociation of intercellular adherens junctions. Of therapeutic interest, new strategies for the development of more effective clinical treatments against the locally aggressive and invasive cancers based on the molecular targeting of deregulated signaling elements in tumorigenic and migrating cancer cells and their local microenvironment are also described.

    Topics: Antineoplastic Combined Chemotherapy Protocols; beta Catenin; Cell Proliferation; Cell Survival; Cell Transformation, Neoplastic; Cytokines; Epidermal Growth Factor; Epithelial Cells; ErbB Receptors; Growth Substances; Hedgehog Proteins; Humans; Integrins; Mesoderm; Neoplasm Metastasis; Neoplasms; Neoplastic Stem Cells; Signal Transduction

2007
Sequestration and segregation of receptor kinases in epithelial cells: implications for ErbB2 oncogenesis.
    Science's STKE : signal transduction knowledge environment, 2007, Apr-10, Volume: 2007, Issue:381

    Cell behaviors are regulated by signaling pathways triggered by the activation of cell surface receptors. A key aspect of receptor signaling is the location of these receptors relative to their ligands and to other receptors, particularly in epithelia, whose cells are polarized by tight junction barriers into apical and basolateral membrane regions. In polarized epithelia, the co-receptor ErbB2 is often localized to the apical surface by its intramembrane ligand Muc4, thus segregating it from its partner ErbB3, which is sequestered at the lateral surface, co-localized with cadherin junctions. The ErbB2-ErbB3 receptor heterodimer, when activated, is a potent stimulator of cell proliferation; thus, the segregation mechanism helps maintain these cells in a differentiated state. Similarly, epidermal growth factor, the ligand for ErbB1, which is present in the apical fluid of some epithelia, is segregated from its receptor by the tight junction barrier. Loss of cell polarity and the tight junction barrier facilitates the interaction of ErbB2 with the hemidesmosome integrin alpha(6)beta(4). This integrin acts as a docking site for signaling pathways to promote cell proliferation and further disruption of cell junctions. The ultimate dissolution of tight junctions may result from activation of transforming growth factor-beta receptors, one subunit of which is directly associated with the junction. This activation triggers degradation of critical tight junction components. These sequestration and segregation phenomena provide a model by which overexpression of the ErbB2 receptor kinase may trigger oncogenesis by initiating junction breakdown. Equally important, these mechanisms may act as a sensor for epithelial damage that can activate repair mechanisms.

    Topics: Animals; Cell Differentiation; Cell Polarity; Cell Proliferation; Cell Transformation, Neoplastic; Epidermal Growth Factor; Epithelial Cells; Humans; Integrins; Ligands; Mucin-4; Mucins; Receptor, ErbB-2; Receptors, Transforming Growth Factor beta; Signal Transduction; Tight Junctions

2007
Rational bases for the development of EGFR inhibitors for cancer treatment.
    The international journal of biochemistry & cell biology, 2007, Volume: 39, Issue:7-8

    Growth factor receptors and their ligands not only regulate normal cell processes but have been also identified as key regulators of human cancer formation. The epidermal growth factor receptor (EGFR/ErbB1/HER1) belongs to the ErbB/HER-family of tyrosine kinase receptors (RTKs). These trans-membrane proteins are activated following binding with peptide growth factors of the EGF-family of proteins. Several evidences suggest that cooperation of multiple ErbB receptors and ligands is required for the induction of cell transformation. In this respect, EGFR, upon activation, sustains a complex and redundant network of signal transduction pathways with the contribution of other trans-membrane receptors. EGFR has been found to be expressed and altered in a variety of malignancies and clearly it plays a significant role in tumor development and progression, including cell proliferation, regulation of apoptotic cell death, angiogenesis and metastatic spread. Moreover, amplification of the EGFR gene and mutations in the EGFR tyrosine kinase domain have been recently reported in human carcinomas. As a result, investigators have developed approaches to inhibit the effects of EGFR activation, with the aim of blocking tumor growth and invasion. A number of agents targeting EGFR, including specific antibodies directed against its ligand-binding domain and small molecules inhibiting its tyrosine kinase activity are either in clinical trials or are already approved for clinical treatment. This article reviews the EGFR role in carcinogenesis and tumor progression as rational bases for the development of specific therapeutic inhibitors.

    Topics: Antibodies, Monoclonal; Antineoplastic Agents; Cell Transformation, Neoplastic; Epidermal Growth Factor; ErbB Receptors; Humans; Mutation; Neoplasms; Protein-Tyrosine Kinases; Signal Transduction

2007
Sprouty proteins: multifaceted negative-feedback regulators of receptor tyrosine kinase signaling.
    Trends in cell biology, 2006, Volume: 16, Issue:1

    Receptor tyrosine kinases (RTKs) control a wide variety of processes in multicellular organisms, including proliferation, differentiation, migration and survival. Their activity is tightly controlled through the coordinated action of both positive and negative regulators that function at multiple levels of the signal transduction cascade, and at different time points within the growth-factor-induced response. When this process goes awry, the outcome can be developmental defects and malignancy. Sprouty (Spry) proteins represent a major class of ligand-inducible inhibitors of RTK-dependent signaling pathways. New biochemical and genetic evidence indicates specific roles of the Spry genes in development and multiple modes of action of the Spry proteins in regulation of the RTK-induced response.

    Topics: Amino Acid Sequence; Animals; Cell Transformation, Neoplastic; Epidermal Growth Factor; Feedback, Physiological; Gene Expression Regulation, Developmental; Genes, Tumor Suppressor; Humans; Membrane Proteins; Receptor Protein-Tyrosine Kinases; Signal Transduction

2006
Epidermal growth factor receptor (EGFR) signaling in cancer.
    Gene, 2006, Jan-17, Volume: 366, Issue:1

    The epidermal growth factor receptor (EGFR) belongs to the ErbB family of receptor tyrosine kinases (RTK). These trans-membrane proteins are activated following binding with peptide growth factors of the EGF-family of proteins. Evidence suggests that the EGFR is involved in the pathogenesis and progression of different carcinoma types. The EGFR and EGF-like peptides are often over-expressed in human carcinomas, and in vivo and in vitro studies have shown that these proteins are able to induce cell transformation. Amplification of the EGFR gene and mutations of the EGFR tyrosine kinase domain have been recently demonstrated to occur in carcinoma patients. Interestingly, both these genetic alterations of the EGFR are correlated with high probability to respond to anti-EGFR agents. However, ErbB proteins and their ligands form a complex system in which the interactions occurring between receptors and ligands affect the type and the duration of the intracellular signals that derive from receptor activation. In fact, proteins of the ErbB family form either homo- or hetero-dimers following ligand binding, each dimer showing different affinity for ligands and different signaling properties. In this regard, evidence suggests that cooperation of multiple ErbB receptors and cognate ligands is necessary to induce cell transformation. In particular, the growth and the survival of carcinoma cells appear to be sustained by a network of receptors/ligands of the ErbB family. This phenomenon is also important for therapeutic approaches, since the response to anti-EGFR agents might depend on the total level of expression of ErbB receptors and ligands in tumor cells.

    Topics: Animals; Carcinoma; Cell Transformation, Neoplastic; Dimerization; Enzyme Inhibitors; Epidermal Growth Factor; ErbB Receptors; Gene Amplification; Gene Expression Regulation, Neoplastic; Humans; Mutation; Protein Structure, Tertiary; Signal Transduction

2006
EGFR as a target: rationale for therapy.
    Seminars in oncology nursing, 2006, Volume: 22, Issue:1 Suppl 1

    To review the biology of the EGFR, its structure, and the associated signal transduction pathways. To provide an overview of the role of EGFR in normal physiology and the pathophysiology of malignancy. Current anti-EGFR treatments are also discussed.. Research articles.. EGFR is a valid target in the treatment of solid tumors. EGFR abnormalities and dysfunction are involved in various aspects of carcinogenesis and tumor progression, and EGFR is overexpressed in several tumor types. The development of anti-EGFR therapies represents an important advance in cancer therapy.. Anti-EGFR therapy is currently available in the clinical setting. Nurses involved in the care of patients with cancer can benefit from an increased understanding of the normal and abnormal function of EGFR in the body and the mechanisms by which anti-EGFR therapies act.

    Topics: Antibodies, Monoclonal; Antineoplastic Agents; Cell Transformation, Neoplastic; Epidermal Growth Factor; ErbB Receptors; Humans; Neoplasms; Oncology Nursing; Protein Kinase Inhibitors; Signal Transduction

2006
[Mechanisms of metastasis and molecular markers of malignant tumor progression. I. Colorectal cancer].
    Postepy higieny i medycyny doswiadczalnej (Online), 2006, Volume: 60

    The ability of neoplastic cells to dissemination from a primary tumor to lymphatic nodes and to adjacent and distant tissues and organs is an inseparable feature of malignant tumors and the main cause of failure in their treatment. Metastasis formation is a multistage process which includes proteolysis, the motility and migration of cells, proliferation, and neoangiogenesis. In the first step, the cells released from the primary tumor have to penetrate to the blood or lymphatic vessels (intravasation), the road which dissemination follows. Circulating cells can then migrate through the walls of vessels to surrounding tissues (extravasation) where they settle, proliferate, and induce angiogenesis, creating metastases. Indispensable in the process of intra- and extravasation is the activation of proteolytic enzymes capable of degrading the extracellular matrix (ECM) surrounding the endothelium or creating the basement membrane of epithelial tissue in different organs. In this stage, the activation of proteolytic enzymes, such as proteinases of the plasmin system, serine proteinases, and matrix metalloproteinases (MMPs), is necessary. Simultaneously, changes occur in the expression of many superficial glycoproteins and factors responsible for cell adhesion (integrins) and intercellular communication (cadherins). Neoangiogenesis is connected with the expression of many markers of this process, among them vascular endothelial growth factor (VEGF), endoglin (CD105), a transmembranous glycoprotein which is a component of the receptor for transforming growth factor beta (TGFbeta), as well as neuropilin (NRP), the co-receptor for VEGF. Conventionally, the prognosis of neoplastic disease and its treatment are based mainly on exact clinical and histopathological staging. This prognosis could, however, be improved by measuring the molecular and cellular markers which play key roles in tumor progression. Understanding the cellular processes responsible for tumor dissemination can be useful not only in the diagnosis and prognosis of treatment results, but also in developing targeted drugs, selectively directed towards those factors responsible for tumor invasiveness, as well as in creating new therapeutic strategies permitting the use of such drugs. In the present review the authors concentrate mainly on one tumor type, colorectal carcinoma, in which distant metastases, predominantly to the liver, are the main cause of failure, in spite of surgical curing of the primar

    Topics: Biomarkers, Tumor; Carcinoma; Cell Movement; Cell Transformation, Neoplastic; Colorectal Neoplasms; Disease Progression; Epidermal Growth Factor; ErbB Receptors; Humans; Lymphatic Metastasis; Neoplasm Invasiveness; Neovascularization, Pathologic

2006
VAV1: a new target in pancreatic cancer?
    Cancer biology & therapy, 2005, Volume: 4, Issue:5

    Pancreatic ductal adenocarcinoma (PDA) is arguably the most lethal malignancy in the United States. Despite the identification of many molecular alterations in PDA, this information has not translated into effective therapeutic strategies to date. A recent report in Cancer Cell (Fernandez-Zapico et al, Cancer Cell 2005, 7:39-49) reveals an unexpected role for the hematopoietic-specific RhoGEF VAV1 in pancreatic tumorigenesis, where ectopic expression of VAV1 as a result of promoter demethylation was identified in the majority of established cell lines and PDA tissue samples. Importantly, VAV1 expression was functionally required for optimal proliferation, transformation and survival of pancreatic cancer cell lines. This study provides the first evidence of VAV1 promoter demethylation as an event in cancer progression, suggesting that aberrant signaling pathways driven by VAV1 are potential therapeutic targets in PDA.

    Topics: Animals; Carcinoma, Pancreatic Ductal; Cell Line, Tumor; Cell Proliferation; Cell Survival; Cell Transformation, Neoplastic; DNA Methylation; Enzyme Activation; Epidermal Growth Factor; Humans; Mice; Pancreatic Neoplasms; Promoter Regions, Genetic; Proto-Oncogene Proteins c-vav; rac1 GTP-Binding Protein; RNA Interference; RNA, Messenger; Signal Transduction

2005
Neural stem cells and the origin of gliomas.
    The New England journal of medicine, 2005, Aug-25, Volume: 353, Issue:8

    Topics: Brain Neoplasms; Cell Differentiation; Cell Lineage; Cell Movement; Cell Proliferation; Cell Transformation, Neoplastic; Epidermal Growth Factor; Glioma; Humans; Neurons; Patched Receptors; Receptors, Cell Surface; Signal Transduction; Stem Cells; Transcription Factors; Zinc Finger Protein GLI1

2005
Cripto-1: a multifunctional modulator during embryogenesis and oncogenesis.
    Oncogene, 2005, Aug-29, Volume: 24, Issue:37

    It is increasingly evident that genes known to perform critical roles during early embryogenesis, particularly during stem cell renewal, pluripotentiality and survival, are also expressed during the development of cancer. In this regard, oncogenesis may be considered as the recapitulation of embryogenesis in an inappropriate temporal and spatial manner. The epidermal growth factor-Cripto-1/FRL1/cryptic family of proteins consists of extracellular and cell-associated proteins that have been identified in several vertebrate species. During early embryogenesis, epidermal growth factor-Cripto-1/FRL1/cryptic proteins perform an obligatory role as coreceptors for the transforming growth factor-beta subfamily of proteins, which includes Nodal. Cripto-1 has also been shown to function as a ligand through a Nodal/Alk4-independent signaling pathway that involves binding to glypican-1 and the subsequent activation through src of phosphoinositol-3 kinase/Akt and ras/mitogen-activated protein kinase intracellular pathways. Expression of Cripto-1 is increased in several human cancers and its overexpression is associated with the development of mammary tumors in mice. Here, we review the role of Cripto-1 during embryogenesis, cell migration, invasion and angiogenesis and how these activities may relate to cellular transformation and tumorigenesis. We also briefly discuss evidence suggesting that Cripto-1 may be involved in stem cell maintenance.

    Topics: Activin Receptors, Type I; Cell Movement; Cell Transformation, Neoplastic; DNA-Binding Proteins; Embryonic Development; Epidermal Growth Factor; GPI-Linked Proteins; Humans; Intercellular Signaling Peptides and Proteins; MAP Kinase Signaling System; Membrane Glycoproteins; Milk Proteins; Neoplasm Proteins; Neoplasms; Neovascularization, Physiologic; Nodal Protein; Protein Serine-Threonine Kinases; Proteins; Proto-Oncogene Proteins; Proto-Oncogene Proteins c-akt; Signal Transduction; Smad2 Protein; Trans-Activators; Transforming Growth Factor beta; Wnt Proteins

2005
Activator protein 1 (AP-1)- and nuclear factor kappaB (NF-kappaB)-dependent transcriptional events in carcinogenesis.
    Free radical biology & medicine, 2000, May-01, Volume: 28, Issue:9

    Generation of reactive oxygen species (ROS) during metabolic conversion of molecular oxygen imposes a constant threat to aerobic organisms. Other than the cytotoxic effects, many ROS and oxidants are also potent tumor promoters linking oxidative stress to carcinogenesis. Clonal variants of mouse epidermal JB6 cells originally identified for their differential susceptibility to tumor promoters also show differential reduction-oxidation (redox) responses providing a unique model to study oxidative events in tumor promotion. AP-1 and NF-kappaB, inducible by tumor promoters or oxidative stimuli, show differential protein levels or activation in response to tumor promoters in JB6 cells. We further demonstrated that AP-1 and NF-kappaB are both required for maintaining the transformed phenotypes where inhibition of either activity suppresses transformation response in JB6 cells as well as human keratinocytes and transgenic mouse. NF-kappaB proteins or extracellular signal-regulated kinase (ERK) but not AP-1 proteins are shown to be sufficient for conversion from transformation-resistant to transformation-susceptible phenotype. Insofar as oxidative events regulate AP-1 and NF-kappaB transactivation, these oxidative events can be important molecular targets for cancer prevention.

    Topics: Animals; Apoptosis Regulatory Proteins; Carcinogens; Cell Transformation, Neoplastic; Clone Cells; Epidermal Cells; Epidermal Growth Factor; Epidermis; Gene Expression Regulation, Neoplastic; Humans; Mice; Mitogen-Activated Protein Kinase 1; Mitogen-Activated Protein Kinase 3; Mitogen-Activated Protein Kinases; Neoplasm Proteins; NF-kappa B; Oxidation-Reduction; Oxidative Stress; Phenotype; Protein Biosynthesis; Proteins; Reactive Oxygen Species; RNA-Binding Proteins; Signal Transduction; Tetradecanoylphorbol Acetate; Transcription Factor AP-1; Transcription, Genetic; Transcriptional Activation; Transfection; Tumor Necrosis Factor-alpha

2000
Growth control mechanisms in normal and transformed intestinal cells.
    Philosophical transactions of the Royal Society of London. Series B, Biological sciences, 1998, Jun-29, Volume: 353, Issue:1370

    The cells populating the intestinal crypts are part of a dynamic tissue system which involves the self-renewal of stem cells, a commitment to proliferation, lineage-specific differentiation, movement and cell death. Our knowledge of these processes is limited, but even now there are important clues to the nature of the regulatory systems, and these clues are leading to a better understanding of intestinal cancers. Few intestinal-specific markers have been described; however, homeobox genes such as cdx-2 appear to be important for morphogenic events in the intestine. There are several intestinal cell surface proteins such as the A33 antigen which have been used as targets for immunotherapy. Many regulatory cytokines (lymphokines or growth factors) influence intestinal development: enteroglucagon, IL-2, FGF, EGF family members. In conjunction with cell-cell contact and/or ECM, these cytokines lead to specific differentiation signals. Although the tissue distribution of mitogens such as EGF, TGF alpha, amphiregulin, betacellulin, HB-EGF and cripto have been studied in detail, the physiological roles of these proteins have been difficult to determine. Clearly, these mitogens and the corresponding receptors are involved in the maintenance and progression of the tumorigenic state. The interactions between mitogenic, tumour suppressor and oncogenic systems are complex, but the tumorigenic effects of multiple lesions in intestinal carcinomas involve synergistic actions from lesions in these different systems. Together, the truncation of apc and activation of the ras oncogene are sufficient to induce colon tumorigenesis. If we are to improve cancer therapy, it is imperative that we discover the biological significance of these interactions, in particular the effects on cell division, movement and survival.

    Topics: Animals; Cell Division; Cell Transformation, Neoplastic; Epidermal Growth Factor; Genes, APC; Humans; Intestinal Mucosa; Intestinal Neoplasms; Lymphokines; Mice; Promoter Regions, Genetic

1998
Autocrine regulation of keratinocytes: the emerging role of heparin-binding, epidermal growth factor-related growth factors.
    The Journal of investigative dermatology, 1998, Volume: 111, Issue:5

    Although originally conceived as a basis for malignant cell growth, autocrine signaling networks are currently known to be activated during tissue repair and with in vitro cultivation. In human epidermal keratinocytes, activation of the epidermal growth factor receptor by cognate ligands mediates the majority of the autonomous replicative capacity of these cells and is necessary to inhibit differentiation and apoptosis. The importance of heparin-binding growth factors in activation of this receptor was first suggested by the strong anti-proliferative effects of soluble heparin-like molecules on keratinocyte growth. This and related evidence led to the identification of amphiregulin as a major autocrine factor for keratinocytes. The binding of amphiregulin and its homolog, heparin-binding epidermal growth factor-like growth factor, to the receptor is potentially amplified by autoinduction and cross-signaling through epidermal growth factor-related polypeptides and by transmodulation of other ErbB-family receptors (HER-2, -3, -4) in cells expressing these receptors. Heparan sulfate proteoglycans and the tetraspanin family of membrane-associated proteins appear to act as cofactors in amphiregulin-driven mitogenesis mediated by the epidermal growth factor receptor, but amphiregulin's immunolocalization to keratinocyte nuclei and to filopodia may indicate other potentially novel effects. Following from the observation that amphiregulin is overexpressed in lesional psoriatic epidermis, the importance of amphiregulin in hyperproliferative skin diseases has been further supported by recent studies of the targeted expression of a transgene encoding keratin 14 promoter-driven human amphiregulin to the basal epidermis of mice. Founder transgenic mice displayed a morphologic and microscopic cutaneous phenotype that shares characteristics with psoriasis. Pharmacologic regulation of amphiregulin's expression and receptor signaling may eventually prove to be an effective strategy in the treatment of hyperproliferative skin diseases.

    Topics: Amphiregulin; Animals; Cell Transformation, Neoplastic; EGF Family of Proteins; Epidermal Growth Factor; Glycoproteins; Growth Substances; Heparin; Heparin-binding EGF-like Growth Factor; Humans; Intercellular Signaling Peptides and Proteins; Keratinocytes

1998
Factors regulating SCC antigen expression in squamous cell carcinoma of the uterine cervix.
    Tumour biology : the journal of the International Society for Oncodevelopmental Biology and Medicine, 1998, Volume: 19, Issue:6

    Expression of squamous cell carcinoma (SCC) antigen emerged concurrently with squamous formation of the uterine cervix and increased during the neoplastic transformation of the cervical squamous epithelium. SCC antigen expression differed considerably among the histomorphologic cell types of cervical carcinoma. Large cell nonkeratinizing carcinoma contained high levels of the antigen. In contrast, no appreciable expression of SCC antigen was observed in small cell nonkeratinizing carcinoma. The pattern of SCC antigen expression closely coincided with EGF receptor (EGF-R) expression in cervical squamous neoplasia. This suggests that the expression of SCC and EGF-R in cervical carcinoma is related to the differentiation or dedifferentiation processes of the tumor cells. SCC production by CaSki cervical epidermoid carcinoma cells was stimulated by EGF. It seems likely that an autocrine system, in which EGF serves as the signal, may exist in cervical squamous carcinoma. 17beta-estradiol and L-triiodothyronine were found to upregulate EGF-R expression, proliferative potential and SCC production in the CaSki cervical carcinoma cells.

    Topics: Antigens, Neoplasm; Carcinoma, Squamous Cell; Cell Transformation, Neoplastic; Cervix Uteri; Epidermal Growth Factor; ErbB Receptors; Female; Humans; Serpins; Tumor Cells, Cultured; Uterine Cervical Neoplasms

1998
The ErbB signaling network in embryogenesis and oncogenesis: signal diversification through combinatorial ligand-receptor interactions.
    FEBS letters, 1997, Jun-23, Volume: 410, Issue:1

    Ligand-induced activation of receptor tyrosine kinases (RTK) results in the initiation of diverse cellular pathways, including proliferation, differentiation and cell migration. The ErbB family of RTKs represents a model for signal diversification through the formation of homo- and heterodimeric receptor complexes. Each dimeric receptor complex will initiate a distinct signaling pathway by recruiting a different set of Src homology 2- (SH2-) containing effector proteins. Further complexity is added due to the existence of an oncogenic receptor that enhances and stabilizes dimerization but has no ligand (ErbB-2), and a receptor that can recruit novel SH-2-containing proteins, but is itself devoid of kinase activity (ErbB-3). The resulting signaling network has important implications for embryonic development and malignant transformation.

    Topics: Animals; Cell Transformation, Neoplastic; Embryonic and Fetal Development; Epidermal Growth Factor; ErbB Receptors; Humans; Ligands; Proto-Oncogene Proteins; Receptor, ErbB-2; Receptor, ErbB-3; Receptor, ErbB-4; Signal Transduction

1997
Potential interplay between luminal growth factors and increased tight junction permeability in epithelial carcinogenesis.
    The Journal of experimental zoology, 1997, Dec-01, Volume: 279, Issue:5

    Topics: Biological Transport; Carcinogens; Cell Membrane Permeability; Cell Transformation, Neoplastic; DNA Damage; Epidermal Growth Factor; Epithelial Cells; Humans; Tight Junctions

1997
ErbB receptors and EGF-like ligands: cell lineage determination and oncogenesis through combinatorial signaling.
    Journal of mammary gland biology and neoplasia, 1997, Volume: 2, Issue:2

    The ErbB/HER family of transmembrane receptor tyrosine kinases includes four members that bind more than two dozens ligands sharing an epidermal growth factor- (EGF)3 like motif. This family plays a pivotal role in cell lineage determination in a variety of tissues, including mesenchyme-epithelial inductive processes and the interactions between neurons and muscle, glia and Schwann cells. Certain ligands and receptors of the family, especially the ErbB-2 receptor tyrosine kinase, contribute to a relatively virulent phenotype of some human tumors; most notable are carcinomas of secretory epithelia. This large variety of biological signals is generated through a combinatorial network of signal transduction in which different ErbB ligands are apparently capable of stabilizing discrete homo- and heterodimeric receptor complexes, each coupled to a specific set of cytoplasmic signaling proteins. Because each receptor is unique in terms of catalytic activity, cellular routing and transmodulation, the resulting network allows not only an enormous potential for signal diversification but also fine tuning and stringent control of cellular functions. ErbB-2 emerges as a master coordinator of the network, prolonging and amplifying signaling by decelerating the dissociation rates of its heterologous ligands. Thus, the tumorigenic action of ErbB-2 may be attributed to its ability to act as a shared signaling subunit, rather than by functioning as a bone fide receptor.

    Topics: Animals; Cell Transformation, Neoplastic; Epidermal Growth Factor; Genes, erbB-2; Humans; Receptor, ErbB-2; Signal Transduction

1997
EGF-related peptides in the pathophysiology of the mammary gland.
    Journal of mammary gland biology and neoplasia, 1997, Volume: 2, Issue:2

    Normal mammary gland development is the result of complex interactions between a number of hormones and growth factors. Normal and malignant human mammary epithelial cells are able to synthesize and to respond to various different, locally acting growth factors and growth inhibitors. Among these, the EGF-related peptides play an important role in regulating the proliferation and differentiation of human mammary epithelial cells. EGF4 and TGF4 are able to stimulate the lobulo-alveolar development of the mammary gland in vivo as well they are involved in the pathogenesis of human breast cancer. Experimental evidence suggests that estrogen-induced proliferation of breast carcinoma cells is mediated in part by EGF-related growth factors. It has also been demonstrated that activation of certain cellular protooncogenes such as c-Ha-ras in human mammary epithelial cells results in cellular transformation and in an increased production of several EGF-related growth factors such as TGFalpha and amphiregulin. Coexpression of both EGF-related peptides and their own receptors frequently occurs in human breast carcinomas and in human breast cancer cell lines, suggesting that an autocrine pathway of uncontrolled cell growth sustains neoplastic transformation.

    Topics: Animals; Breast; Breast Neoplasms; Cell Transformation, Neoplastic; Epidermal Growth Factor; Epithelial Cells; Female; Humans; Mammary Glands, Animal; Transforming Growth Factor alpha

1997
Epidermal growth factor-related peptides and their receptors in human malignancies.
    Critical reviews in oncology/hematology, 1995, Volume: 19, Issue:3

    Topics: Animals; Brain Neoplasms; Breast Neoplasms; Cell Division; Cell Transformation, Neoplastic; Digestive System Neoplasms; Epidermal Growth Factor; Gene Expression Regulation, Neoplastic; Growth Substances; Head and Neck Neoplasms; Humans; Lung Neoplasms; Melanoma; Neoplasm Proteins; Neoplasms; Neoplasms, Experimental; Oncogenes; Rats; Receptor Protein-Tyrosine Kinases; Receptors, Growth Factor; Signal Transduction; Urogenital Neoplasms

1995
Transforming growth factor alpha.
    Cell biology international, 1995, Volume: 19, Issue:5

    Transforming growth factor alpha (TGF alpha) is a close relative of epidermal growth factor (EGF), the first polypeptide mitogen discovered in 1962 (Cohen, 1962). TGF alpha, like EGF, exerts its effect on cells through binding to the EGF-Receptor (EGF-R). Here we review the molecular and cell biology of TGF alpha before proceeding to describe our own work on signaling molecules induced in response to activation of the EGF-R.

    Topics: Amino Acid Sequence; Animals; Cell Transformation, Neoplastic; Epidermal Growth Factor; ErbB Receptors; Humans; Models, Biological; Molecular Sequence Data; Protein Structure, Secondary; Psoriasis; ras Proteins; Signal Transduction; Transforming Growth Factor alpha

1995
[Growth factors and oncogenes in development and carcinogenesis. Role of the epidermal growth factor system].
    Minerva medica, 1994, Volume: 85, Issue:9

    The processes of cellular proliferation and progressive acquisition of a specialized phenotype show a remarkable degree of coordination that involves both intracellular programming and intercellular communication. One of the major incentives for studying factors that regulate the processes of cellular proliferation and differentiation is the recognition of their potential contribution to tumorigenesis. In normal cells, stimulatory and inhibitory events are believed to be under the control of growth factors and growth inhibitory factors, which are known to be protooncogene products. Growth regulatory mechanisms usually involve the binding of a growth factor to a specific receptor on the cell surface, which then through an intracellular biochemical cascade leads to cell division. The cell regulation pathways initiated by growth factors may be subverted at several distinct levels in cancer cells. Studies of oncogenes have shown that they may function as abnormal growth factors or abnormal receptors, induce expression of potential signal regulators or encode proteins which modulate gene transcription. The purpose of the present paper is to examine the role of growth factors, growth factor receptors and intracellular proteins involved in signal transduction (with particular regard to the epidermal growth factor receptor system) in the control of normal growth and differentiation, and their contribution to transformation and tumorigenesis. We also review the classical theories of neoplasia and various other models. Chemical carcinogenesis and Vogelstein-Lane model are presented.

    Topics: Amino Acid Sequence; Cell Division; Cell Transformation, Neoplastic; Epidermal Growth Factor; ErbB Receptors; Genes, Tumor Suppressor; Growth; Growth Substances; Humans; Molecular Sequence Data; Neoplasm Metastasis; Neoplasms; Oncogenes; Phenotype; Receptors, Growth Factor; Signal Transduction; Transcription, Genetic; Transforming Growth Factor alpha

1994
Malignant transformation by G protein-coupled hormone receptors.
    Molecular and cellular endocrinology, 1994, Volume: 100, Issue:1-2

    Topics: 3T3 Cells; Animals; Cell Nucleus; Cell Transformation, Neoplastic; Epidermal Growth Factor; ErbB Receptors; GTP-Binding Proteins; Humans; Mice; Mice, Nude; Mitosis; Oncogenes; Protein Kinases; Proto-Oncogene Mas; Proto-Oncogene Proteins; Proto-Oncogene Proteins p21(ras); Receptors, Angiotensin; Receptors, Cell Surface; Receptors, G-Protein-Coupled; Signal Transduction; Type C Phospholipases

1994
Insights into the physiology of TGF alpha and signaling through the EGF receptor revealed by gene targeting and acts of nature.
    Princess Takamatsu symposia, 1994, Volume: 24

    Transforming growth factor alpha (TGF alpha) is one of a group of structurally-related growth factors (the epidermal growth factor family of ligands) that interact with one or other members of the epidermal growth factor family of protein tyrosine kinase receptors (EGF-R's). A number of excellent reviews detailing our knowledge of this area have been recently published (Carpenter and Wahl, 1991; Derynck, 1992; Prigent and Lemoine, 1992). Rather than add to their number, this review focuses on new insights into the importance of TGF alpha and signaling through the EGF receptor considered in the context of the laboratory mouse. The new information has emerged from analysis of mutant mice generated either by classical gene targeting in embryonic stem (ES) cells or by accidents of nature. In addition to their intrinsic interest, these mice are proving invaluable in determining the importance of EGF receptor signaling in wound healing and as a contributing factor in the conversion of a normal cell into its tumorigenic counterpart.

    Topics: 9,10-Dimethyl-1,2-benzanthracene; Animals; Cell Transformation, Neoplastic; Cocarcinogenesis; Epidermal Growth Factor; ErbB Receptors; Gene Targeting; Hair; Mice; Mice, Knockout; Mice, Mutant Strains; Multigene Family; Mutagenesis, Insertional; Papilloma; Recombination, Genetic; Signal Transduction; Skin Neoplasms; Transforming Growth Factor alpha; Vibrissae; Wound Healing

1994
A chimeric EGFR/neu receptor in studies of neu function.
    Cancer treatment and research, 1992, Volume: 61

    Topics: 3T3 Cells; Animals; Cell Division; Cell Transformation, Neoplastic; Enzyme Activation; Epidermal Growth Factor; ErbB Receptors; Gene Expression Regulation; Genes, Synthetic; Mice; Phosphorylation; Promoter Regions, Genetic; Protein Kinase C; Protein Processing, Post-Translational; Protein-Tyrosine Kinases; Proto-Oncogene Proteins; Proto-Oncogenes; Receptor, ErbB-2; Recombinant Fusion Proteins; Transcription Factors; Transforming Growth Factor alpha

1992
The physiology of transforming growth factor-alpha.
    Advances in cancer research, 1992, Volume: 58

    Topics: Amino Acid Sequence; Animals; Binding, Competitive; Cell Division; Cell Transformation, Neoplastic; Embryonic and Fetal Development; Epidermal Growth Factor; ErbB Receptors; Gene Expression Regulation, Neoplastic; Growth Substances; Humans; Membrane Glycoproteins; Mice; Molecular Sequence Data; Multigene Family; Neoplasm Proteins; Neoplasms; Protein Precursors; Protein Processing, Post-Translational; Rats; Signal Transduction; Transforming Growth Factor alpha

1992
[Role of oncogenes and anti-oncogenes in dermatology].
    Pathologie-biologie, 1992, Volume: 40, Issue:2

    Cell growth is controlled by two types of genes, i.e., activating genes (oncogenes) and negative regulator genes (antioncogenes). Studies have shown that malignant transformation of a cell can result from either increased oncogene activity or decreased antioncogene activity. Current knowledge of genes relevant to dermatology is discussed.

    Topics: Animals; Cell Transformation, Neoplastic; Epidermal Growth Factor; Genes, p53; Genes, Tumor Suppressor; Humans; Mice; Proto-Oncogenes; Psoriasis; Skin Neoplasms

1992
Polypeptide modulators of prostatic growth and development.
    Cancer surveys, 1991, Volume: 11

    Normal and abnormal developmental events in the prostate are strongly influenced by androgens. There is abundant evidence, however, that androgens are not the only substances present that have the capacity to influence prostatic growth. A number of polypeptides that either stimulate or inhibit growth have now been identified in the prostate. These include members of the HBGF family, TGF-beta family, EGF and TGF-alpha, PDGF, NGF, and the less well characterized osteoblast growth factors. In some cases, the prostatic cell population, stromal or epithelial, that synthesizes the growth factor and its receptor is known. This information and the properties of the growth factors suggest ways in which these polypeptides may be involved in regulating growth of the prostate, including benign prostatic hyperplasia and prostate cancer.

    Topics: Bone Neoplasms; Cell Transformation, Neoplastic; Epidermal Growth Factor; Fibroblast Growth Factors; Growth Substances; Humans; Male; Neovascularization, Pathologic; Prostate; Receptors, Fibroblast Growth Factor; Transforming Growth Factor alpha; Transforming Growth Factor beta; Wound Healing

1991
Alterations in growth factor pathways in multistage carcinogenesis of rat tracheal epithelial cells.
    Journal of radiation research, 1991, Volume: 32 Suppl 2

    The role of peptide growth factors in the process of multistage carcinogenesis of rat tracheal epithelial (RTE) cells was assessed by examining growth factor requirements and expression of growth factors and their receptors in normal and transformed RTE cells. Transformed RTE cell lines show decreased requirements for bovine pituitary extract, insulin and epidermal growth factor compared to primary RTE cells in culture. An autocrine role for TGF alpha in transformed RTE cells is suggested by data showing TGF alpha production and decreased proliferation in the presence of TGF alpha antisera and TGF alpha/EGF receptor kinase inhibitor. Therefore, decreased EGF requirements in transformed RTE cells could be explained by autocrine TGF alpha regulation. In contrast, no evidence for an insulin/IGF-I autocrine pathway could be detected in transformed RTE cells. These data indicate that multiple alterations in growth factor pathways occur in transformed RTE cells.

    Topics: Animals; Cell Transformation, Neoplastic; Epidermal Growth Factor; Epithelial Cells; Gene Expression; Growth Substances; Rats; Trachea; Transforming Growth Factor alpha

1991
Transforming growth factor-alpha: an oncodevelopmental growth factor.
    Cancer cells (Cold Spring Harbor, N.Y. : 1989), 1990, Volume: 2, Issue:12

    Transforming growth factor-alpha (TGF-alpha) is a 50-amino-acid mitogenic peptide that is structurally and, in some cases, functionally related to members of the epidermal growth factor (EGF) family of peptides. TGF-alpha is initially synthesized as a high-molecular-weight, glycosylated, membrane-associated precursor of approximately 160 amino acids. The low-molecular-weight TGF-alpha peptide as well as the precursor are biologically active in a number of systems and can function as transforming proteins when overexpressed. TGF-alpha binds to and activates the EGF receptor, and TGF-alpha and the EGF receptor are coexpressed in a number of human and rodent tumors and tumor cell lines--which suggests that TGF-alpha can function as an autocrine or paracrine growth factor. TGF-alpha is transiently expressed in some fetal and adjacent maternal tissues during development and is also expressed in a number of adult tissues; this pattern of expression suggests that the growth factor is involved in several distinct physiological functions.

    Topics: Amino Acid Sequence; Animals; Cell Differentiation; Cell Division; Cell Transformation, Neoplastic; Epidermal Growth Factor; ErbB Receptors; Gene Expression; Humans; Molecular Sequence Data; Multigene Family; Transforming Growth Factor alpha; Tumor Cells, Cultured

1990
Sequential changes in epidermal growth factor receptor/ligand function in cultured rat liver epithelial cells transformed chemically in vitro.
    Pathobiology : journal of immunopathology, molecular and cellular biology, 1990, Volume: 58, Issue:1

    Diploid WB rat liver epithelial cells contain abundant, rapidly internalized epidermal growth factor receptors, and respond pleiotropically to ligand binding. Signal transduction pathways downstream from the EGF receptor involve activation of elements that are both dependent on and independent of protein kinase C activation. Neoplastic transformation of wild-type WB rat liver epithelial cells by exposure to N-methyl-N'-nitro-N-nitrosoguanidine is associated with progressive alterations in the responses of affected cells to binding of EGF to EGF receptors, including heightened cell proliferation and the expression of several other phenotypic properties. Tumorigenic rat liver epithelial cells acquire the ability to express transforming growth factor-alpha (TGF-alpha), and to secrete this growth factor in a regulated and then unregulated manner. TGF-alpha expression, together with the presence of abundant EGF receptors, provides affected cells with an autocrine growth cycle. The ability of transformed WB rat liver epithelial cells to produce tumors cosegregates clonally with TGF-alpha expression and with heightened expression of c-myc, c-Ha-ras and c-Ki-ras proto-oncogenes.

    Topics: Animals; Cell Line; Cell Line, Transformed; Cell Transformation, Neoplastic; Epidermal Growth Factor; Epithelium; ErbB Receptors; Gene Expression Regulation, Neoplastic; Liver; Methylnitronitrosoguanidine; Rats; Transforming Growth Factors

1990
Stimulatory and inhibitory growth factors and breast cancer.
    The Journal of steroid biochemistry and molecular biology, 1990, Dec-20, Volume: 37, Issue:6

    While steroid hormones act as endocrine effectors of growth and development of normal breast and of carcinogenesis and progression of malignant breast, recent evidence suggests that local hormonal effectors also exist. These are the growth regulatory growth factors. This article summarizes current status of our understanding of structure and function of growth factors secreted by the normal and malignant mammary epithelium. While growth inhibitory factors and their receptors generally suppress development of the transformed phenotype and promote differentiation, growth stimulatory factors and their receptors may be necessary for both normal proliferation and early stages of malignant progression of breast cancer. Overexpression of two receptors, c-erbB-2 and EGF receptor, have also been associated with poor prognosis in the clinical disease.

    Topics: Antineoplastic Agents; Breast Neoplasms; Cell Transformation, Neoplastic; Epidermal Growth Factor; Growth Substances; Humans; Oncogenes; Transforming Growth Factor alpha

1990
Growth factors and oncogenes.
    Biomedicine & pharmacotherapy = Biomedecine & pharmacotherapie, 1989, Volume: 43, Issue:9

    Recent findings reporting the structure, the mechanism of action of peptide growth factors and their receptor as well as their presence in human tumors have led to a better knowledge of the events involved in cell growth regulation. The ability of cancer cells to produce and to respond to their own growth factors confers on them a growth autonomy and strengthens the link between growth factors and oncogenes. Oncogenes render the cells growth factor-independent not only by coding for autocrine growth factors of their receptor, but also by amplifying the mitogenic signals generated by the binding of the peptide growth factor to its membrane receptor.

    Topics: Animals; Cell Transformation, Neoplastic; Epidermal Growth Factor; Estrogens; Growth Inhibitors; Growth Substances; Humans; Mice; Oncogenes; Rats; Receptors, Cell Surface

1989
Peptide regulatory factors and malignancy.
    Lancet (London, England), 1989, Jul-01, Volume: 2, Issue:8653

    Topics: Cell Differentiation; Cell Division; Cell Transformation, Neoplastic; Epidermal Growth Factor; ErbB Receptors; Female; Growth Substances; Humans; Interferon Type I; Neoplasms; Proto-Oncogenes; Signal Transduction; Transforming Growth Factors

1989
[Epidermal growth factor and cancer].
    Sheng li ke xue jin zhan [Progress in physiology], 1989, Volume: 20, Issue:1

    Topics: Animals; Cell Transformation, Neoplastic; Epidermal Growth Factor; Oncogenes

1989
The role of the neu oncogene product in cell transformation and normal development.
    Princess Takamatsu symposia, 1988, Volume: 19

    p185neu, the protein product of the neu gene, is a tyrosine kinase receptor with structural similarity to the epidermal growth factor (EGF) receptor. The cognate ligand for the p185neu receptor remains unknown. We have defined: 1) stage and tissue-specific expression patterns of the neu gene product in developing tissues; 2) p185neu phosphorylation and the regulation of p185neu tyrosine kinase activity by EGF. 3) Synergistic interactions of cellular rat p185neu and EGF receptor leading to cell transformation; 4) structural and functional differences of normal and oncogenic p185neu. These observations explain some features of how p185neu is involved in normal development and neoplastic transformation.

    Topics: Animals; Cell Transformation, Neoplastic; Epidermal Growth Factor; ErbB Receptors; Gene Expression; Phosphorylation; Protein-Tyrosine Kinases; Proto-Oncogene Proteins; Proto-Oncogenes; Rats; Receptor, ErbB-2

1988
Transforming growth factor alpha.
    Cell, 1988, Aug-26, Volume: 54, Issue:5

    Topics: Animals; Cell Transformation, Neoplastic; Epidermal Growth Factor; Peptides; Transforming Growth Factors

1988
[Effect of epidemic growth factor on cell growth and proliferation].
    Sheng li ke xue jin zhan [Progress in physiology], 1988, Volume: 19, Issue:1

    Topics: Animals; Cell Division; Cell Transformation, Neoplastic; Cells, Cultured; Cornea; Epidermal Growth Factor; Mice; Skin

1988
Proteinases and eukaryotic cell growth.
    Comparative biochemistry and physiology. B, Comparative biochemistry, 1987, Volume: 87, Issue:1

    Topics: Animals; Cell Membrane; Cell Physiological Phenomena; Cell Transformation, Neoplastic; Cells; Endopeptidases; Epidermal Growth Factor; Fibroblasts; Growth; Humans; Mitogens; Nerve Growth Factors; Phosphatidylinositols; Receptors, Cell Surface

1987
[Oncogenes, their implications in cell growth, differentiation and carcinogenesis].
    Gan to kagaku ryoho. Cancer & chemotherapy, 1987, Volume: 14, Issue:2

    Cancer Research has changed substantially over the past several years since oncogenes were isolated from cancer cells. More than 40 oncogenes have been identified to date from tumor viruses and cancer cells. Many of the gene products seem related to signalling pathways that determine growth and differentiation of cells. This review attempts to summarize much of the currently available data and to gain future perspectives.

    Topics: 9,10-Dimethyl-1,2-benzanthracene; Animals; Carcinogens; Cell Transformation, Neoplastic; Cell Transformation, Viral; Epidermal Growth Factor; Genes, Viral; Humans; Mammary Neoplasms, Experimental; Neoplasms; Platelet-Derived Growth Factor; Retroviridae; Transfection; Urinary Bladder Neoplasms

1987
Epidermal growth factor and its receptor.
    Molecular and cellular endocrinology, 1987, Volume: 51, Issue:3

    Epidermal growth factor (EGF) binds with high affinity and specificity to a single site on the external domain of its transmembrane receptor to activate the tyrosine protein kinase activity of its cytoplasmic portion. The EGF receptor gene is amplified and over-expressed in several human tumors, suggesting that increased concentrations of the proto-oncogene leads to constitutive activity similar to that seen with oncogene erb B. Synthesis and degradation of the EGF receptor are regulated, in addition, covalent modification by phosphorylation regulates activity of the receptor protein. Intramolecular self-phosphorylation of Tyr1173 removes a competitive inhibitory constraint to enhance phosphorylation of substrates. Phosphorylation of Thr654 by protein kinase C decreases high affinity EGF binding and EGF-stimulated tyrosine protein kinase activity, providing a mechanism for heterologous regulation of the EGF receptor by tumor promoters and other ligand X receptor complexes. Extensive regulation contributes to normal growth control, abrogation of regulatory controls contributes to uncontrolled growth as seen with erb B transformation and EGF receptor gene amplification in human tumors.

    Topics: Cell Division; Cell Transformation, Neoplastic; Enzyme Activation; Epidermal Growth Factor; ErbB Receptors; Gene Amplification; Humans; Phosphorylation; Protein Kinase C; Protein-Tyrosine Kinases; Proto-Oncogene Mas; Proto-Oncogenes; Threonine; Tyrosine

1987
Polypeptide growth factors: roles in normal and abnormal cell growth.
    Annual review of cell biology, 1987, Volume: 3

    An increasing number of polypeptide growth factors have been identified that regulate not only cell proliferation but an extraordinary range of cell activities, including matrix protein deposition and resolution, the maintenance of cell viability, cell differentiation, inflammation, and tissue repair. Normal cells appear to require growth factors for proliferation and for maintenance of viability. Cells that secrete a polypeptide growth factor have an advantage in growth. These factors can act either externally through cell surface receptors or perhaps internally during the transport of receptors and growth factors through the ER and Golgi, causing autocrine stimulation of cell growth. Depending on the cell type, growth factors can also be potent inhibitors of cell growth rather than stimulating growth, and the effects can depend on the presence or absence of other growth factors. Platelet-derived growth factor has been shown to be nearly identical to the product of the v-sis gene of the simian sarcoma virus, which appears to cause cell transformation through its interactions with the PDGF receptor activating the tyrosine kinase activity of the PDGF receptor. Similarly, two proto-oncogenes, c-erbB and c-fms, encode growth factor receptors. The EGF receptor activity of the v-erb oncogene product appears to be constitutively activated without the need for growth factor, perhaps because of the truncation at the amino terminus deleting the EGF binding domain. The induction of the myc and the fos proteins by growth factor stimulation of quiescent cells, as well as the potential for the p21 product of the ras oncogene to act as an intermediate in transducing adrenergic signals, provide direct evidence that these pathways are important for stimulation of cell growth. Cells transformed by the v-sis oncogene always appear to bear PDGF cell surface receptors, which suggests that this oncogene has a specific requirement of the PDGF receptor for transformation. In contrast, cells transformed by the v-erbB and v-fms oncogenes are not stimulated by EGF or by CSF-1. Thus it seems likely that the tyrosine kinase activity of the corresponding receptor is ubiquitously expressed in these cases. Major questions remain unanswered. In particular, what are the mechanisms by which growth factors initiate pathways leading to DNA synthesis? What are the physiological substrates of the growth factor receptor tyrosine kinase? Considerable effort also is needed to further define the

    Topics: Animals; Cell Division; Cell Transformation, Neoplastic; Epidermal Growth Factor; Growth Substances; Humans; Peptides; Platelet-Derived Growth Factor

1987
Role of epidermal growth factor in carcinogenesis.
    Cancer research, 1986, Volume: 46, Issue:3

    For cell growth and division to occur, a large variety of metabolic processes must be carefully coordinated in the cell. Through evolutionary pressures, specific hormones and growth factors have acquired the ability to trigger a complex coordinated "pleiotropic growth response" in their target cells. This complex response is mediated by specific cellular receptors and intracellular messengers. Teleologically then, it makes sense that in oncogenesis this growth regulating network is utilized by the production of proteins which mimic growth factors, the activated form of their receptors or, the messengers themselves. Several lines of evidence indicate that the epidermal growth factor-stimulated growth regulatory system is involved in cellular proliferation, both normal and neoplastic. Some of the effects of epidermal growth factor in carcinogenesis are separable from its direct, growth stimulatory effects. Thus, the role of epidermal growth factor in carcinogenesis is more complex than is its role in stimulating growth.

    Topics: Animals; Carcinogens; Cell Transformation, Neoplastic; Cell Transformation, Viral; Epidermal Growth Factor; ErbB Receptors; Humans; Immune Tolerance; Neoplasms; Oncogene Proteins, Viral; Oncogenes; Peptides; Receptors, Cell Surface; Transforming Growth Factors

1986
Growth factors in cancer and their relationship to oncogenes.
    Cancer investigation, 1986, Volume: 4, Issue:1

    Topics: Bombesin; Cell Line; Cell Transformation, Neoplastic; Epidermal Growth Factor; ErbB Receptors; Gastrins; Growth Substances; Humans; Neoplasms; Nerve Growth Factors; Oncogenes; Platelet-Derived Growth Factor; Receptors, Cell Surface; Somatomedins; Transferrin

1986
Paracrine action of transforming growth factors.
    Clinics in endocrinology and metabolism, 1986, Volume: 15, Issue:1

    Polypeptide growth factors form a class of regulatory molecules which exert their effects by binding to specific receptors present on the cell surface. Most of the time the exact role of these factors in the healthy body is unknown. Some, like PDGF and TGF beta, seem to be involved in wound healing. Others, like EGF, promote epithelial cell growth and differentiation. The site of synthesis of most polypeptide growth factors is unknown. Their target can be identified by detecting the cells which present the specific receptors at their surface. It is though that polypeptide growth factors have a paracrine mode of action. Many different cancerous cells produce polypeptide growth factors and the appropriate receptors. Thus, they are able to stimulate their own growth in an autocrine fashion. Recently, some polypeptide growth factors and receptor genes or cDNAs have been molecularly cloned. Growth factor genes and messengers are much more complex than would be expected from the size of the polypeptide. Some cDNAs have been introduced into bacterial expression vectors and large amounts of the factors have been produced by bacteria. New tools, such as molecular probes and specific antibodies, are thus now available to investigate the production of the growth factors and their receptors. The same tools will facilitate the identification and understanding of the molecular mechanism whereby cancerous cells produce the growth factors and the appropriate receptors simultaneously. The importance of growth factors and receptors in cancer is stressed by the finding that three oncogenes are in fact the genes coding for one growth factor and two receptors. Finally, the molecular probes and the specific antibodies raised against these molecules can be used to identify precisely the growth factor(s) and receptor(s) produced abnormally in cancers. Antibodies that inhibit specifically the interaction of this very growth factor with its receptor could then be developed, thus allowing human tumour cell growth to be controlled.

    Topics: Animals; Cell Transformation, Neoplastic; Cloning, Molecular; Endocrine Glands; Epidermal Growth Factor; ErbB Receptors; Growth Substances; Humans; Neoplasms; Oncogenes; Peptides; Platelet-Derived Growth Factor; Receptors, Cell Surface; Receptors, Platelet-Derived Growth Factor; Transforming Growth Factors

1986
[Interaction of growth factors and oncogenes in the tumorous transformation of cells].
    Eksperimental'naia onkologiia, 1986, Volume: 8, Issue:2

    Different aspects of the interaction of polypeptide growth factors with their receptors are reviewed. The problem of structural and functional interactions of the normal and transforming growth factors as well as the protein products of oncogenes during transmittance of a mitogenic signal and the proliferation regulation of normal and tumour cells is discussed.

    Topics: Animals; Cell Transformation, Neoplastic; Cell Transformation, Viral; Cells, Cultured; DNA, Neoplasm; DNA, Viral; Drug Interactions; Epidermal Growth Factor; Gene Expression Regulation; Genes, Viral; Growth Substances; Male; Mice; Oncogenes; Oncogenic Viruses; Peptides; Platelet-Derived Growth Factor; Protein Biosynthesis; Receptors, Cell Surface; Transforming Growth Factors; Translocation, Genetic

1986
[The role of hormones in neoplastic transformation].
    Nihon rinsho. Japanese journal of clinical medicine, 1986, Volume: 44, Issue:2

    Topics: 9,10-Dimethyl-1,2-benzanthracene; Animals; Cell Transformation, Neoplastic; Epidermal Growth Factor; Hormones; Mammary Glands, Animal; Mammary Neoplasms, Experimental; Neoplasms, Hormone-Dependent; Prolactin; Receptors, Cell Surface; Thyrotropin; Urethane

1986
Functional and structural characteristics of EGF and its receptor and their relationship to transforming proteins.
    Journal of cellular biochemistry, 1986, Volume: 31, Issue:2

    Epidermal growth factor (EGF) is a peptide which effects the growth and/or differentiated functions of many cell types. Several pieces of evidence indicate that EGF and its receptor may play a role in carcinogenesis. Functional and structural characteristics of EGF and its receptor and their relationship to transforming proteins are discussed. EGF has extensive homology with alpha-transforming growth factor (alpha-TGF), which may actually be an embryonic form of EGF. Nevertheless, both EGF and alpha-TGF elicit transformation-associated phenotypes in target cells under certain conditions. EGF effects are mediated by a receptor present on the plasma membrane. The EGF receptor is a highly complex protein having several functions in addition to binding EGF in a highly specific manner. One of these functions is to phosphorylate tyrosyl residues on certain proteins. This activity is similar to that expressed by the src family of oncogene-encoded proteins. Besides sharing functional homology the EGF receptor also exhibits structural homology to several oncogene-encoded proteins. The v-erb-B-transforming protein has a striking extent of homology (95%) to the cytoplasmic portion of the EGF receptor. These data support the concept that some aspect of EGF-stimulated metabolism is involved in cellular transformation.

    Topics: Animals; Cell Line; Cell Transformation, Neoplastic; Epidermal Growth Factor; ErbB Receptors; Humans; Molecular Weight; Neoplasm Proteins; Peptides; Receptors, Cell Surface; Transforming Growth Factors

1986
Transformation-related growth factors and their receptors.
    International journal of cell cloning, 1986, Volume: 4, Issue:4

    Cellular transformation may be accomplished in vitro and in vivo through the concerted action of growth factors and oncogenes. This association has demonstrated that malignant growth results from aberrations in growth factor-signal transduction pathways that normally operate to control proliferation. Activation of genes that code for growth factors and/or their receptors provides tumor cells with potential mechanisms to maintain their proliferative state. Tumor cells have been shown to produce endogenous substances that augment their growth (autocrine stimulation), as well as responding to exogenous substances (paracrine stimulation). With solid tumor cells these responses have been shown to involve aberrant expression of growth factor and/or receptor genes. The study of the interrelationship of these various growth regulatory molecules is important not only in the identification of gene products essential to cellular proliferation, but also in providing clues as to what forces are driving tumor cell growth.

    Topics: Animals; Cell Division; Cell Transformation, Neoplastic; Epidermal Growth Factor; ErbB Receptors; Gene Expression Regulation; Growth Substances; Humans; Oncogenes; Peptides; Phenotype; Receptors, Cell Surface; Transforming Growth Factors

1986
The growth factor-platelet-transformation connection.
    Developmental biology (New York, N.Y. : 1985), 1986, Volume: 3

    Topics: Animals; Blood Platelets; Cell Transformation, Neoplastic; Epidermal Growth Factor; Humans; Platelet-Derived Growth Factor; Transforming Growth Factors

1986
[Abnormal control of cell division and neoplastic transformation].
    Nihon rinsho. Japanese journal of clinical medicine, 1986, Volume: 44, Issue:2

    Topics: Animals; Cell Division; Cell Transformation, Neoplastic; Epidermal Growth Factor; Interleukin-6; Liver Neoplasms, Experimental; Liver Regeneration; Monocytes; Proteins

1986
Transforming growth factor-alpha: structure and biological activities.
    Journal of cellular biochemistry, 1986, Volume: 32, Issue:4

    Topics: Amino Acid Sequence; Animals; Cell Transformation, Neoplastic; Epidermal Growth Factor; Humans; Mice; Peptides; Protein Precursors; Rats; Structure-Activity Relationship; Transforming Growth Factors

1986
The role of progenitor cells in cellular differentiation, cellular senescence and neoplastic transformation.
    Progress in clinical and biological research, 1986, Volume: 226

    Topics: Adipose Tissue; Animals; Cell Differentiation; Cell Survival; Cell Transformation, Neoplastic; Cricetinae; Epidermal Growth Factor; Fibroblast Growth Factors; Humans; Mesoderm; Muscles; Phorbol Esters; Stem Cells

1986
Transforming growth factors.
    Cancer surveys, 1985, Volume: 4, Issue:4

    The term transforming growth factor (TGF) has been applied to peptides that have the ability to confer the transformed phenotype on untransformed fibroblastic indicator cells in vitro. Peptides representing two distinct classes of TGFs have been purified to homogeneity. Type alpha and type beta TGFs are distinguished both chemically by their unique amino acid sequences and biologically by their different activities on cells. Type alpha TGFs are single chain peptides of 50-53 amino acids cross-linked by three disulphide bonds. They have strong homology to epidermal growth factor with which they compete for receptor binding. Type beta TGFs have a homodimeric structure comprised of two chains of 112 amino acids, each containing nine cysteine residues; TGF beta binds to a unique cell surface receptor. Type alpha TGFs are usually mitogenic for fibroblasts, whereas type beta TGFs have bifunctional effects on cell growth and can either stimulate or inhibit growth of the same cells, depending on conditions. The interactions of type alpha and beta TGFs can be either synergistic or antagonistic. Though the development of peptide antagonists to type alpha TGFs may have therapeutic potential for the treatment of malignancy, type beta TGFs may inhibit tumorigenesis.

    Topics: Amino Acid Sequence; Animals; Cell Transformation, Neoplastic; Cloning, Molecular; Epidermal Growth Factor; Humans; Molecular Sequence Data; Peptides; Protein Conformation; Receptors, Cell Surface; Receptors, Transforming Growth Factor beta; Species Specificity; Transforming Growth Factors

1985
A possible purification of mullerian inhibiting substance and a model for its mechanism of action.
    Progress in clinical and biological research, 1985, Volume: 171

    Topics: Animals; Anti-Mullerian Hormone; Cattle; Cell Transformation, Neoplastic; Chromatography, Affinity; Epidermal Growth Factor; ErbB Receptors; Glycoproteins; Growth Inhibitors; Growth Substances; Insulin; Male; Manganese; Membrane Proteins; Models, Biological; Mullerian Ducts; Phosphoprotein Phosphatases; Phosphorylation; Protein Kinase Inhibitors; Protein Tyrosine Phosphatases; Protein-Tyrosine Kinases; Testicular Hormones; Testis

1985
Cytoskeletal events during calcium- or EGF-induced initiation of DNA synthesis in cultured cells. Role of protein phosphorylation and clues in the transformation process.
    Cell and muscle motility, 1985, Volume: 6

    The possible relationship between cytoskeletal events and growth regulation in response to stimulation by calcium and by growth factors such as EGF can be summarized as follows: An elaborate interaction exists between calcium and serum growth factors, such as EGF, in the initiation of DNA synthesis in quiescent cells. This implies that many processes between the external signals delivered at the cell surface and the sequential intracellular events that lead to chromosomal replication, and ultimately to cell division, must be coordinated in a reproducible manner. It is now apparent that because of its possible role as a dynamic integrator of the cytoplasm, the cytoskeleton could represent the coordinator of the events that lead to replication. Calcium (with its intracellular acceptor, calmodulin) and cAMP (which can act by opposing mechanisms) are extensively involved in the control of the integrity of the cytoskeleton. Distinct protein kinases are activated by calcium/calmodulin, EGF, and cAMP as aspects of the prereplicative response, and many of the substrates for phosphorylation are cytoskeletal proteins. The emerging picture seems to include a direct involvement of these protein kinases in the cascade of regulatory events that leads to the initiation of DNA synthesis. Thus, the cytoskeleton has a direct role in the transmission of proliferative signals from external receptor sites to the nucleus. A means by which neoplastic cells can bypass the normal regulatory pathways is proposed in the light of recent data showing that the product of oncogenes are protein kinases or proteins that intimately interact with cellular protein kinases.

    Topics: Animals; Calcium; Calmodulin; Cell Adhesion; Cell Cycle; Cell Transformation, Neoplastic; Cells, Cultured; Cyclic AMP; Cytoskeletal Proteins; Cytoskeleton; DNA; Epidermal Growth Factor; Humans; Microtubules; Phosphorylation; Protein Kinases; Receptors, Cell Surface

1985
Growth factors, oncogenes and transformation. Part I: Growth factors and cell cycle control.
    Arzneimittel-Forschung, 1985, Volume: 35, Issue:4

    Topics: Animals; Cell Division; Cell Transformation, Neoplastic; Cell Transformation, Viral; Cells, Cultured; Epidermal Growth Factor; ErbB Receptors; Gene Expression Regulation; Growth Substances; Hormones; Humans; Lymphocyte Activation; Mitogens; Oncogenes; Oncogenic Viruses; Receptors, Cell Surface

1985
Growth factors and oncogenes in human malignant glioma.
    Neurologic clinics, 1985, Volume: 3, Issue:4

    Normal cell replication is regulated by growth factors such as epidermal growth factor (EGF) and platelet-derived growth factor (PDGF) that act through binding to specific surface receptors on target cells. Oncogenes may exert their transforming activity by encoding proteins that mimic the function of the normal regulatory factors along the mitogenic pathway, growth factors, their receptors or elements along the postreceptor signaling system. This may be exemplified by the human malignant glioma, in which the sis gene (encoding a growth factor homologous to PDGF) and the erb B gene (encoding a membrane protein homologous to the EGF receptor) have been implicated.

    Topics: Animals; Cell Cycle; Cell Line; Cell Transformation, Neoplastic; Epidermal Growth Factor; ErbB Receptors; Gene Amplification; Glioblastoma; Glioma; Growth Substances; Humans; Middle Aged; Mitosis; Oncogenes; Platelet-Derived Growth Factor; Receptors, Cell Surface; Receptors, Platelet-Derived Growth Factor

1985
The 1985 Walter Hubert lecture. Malignant cell differentiation as a potential therapeutic approach.
    British journal of cancer, 1985, Volume: 52, Issue:3

    Most drugs available for cancer chemotherapy exert their effects through cytodestruction. Although significant advances have been attained with these cytotoxic agents in several malignant diseases, response is often accompanied by significant morbidity and many common malignant tumours respond poorly to existing cytotoxic therapy. Development of chemotherapeutic agents with non-cytodestructive actions appears desirable. Considerable evidence exists which indicates that (a) the malignant state is not irreversible and represents a disease of altered maturation, and (b) some experimental tumour systems can be induced by chemical agents to differentiate to mature end-stage cells with no proliferative potential. Thus, it is conceivable that therapeutic agents can be developed which convert cancer cells to benign forms. To study the phenomenon of blocked maturation, squamous carcinoma SqCC/Y1 cells were employed in culture. Using this system it was possible to demonstrate that physiological levels of retinoic acid and epidermal growth factor were capable of preventing the differentiation of these malignant keratinocytes into a mature tissue-like structure. The terminal differentiation caused by certain antineoplastic agents was investigated in HL-60 promyelocytic leukaemia cells to provide information on the mechanism by which chemotherapeutic agents induce cells to by-pass a maturation block. The anthracyclines aclacinomycin A and marcellomycin were potent inhibitors of N-glycosidically linked glycoprotein biosynthesis and transferrin receptor activity, and active inducers of maturation; temporal studies suggested that the biochemical effects were associated with the differentiation process. 6-Thioguanine produced cytotoxicity in parental cells by forming analog nucleotide. In hypoxanthine-guanine phosphoribosyltransferase negative HL-60 cells the 6-thiopurine initiated maturation; this action was due to the free base (and possibly the deoxyribonucleoside), a finding which separated termination of proliferation due to cytotoxicity from that caused by maturation.

    Topics: Antibiotics, Antineoplastic; Antineoplastic Agents; Carcinoma, Squamous Cell; Cell Differentiation; Cell Division; Cell Line; Cell Survival; Cell Transformation, Neoplastic; Epidermal Growth Factor; Humans; Leukemia, Myeloid; Models, Biological; Naphthacenes; Thioguanine; Tretinoin

1985
Oncogenes: growth regulation and the papovaviruses polyoma and SV40.
    Journal of cellular biochemistry, 1984, Volume: 26, Issue:2

    Cellular oncogenes and their activated and retrovirus-coded counterparts play an important role in cellular regulation. Here the relationship between such oncogenes and the genes coding for the transforming proteins of the papovaviruses, polyoma viruses, and simian virus 40 (SV40) is discussed. It is concluded that polyoma virus may transform established cells by a mechanism involving activation of a cellular oncogene product, whereas SV40 may transform by a mechanism involving a previously little studied cytoplasmic form of the transforming protein.

    Topics: Animals; Antigens, Polyomavirus Transforming; Antigens, Viral, Tumor; Base Sequence; Cell Division; Cell Line; Cell Transformation, Neoplastic; Cell Transformation, Viral; DNA; DNA-Binding Proteins; DNA, Viral; Epidermal Growth Factor; Humans; Neoplasms; Oncogenes; Papillomaviridae; Platelet-Derived Growth Factor; Polyomaviridae; Polyomavirus; Protein Kinases; Protein-Tyrosine Kinases; Simian virus 40; Transcription, Genetic; Viral Proteins

1984
Large bowel growth.
    Scandinavian journal of gastroenterology. Supplement, 1984, Volume: 93

    Developmental events in the large bowel have been studied pre- and post-natally and indicate that by birth, crypt organisation and kinetic activity are organised along adult lines. The period from birth to maturity is marked by an increase in crypt size and a massive increase in their number, with new crypts developing by a process of longitudinal fission from the base of existing ones. We know little of the fate of crypt size and number thereafter. Adaptive responses to resection or bypass of intestine are much less marked in large bowel when compared to small bowel, but in general postoperative responses have not been as extensively examined. Of the factors maintaining mass and cell turnover in large bowel mucosa simple luminal bulk seems to be most important, although a role exists for endocrine and neurovascular influences. Knowledge of growth, kinetic activity and adaptive responses in human large bowel is scanty and represents a large area for further study.

    Topics: Animals; Carcinogens; Cell Division; Cell Transformation, Neoplastic; Colectomy; Colon; Colostomy; Diet; Epidermal Growth Factor; Glucagon-Like Peptides; Humans; Intestinal Mucosa; Intestine, Large; Rats

1984
A model of cell cycle control: sequential events regulated by growth factors.
    Molecular and cellular endocrinology, 1983, Volume: 31, Issue:2-3

    PDGF is a potent mitogen that initiates the proliferation of quiescent fibroblastic cells. EGF and somatomedin C (or insulin) can replace the requirement for plasma to function synergistically with PDGF to stimulate DNA synthesis. PDGF, EGF and somatomedin C control discrete cellular events in the cell cycle. Cyclic AMP can potentiate the effects of polypeptide mitogens. The down-regulation of EGF receptors by PDGF and cyclic AMP brings about a loss of the requirement for exogenous EGF. The transient treatment of density-arrested fibroblasts with PDGF allows better study of synergistic actions of PDGF and plasma-derived factors. These synergistic interactions are important to understand in determining how multiple growth factors regulate cellular proliferation.

    Topics: Animals; Blood; Calcium; Cell Cycle; Cell Division; Cell Transformation, Neoplastic; Cell Transformation, Viral; Cells, Cultured; Cyclic AMP; DNA; Drug Synergism; Epidermal Growth Factor; ErbB Receptors; Fibroblast Growth Factors; Fibroblasts; Growth Substances; Humans; Insulin-Like Growth Factor I; Mice; Models, Biological; Platelet-Derived Growth Factor; Protein Biosynthesis; Receptors, Cell Surface; Receptors, Platelet-Derived Growth Factor; Somatomedins

1983
[Oncogenes and cell transformation].
    Voprosy onkologii, 1983, Volume: 29, Issue:7

    Topics: Animals; Antigens, Viral; Cell Transformation, Neoplastic; Cricetinae; Epidermal Growth Factor; Humans; Laryngeal Neoplasms; Mice; Neoplasms, Radiation-Induced; Oncogenes; Oncogenic Viruses; Osteosarcoma; Peptides; Repetitive Sequences, Nucleic Acid; Transforming Growth Factors

1983
Molecular aspects of growth factor action: receptors and intracellular signals.
    The Journal of pathology, 1983, Volume: 141, Issue:3

    An increasing number of factors have been described which influence the proliferative responses of normal and neoplastic cells. A review of such moieties, their occurrence and possible mechanisms of their action and interaction is presented together with a discussion of their potential importance in the genesis and progression of neoplasms.

    Topics: Animals; Cell Division; Cell Line; Cell Membrane; Cell Transformation, Neoplastic; Epidermal Growth Factor; Feedback; Growth Substances; Humans; Mice; Mitogens; Phorbol 12,13-Dibutyrate; Phorbol Esters; Rats; Receptors, Drug

1983
Chemical-viral interactions in cell transformation.
    Cancer investigation, 1983, Volume: 1, Issue:6

    Topics: Animals; Carcinogens; Cell Adhesion; Cell Transformation, Neoplastic; Cell Transformation, Viral; Cells, Cultured; Cocarcinogenesis; DNA, Neoplasm; Epidermal Growth Factor; Humans; Methylation; Phenotype

1983
Role of polypeptide growth factors in normal and abnormal growth.
    Kidney international, 1983, Volume: 23, Issue:4

    In the preceding sections we have shown evidence that growth-promoting factors are involved in three basic situations. In normal embryonic development and function of mature organisms, growth factors such as NGF and EGF are of prime importance in supporting the necessary embryonic cell proliferation and the development of specific cell types. Other factors operate on subsets of mature cells during specialized functions such as inflammation. Included in this set would be factors such as CSF/MGF and Interleukin-2. Another basic function of growth factors has been shown to be wound repair and organ regeneration. This includes the well characterized PDGF and FGF as well as the various renotropic factors and liver growth factors. As these factors must operate in mature organisms with many different cell types and similar cell types in many locations, more specificity is needed than in embryonic growth. This has resulted in the organ specific factors such as the renotropins and in the unique delivery system of the PDGF. The recent discovery and characterization of the transforming growth factors has provided a possible connection between embryonic and normal developmental growth and the rapid cellular proliferation characteristic of tumor cells. The TGF not only interacts with receptors for normal growth factors such as EGF but are also detectable in low levels in normal tissue and embryos. The exact relationships between these various factors will have to await the determinations of more amino acid sequences for comparisons. The other tumor-related product, tumor angiogenesis factor, is also found in normal tissue and inflammatory reaction sites.(ABSTRACT TRUNCATED AT 250 WORDS)

    Topics: Angiogenesis Inducing Agents; Animals; Cell Division; Cell Transformation, Neoplastic; Epidermal Growth Factor; Fibroblast Growth Factors; Growth Substances; Humans; Hypertrophy; Interleukin-2; Interleukin-3; Neoplasms; Nerve Growth Factors; Peptides; Platelet-Derived Growth Factor; Transforming Growth Factors; Wound Healing

1983
[Polypeptide growth factors of normal and neoplastic cells].
    Postepy biochemii, 1982, Volume: 28, Issue:1-2

    Topics: Animals; Blood Platelets; Cell Cycle; Cell Transformation, Neoplastic; Cells, Cultured; Chickens; Embryo, Mammalian; Embryo, Nonmammalian; Epidermal Growth Factor; Female; Fibroblasts; Growth Substances; Humans; In Vitro Techniques; Insulin; Kidney; Liver; Mice; Neoplasms; Nerve Growth Factors; Peptides; Pituitary Gland; Pregnancy; Rats; Somatomedins

1982
Epidermal growth factor receptors.
    Molecular and cellular biochemistry, 1981, Feb-11, Volume: 34, Issue:3

    EGF-Rs are cell membrane glycoproteins of wide distribution. They have not yet been fully characterized or purified but are probably molecules of 170-190,000 mol. wt. in most cells. The growth factor EGF binds and will saturate cell surface receptors with a KA of about 5 X 10(9) M-1 although a receptor class with an affinity in excess of 10(10) M-1 has been detected in some cells. The number of receptors on a cell does not determine the level of its response. Some cell types have receptors which bind EGF, but with no mitogenic response. The ways in which receptor affinity and/or number is modulated are described. This and other evidence is reviewed in a search for a suitable model of a mechanism of action on the cell, which best fits the current data. There is ample evidence that EGF binds to the receptor; that ligand-receptor complexes cluster or aggregate; and then are internalized and degraded, but evidence for a direct connection between internalization and the subsequent mitogenic response is lacking. Good correlations between internalization and mitogenic responses have been observed and developed into a theory of endocytic activation, but there is a body of evidence which cannot be accommodated by this theory. Instead, an alternative model is suggested.

    Topics: Animals; Binding Sites; Cell Membrane; Cell Transformation, Neoplastic; Cell Transformation, Viral; DNA; Epidermal Growth Factor; ErbB Receptors; Female; Humans; Kinetics; Models, Biological; Molecular Weight; Organ Specificity; Peptides; Pregnancy; Receptors, Cell Surface; Species Specificity; Teratoma; Tissue Distribution; Tretinoin

1981
Membrane phosphorylation: a crucial role in the action of insulin, EGF, and pp60src?
    Bioscience reports, 1981, Volume: 1, Issue:1

    Topics: Adipose Tissue; Animals; Cell Transformation, Neoplastic; Cell Transformation, Viral; Cyclic AMP; Epidermal Growth Factor; ErbB Receptors; Insulin; Membrane Proteins; Models, Biological; Oncogene Protein pp60(v-src); Phosphorylation; Protein Kinases; Proteins; Receptor, Insulin; Receptors, Cell Surface; Sodium-Potassium-Exchanging ATPase; Viral Proteins

1981
Epidermal growth factor-urogastrone, a polypeptide acquiring hormonal status.
    Vitamins and hormones, 1979, Volume: 37

    Topics: Animals; Binding Sites; Cattle; Cell Transformation, Neoplastic; Cells, Cultured; Cricetinae; Cross-Linking Reagents; Epidermal Growth Factor; Female; Gastric Juice; Gastrointestinal Hormones; Hormones; Humans; Liver; Male; Mice; Peptides; Placenta; Skin; Structure-Activity Relationship; Submandibular Gland

1979
Factors involved in the modulation of cell proliferation in vivo and in vitro: the role of fibroblast and epidermal growth factors in the proliferative response of mammalian cells.
    In vitro, 1978, Volume: 14, Issue:1

    The control of proliferation of mesoderm-derived cells by EGF and FGF has been examined taking, as an example, the vascular endothelium. The mechanisms by which cell proliferation can be brought to a stop in vivo and in vitro have been reviewed.

    Topics: Adrenocorticotropic Hormone; Angiogenesis Inducing Agents; Cartilage; Cell Adhesion; Cell Division; Cell Transformation, Neoplastic; Cells, Cultured; Contact Inhibition; DNA; Epidermal Growth Factor; Fibroblasts; Growth Substances; Membrane Proteins; Mitogens; Peptides; Thrombin

1978

Other Studies

491 other study(ies) available for epidermal-growth-factor and Cell-Transformation--Neoplastic

ArticleYear
Chromium (VI)-induced ALDH1A1/EGF axis promotes lung cancer progression.
    Clinical and translational medicine, 2022, Volume: 12, Issue:12

    Cr(VI) is broadly applied in industry. Cr(VI) exposure places a big burden on public health, thereby increasing the risk of lung squamous cell carcinoma (LUSC). The mechanisms underlying Cr(VI)-induced LUSC remain largely elusive. Here, we report that the cancer stem cell (CSC)/tumour-initiating cell (TIC)-like subgroup within Cr(VI)-transformed bronchial epithelial cells (CrT) promotes lung cancer tumourigenesis. Mechanistically, Cr(VI) exposure specifically increases the expression levels of aldehyde dehydrogenase 1A1 (ALDH1A1), a CSC marker, through KLF4-mediated transcription. ALDH1A1 maintains self-renewal of CrT/TICs and facilitates the expression and secretion of EGF from CrT/TICs, which subsequently promotes the activation of EGFR signalling in differentiated cancer cells and tumour growth of LUSC. In addition, the ALDH1A1 inhibitor A37 and gemcitabine synergistically suppress LUSC progression. Importantly, high ALDH1A1 expression levels are positively correlated with advanced clinical stages and predict poor survival in LUSC patients. These findings elucidate how ALDH1A1 modulates EGF secretion from TICs to facilitate LUSC tumourigenesis, highlighting new therapeutic strategies for malignant lung cancers.

    Topics: Aldehyde Dehydrogenase; Aldehyde Dehydrogenase 1 Family; Carcinogenesis; Carcinoma, Non-Small-Cell Lung; Carcinoma, Squamous Cell; Cell Transformation, Neoplastic; Epidermal Growth Factor; Humans; Lung; Lung Neoplasms; Neoplastic Processes; Retinal Dehydrogenase; Tics

2022
Structure and bioactivity of sulfated α-D-mannan from marine bacterium Halomonas halocynthiae KMM 1376
    Carbohydrate polymers, 2020, Feb-01, Volume: 229

    Halomonas halocynthiae KMM 1376

    Topics: Acetates; Animals; Cell Line, Tumor; Cell Survival; Cell Transformation, Neoplastic; Epidermal Growth Factor; Halomonas; Humans; Hydrolysis; Lipopolysaccharides; Mannans; Mice; Polysaccharides, Bacterial; Sulfates

2020
Epimagnolin targeting on an active pocket of mammalian target of rapamycin suppressed cell transformation and colony growth of lung cancer cells.
    Molecular carcinogenesis, 2019, Volume: 58, Issue:7

    Mammalian target of rapamycin (mTOR) has a pivotal role in carcinogenesis and cancer cell proliferation in diverse human cancers. In this study, we observed that epimagnolin, a natural compound abundantly found in Shin-Yi, suppressed cell proliferation by inhibition of epidermal growth factor (EGF)-induced G1/S cell-cycle phase transition in JB6 Cl41 cells. Interestingly, epimagnolin suppressed EGF-induced Akt phosphorylation strongly at Ser473 and weakly at Thr308 without alteration of phosphorylation of MAPK/ERK kinases (MEKs), extracellular signal-regulated kinase (ERKs), and RSK1, resulting in abrogation of the phosphorylation of GSK3β at Ser9 and p70S6K at Thr389. Moreover, we found that epimagnolin suppressed c-Jun phosphorylation at Ser63/73, resulting in the inhibition of activator protein 1 (AP-1) transactivation activity. Computational docking indicated that epimagnolin targeted an active pocket of the mTOR kinase domain by forming three hydrogen bonds and three hydrophobic interactions. The prediction was confirmed by using in vitro kinase and adenosine triphosphate-bead competition assays. The inhibition of mTOR kinase activity resulted in the suppression of anchorage-independent cell transformation. Importantly, epimagnolin efficiently suppressed cell proliferation and anchorage-independent colony growth of H1650 rather than H460 lung cancer cells with dependency of total and phosphorylated protein levels of mTOR and Akt. Inhibitory signaling of epimagnolin on cell proliferation of lung cancer cells was observed mainly in mTOR-Akt-p70S6K and mTOR-Akt-GSK3β-AP-1, which was similar to that shown in JB6 Cl41 cells. Taken together, our results indicate that epimagnolin potentiates as chemopreventive or therapeutic agents by direct active pocket targeting of mTOR kinase, resulting in sensitizing cancer cells harboring enhanced phosphorylation of the mTORC2-Akt-p70S6k signaling pathway.

    Topics: Animals; Cell Line, Tumor; Cell Proliferation; Cell Transformation, Neoplastic; Chemoprevention; Drugs, Chinese Herbal; Epidermal Growth Factor; Extracellular Signal-Regulated MAP Kinases; G1 Phase Cell Cycle Checkpoints; Glycogen Synthase Kinase 3 beta; HEK293 Cells; Humans; JNK Mitogen-Activated Protein Kinases; Lignans; Lung Neoplasms; Mice; Molecular Docking Simulation; Phosphorylation; Protein Conformation; Proto-Oncogene Proteins c-akt; Ribosomal Protein S6 Kinases, 70-kDa; Ribosomal Protein S6 Kinases, 90-kDa; RNA Interference; RNA, Small Interfering; TOR Serine-Threonine Kinases

2019
Carvedilol inhibits EGF-mediated JB6 P+ colony formation through a mechanism independent of adrenoceptors.
    PloS one, 2019, Volume: 14, Issue:5

    Carvedilol is reported to prevent cancers in humans and animal models. However, a molecular mechanism has yet to be established, and the extent to which other β-blockers are chemopreventive remains relatively unknown. A comparative pharmacological approach was utilized with the expectation that a mechanism of action could be devised. JB6 Cl 41-5a (JB6 P+) murine epidermal cells were used to elucidate the chemopreventative properties of β-blockers, as JB6 P+ cells recapitulate in vivo tumor promotion and chemoprevention. The initial hypothesis was that β-blockers that are GRK/β-arrestin biased agonists, like carvedilol, are chemopreventive. Sixteen β-blockers of different classes, isoproterenol, and HEAT HCl were individually co-administered with epidermal growth factor (EGF) to JB6 P+ cells to examine the chemopreventative properties of each ligand. Cytotoxicity was examined to ensure that the anti-transformation effects of each ligand were not due to cellular growth inhibition. Many of the examined β-blockers suppressed EGF-induced JB6 P+ cell transformation in a non-cytotoxic and concentration-dependent manner. However, the IC50 values are high for the most potent inhibitors (243, 326, and 431 nM for carvedilol, labetalol, and alprenolol, respectively) and there is no correlation between pharmacological properties and inhibition of transformation. Therefore, the role of α1- and β2-adrenergic receptors (AR) was examined by standard competition assays and shRNA targeting β2-ARs, the only β-AR expressed in JB6 P+ cells. The results reveal that pharmacological inhibition of α1- and β2-ARs and genetic knockdown of β2-ARs did not abrogate carvedilol-mediated inhibition of EGF-induced JB6 P+ cell transformation. Furthermore, topical administration of carvedilol protected mice from UV-induced skin damage, while genetic ablation of β2-ARs increased carvedilol-mediated effects. Therefore, the prevailing hypothesis that the chemopreventive property of carvedilol is mediated through β-ARs is not supported by this data.

    Topics: Adrenergic beta-Antagonists; Alprenolol; Animals; Carvedilol; Cell Line; Cell Proliferation; Cell Transformation, Neoplastic; Epidermal Growth Factor; Inhibitory Concentration 50; Labetalol; Ligands; Mice; Mice, Inbred C57BL; Receptors, Adrenergic; Receptors, Adrenergic, beta; RNA, Small Interfering; Signal Transduction; Skin; Skin Neoplasms; Ultraviolet Rays

2019
β-Catenin nuclear localization positively feeds back on EGF/EGFR-attenuated AJAP1 expression in breast cancer.
    Journal of experimental & clinical cancer research : CR, 2019, Jun-06, Volume: 38, Issue:1

    Adherent junction associated protein 1 (AJAP1), a typical molecule of adherent junctions, has been found to be a tumor suppressor in many cancer types. Aberrant activation of β-catenin has been demonstrated to be associated with malignant biological properties of tumors including breast cancer. This study aimed to investigate the function and mechanism of AJAP1-mediated β-catenin activity of breast cancer lines in vitro and in breast cancer patients.. AJAP1 and β-catenin expressions in breast cancer tissues and cell lines were detected by immunohistochemistry, western blotting and qRT-PCR. The EGF/EGFR axis-mediated AJAP1 attenuated β-catenin nuclear location was measured by western blotting, immunofluorescence assay, co-immunoprecipitation, luciferase assay and ubiquitination assays. Furthermore, the function of AJAP1 and β-catenin regulated breast cancer progression was explored both in vivo and in vitro.. It was found that AJAP1 had a high negative correlation with β-catenin nuclear expression and was a novel tumor suppressor in breast cancer. AJAP1 loss can mediate β-catenin accumulated in cytoplasm and then transferred it to the nucleus, activating β-catenin transcriptional activity and downstream genes. Additionally, β-catenin can reverse the invasion, proliferation ability and tumorigenicity of the depletion of AJAP1 caused both in vivo and in vitro. Besides, EGF/EGFR also involved in the process of AJAP1-depiction induced β-catenin transactivation to the nucleus. More importantly, EGFR depletion/AJAP1 knocked down promoted the progression of breast cancer by regulating the activity of β-catenin nuclear transactivation.. This study demonstrated that AJAP1 acted as a putative tumor suppressor while β-catenin nuclear localization positively fed back on EGF/EGFR-attenuated AJAP1 expression in breast cancer, which might be beneficial to develop new therapeutic targets for decreasing nuclear β-catenin-mediated malignancy in breast cancer.

    Topics: Adult; Aged; Animals; beta Catenin; Breast Neoplasms; Case-Control Studies; Cell Adhesion Molecules; Cell Line, Tumor; Cell Proliferation; Cell Transformation, Neoplastic; Disease Models, Animal; Epidermal Growth Factor; ErbB Receptors; Female; Gene Expression Regulation, Neoplastic; Heterografts; Humans; Immunohistochemistry; Kaplan-Meier Estimate; Mice; Middle Aged; Models, Biological; Neoplasm Grading; Neoplasm Staging; Prognosis; Protein Binding

2019
The EGF/hnRNP Q1 axis is involved in tumorigenesis via the regulation of cell cycle-related genes.
    Experimental & molecular medicine, 2018, 06-06, Volume: 50, Issue:6

    Heterogeneous nuclear ribonucleoprotein (hnRNP) Q1, an RNA-binding protein, has been implicated in many post-transcriptional processes, including RNA metabolism and mRNA splicing and translation. However, the role of hnRNP Q1 in tumorigenesis remains unclear. We previously performed RNA immunoprecipitation (RIP)-seq analysis to identify hnRNP Q1-interacting mRNAs and found that hnRNP Q1 targets a group of genes that are involved in mitotic regulation, including Aurora-A. Here, we demonstrate that altering the hnRNP Q1 level influences the expression of the Aurora-A protein, but not its mRNA. Stimulation with epidermal growth factor (EGF) enhances both binding between hnRNP Q1 and Aurora-A mRNA as well as the efficacy of the hnRNP Q1-induced translation of Aurora-A mRNA. The EGF/hnRNP Q1-induced translation of Aurora-A mRNA is mediated by the mTOR and ERK pathways. In addition, we show that hnRNP Q1 up-regulates the translation of a group of spindle assembly checkpoint (SAC) genes. hnRNP Q1 overexpression is positively correlated with the levels of Aurora-A and the SAC genes in human colorectal cancer tissues. In summary, our data suggest that hnRNP Q1 plays an important role in regulating the expression of a group of cell cycle-related genes. Therefore, it may contribute to tumorigenesis by up-regulating the translation of these genes in colorectal cancer.

    Topics: Cell Transformation, Neoplastic; Colorectal Neoplasms; Epidermal Growth Factor; HCT116 Cells; Heterogeneous-Nuclear Ribonucleoproteins; Humans; MAP Kinase Signaling System; Mitosis; Neoplasm Proteins

2018
High CD90 (THY-1) expression positively correlates with cell transformation and worse prognosis in basal-like breast cancer tumors.
    PloS one, 2018, Volume: 13, Issue:6

    Breast cancer is the most prevalent cancer among women, with the basal-like triple negative (TNBC) being the most agressive one, displaying the poorest prognosis within the ductal carcinoma subtype. Due to the lack of adequate molecular targets, the diagnosis and treatment of patients with the TNBC phenotype has been a great challenge. In a previous work, we identified CD90/Thy-1 as being highly expressed in the aggressive high malignancy grade Hs578T basal-like breast tumor cell line, pointing to this molecule as a promising breast tumor marker, which should be further investigated. Here, CD90 expression was analyzed in human breast cancer samples and its functional role was investigated to better assess the oncogenic nature of CD90 in mammary cells. Quantification of CD90 expression in human breast cancer samples, by tissue microarray, showed that high CD90 positivity correlates with metastasis and poor patient survival in the basal-like subtype. The functional genetic approach, by overexpression in the CD90 cDNA in a basal-like normal mammary cell line (MCF10A) and knockdown in a highly malignant cell line (Hs578T), allowed us to demonstrate that CD90 is involved with several cellular processes that lead to malignant transformation, such as: morphological change, increased cell proliferation, invasiveness, metastasis and activation of the EGFR pathway. Therefore, our results reveal that CD90 is involved with malignant transformation in breast cancer cell lines and is correlated with metastasis and poor patient survival in the basal-like subtype, being considered as a promising new breast cancer target.

    Topics: Animals; Biomarkers, Tumor; Brazil; Cell Line, Tumor; Cell Movement; Cell Transformation, Neoplastic; Epidermal Growth Factor; Epithelial-Mesenchymal Transition; Female; Fluorescent Antibody Technique; Gene Amplification; Gene Expression; Gene Expression Profiling; Humans; Immunohistochemistry; Kaplan-Meier Estimate; Neoplasms, Basal Cell; Prognosis; Rats; Signal Transduction; Thy-1 Antigens; Tissue Array Analysis

2018
Exposure to EGF and 17β‑estradiol irreversibly affects the proliferation and transformation of MCF7 cells but is not sufficient to promote tumor growth in a xenograft mouse model upon withdrawal of exposure.
    International journal of molecular medicine, 2018, Volume: 42, Issue:3

    Epidermal growth factor (EGF) and estrogen are potent regulators of breast tumorigenesis. Their short‑term actions on human breast epithelial cells have been investigated extensively. However, the consequence of a long‑term exposure to EGF and estrogen remains to be fully elucidated. The present study examined the effects of long‑term exposure to EGF and 17β‑estradiol on the proliferation, transformation, expression of markers of stemness, and tumorigenesis of MCF7 human breast adenocarcinoma cells. Exposure to EGF and/or 17β‑estradiol irreversibly enhanced the proliferation rate of MCF7 cells, even following withdrawal. However, in a mouse xenograft experiment, no significant difference in tumor volume was observed between tumors derived from cells exposed to EGF, 17β‑estradiol or EGF + 17β‑estradiol. Immunohistochemistry performed on tumors derived from 17β‑estradiol‑exposed cells revealed reduced cell proliferation and vessel scores, according to the results obtained using Ki67 and von Willebrand factor staining, respectively. The EGF‑ and/or 17β‑estradiol‑treated cells exhibited an increased ratio of cluster of differentiation (CD)44+/CD24‑ cells and enhanced ability to form mammospheres. Furthermore, the long‑term exposure of MCF7 cells to EGF and 17β‑estradiol altered their responsiveness to short‑term stimulatory or inhibitory treatments with EGF, 17β‑estradiol, transforming growth factor‑β1 (TGFβ1), Iressa and SB431542. Therefore, the findings indicated that sustained exposure of MCF7 cells to EGF and/or 17β‑estradiol resulted in enhanced cell proliferation and mammosphere formation, an increased ratio of CD44+/CD24‑ cells, and altered responses to short‑term treatments with EGF, 17β‑estradiol, TGFβ1, and drugs inhibiting these signaling pathways. However, this sustained exposure was not sufficient to affect tumor take or volume in a xenograft mouse model.

    Topics: Animals; Benzamides; Carcinogenesis; Cell Proliferation; Cell Transformation, Neoplastic; Dioxoles; Epidermal Growth Factor; Estradiol; Female; Gefitinib; Humans; MCF-7 Cells; Mice, SCID; Models, Biological; Neoplastic Stem Cells; Phenotype; Quinazolines; Spheroids, Cellular; Tamoxifen; Xenograft Model Antitumor Assays

2018
Aloe emodin suppresses EGF‑induced neoplastic cell transformation by inhibiting the ERK/MSK1 and AKT/GSK3β signaling pathways.
    Molecular medicine reports, 2018, Volume: 18, Issue:6

    Natural compounds which can block cell transformation due to potential for chemoprevention have received increased attention. The present study aimed to investigate whether aloe emodin, which is present in aloe latex or the roots of the Rheum palmatum L. are able to block epidermal growth factor (EGF)‑ and tissue plasminogen activator‑induced JB6 C141 cell transformation. The aloe emodin treatment was applied to the JB6 C141 cell neoplastic model. The toxicity of aloe emodin was determined. The present study detected the expression level of AKT serine/threonine kinase 1 (AKT), lysine‑tRNA ligase MSK1 (MSK1) and cyclin D1 using western blotting. The cell proliferation and cell cycle distribution were also monitored. And when 95‑maximal effective dose ranged between 1 and 15 µM, the cell death was evident. Aloe emodin‑treated cells had an impaired anchorage‑independent growth capability, leading to a dose‑dependent reduction of colony formation. Western blotting revealed that aloe emodin had a significant effect on phosphorylation of pyruvate dehydrogenase kinase 1 and glycogen synthase kinase 3β (GSK3β) and AKT was inhibited. The present study determined that the proliferation of JB6 C141 cells was reduced in a dose‑dependent manner and the effect may be associated with its inhibition of the G1/S cell cycle transition. Cyclin D1 transcriptional activity was reduced to 25%, 24 h following aloe emodin treatment. The protein expression of cyclin D1 was inhibited. The findings of the present study indicated that aloe emodin may be able to suppress neoplastic cell transformation by inhibiting the extracellular‑signal regulated kinase/MSK1 and AKT/GSK3β signaling pathways. It may be a potential natural compound for chemoprevention.

    Topics: Anthraquinones; Antineoplastic Agents; Cell Transformation, Neoplastic; Cyclin D1; Dose-Response Relationship, Drug; Epidermal Growth Factor; Extracellular Signal-Regulated MAP Kinases; Gene Expression Regulation; Glycogen Synthase Kinase 3 beta; Proto-Oncogene Proteins c-akt; Ribosomal Protein S6 Kinases, 90-kDa; Signal Transduction

2018
Carcinogenic epithelial-mesenchymal transition initiated by oral cancer exosomes is inhibited by anti-EGFR antibody cetuximab.
    Oral oncology, 2018, Volume: 86

    Overexpression and increased signaling from the epidermal growth factor receptor (EGFR) often changes oral squamous cell carcinoma (OSCC) and thus EGFR is frequently targeted molecularly by the therapeutic antibody cetuximab. We assessed the roles of OSCC-derived extracellular vesicles (EVs), including exosomes in the trafficking of cetuximab and in epithelial-mesenchymal transition (EMT) of epithelial cells. OSCC cells abundantly expressed EGFR, which was secreted from cells with OSCC-EVs upon EGF stimulations. The OSCC-EGFR-EVs were then able to enter into and transform epithelial cells leading to increased mesenchymal traits with increased vimentin and spindle-like shapes. EGF priming of OSCC cells further increased this EMT-initiating effect of the OSCC-EVs. The internalization and pro-EMT effects of the OSCC-EVs were largely blocked by cetuximab. Thus, OSCC-derived EVs transform normal epithelial cells into a mesenchymal phenotype and anti-EGFR therapeutic antibody cetuximab inhibits such a carcinogenic effect of the OSCC-EVs.

    Topics: Cell Line, Tumor; Cell Transformation, Neoplastic; Cetuximab; Epidermal Growth Factor; Epithelial Cells; Epithelial-Mesenchymal Transition; ErbB Receptors; Exosomes; Humans; Mouth Neoplasms; Squamous Cell Carcinoma of Head and Neck

2018
EGF hijacks miR-198/FSTL1 wound-healing switch and steers a two-pronged pathway toward metastasis.
    The Journal of experimental medicine, 2017, Oct-02, Volume: 214, Issue:10

    Epithelial carcinomas are well known to activate a prolonged wound-healing program that promotes malignant transformation. Wound closure requires the activation of keratinocyte migration via a dual-state molecular switch. This switch involves production of either the anti-migratory microRNA miR-198 or the pro-migratory follistatin-like 1 (FSTL1) protein from a single transcript; miR-198 expression in healthy skin is down-regulated in favor of FSTL1 upon wounding, which enhances keratinocyte migration and promotes re-epithelialization. Here, we reveal a defective molecular switch in head and neck squamous cell carcinoma (HNSCC). This defect shuts off miR-198 expression in favor of sustained FSTL1 translation, driving metastasis through dual parallel pathways involving DIAPH1 and FSTL1. DIAPH1, a miR-198 target, enhances directional migration through sequestration of Arpin, a competitive inhibitor of Arp2/3 complex. FSTL1 blocks Wnt7a-mediated repression of extracellular signal-regulated kinase phosphorylation, enabling production of MMP9, which degrades the extracellular matrix and facilitates metastasis. The prognostic significance of the FSTL1-DIAPH1 gene pair makes it an attractive target for therapeutic intervention.

    Topics: Animals; Blotting, Western; Carcinoma, Squamous Cell; Cell Proliferation; Cell Transformation, Neoplastic; Epidermal Growth Factor; Female; Follistatin-Related Proteins; Genes, Switch; Head and Neck Neoplasms; Immunoprecipitation; Mass Spectrometry; Mice, Inbred NOD; MicroRNAs; Wound Healing

2017
Neuroblastoma patient-derived xenograft cells cultured in stem-cell promoting medium retain tumorigenic and metastatic capacities but differentiate in serum.
    Scientific reports, 2017, 08-31, Volume: 7, Issue:1

    Cultured cancer cells serve as important models for preclinical testing of anti-cancer compounds. However, the optimal conditions for retaining original tumor features during in vitro culturing of cancer cells have not been investigated in detail. Here we show that serum-free conditions are critical for maintaining an immature phenotype of neuroblastoma cells isolated from orthotopic patient-derived xenografts (PDXs). PDX cells could be grown either as spheres or adherent on laminin in serum-free conditions with retained patient-specific genomic aberrations as well as tumorigenic and metastatic capabilities. However, addition of serum led to morphological changes, neuronal differentiation and reduced cell proliferation. The epidermal growth factor (EGF) and basic fibroblast growth factor (bFGF) were central for PDX cell proliferation and MYCN expression, and also hindered the serum-induced differentiation. Although serum induced a robust expression of neurotrophin receptors, stimulation with their cognate ligands did not induce further sympathetic differentiation, which likely reflects a block in PDX cell differentiation capacity coupled to their tumor genotype. Finally, PDX cells cultured as spheres or adherent on laminin responded similarly to various cytotoxic drugs, suggesting that both conditions are suitable in vitro screening models for neuroblastoma-targeting compounds.

    Topics: Animals; Biomarkers, Tumor; Biopsy; Cell Differentiation; Cell Line, Tumor; Cell Movement; Cell Proliferation; Cell Transformation, Neoplastic; Culture Media, Conditioned; Disease Models, Animal; Epidermal Growth Factor; Fibroblast Growth Factor 2; Heterografts; Humans; Immunohistochemistry; Mice; N-Myc Proto-Oncogene Protein; Neoplasm Metastasis; Neural Stem Cells; Neuroblastoma

2017
Novel role of cannabinoid receptor 2 in inhibiting EGF/EGFR and IGF-I/IGF-IR pathways in breast cancer.
    Oncotarget, 2017, May-02, Volume: 8, Issue:18

    Breast cancer is the second leading cause of cancer deaths among women. Cannabinoid receptor 2 (CNR2 or CB2) is an integral part of the endocannabinoid system. Although CNR2 is highly expressed in the breast cancer tissues as well as breast cancer cell lines, its functional role in breast tumorigenesis is not well understood. We observed that estrogen receptor-α negative (ERα-) breast cancer cells highly express epidermal growth factor receptor (EGFR) as well as insulin-like growth factor-I receptor (IGF-IR). We also observed IGF-IR upregulation in ERα+ breast cancer cells. In addition, we found that higher CNR2 expression correlates with better recurrence free survival in ERα- and ERα+ breast cancer patients. Therefore, we analyzed the role of CNR2 specific agonist (JWH-015) on EGF and/or IGF-I-induced tumorigenic events in ERα- and ERα+ breast cancers. Our studies showed that CNR2 activation inhibited EGF and IGF-I-induced migration and invasion of ERα+ and ERα- breast cancer cells. At the molecular level, JWH-015 inhibited EGFR and IGF-IR activation and their downstream targets STAT3, AKT, ERK, NF-kB and matrix metalloproteinases (MMPs). In vivo studies showed that JWH-015 significantly reduced breast cancer growth in ERα+ and ERα- breast cancer mouse models. Furthermore, we found that the tumors derived from JWH-015-treated mice showed reduced activation of EGFR and IGF-IR and their downstream targets. In conclusion, we show that CNR2 activation suppresses breast cancer through novel mechanisms by inhibiting EGF/EGFR and IGF-I/IGF-IR signaling axes.

    Topics: Animals; Breast Neoplasms; Cell Line, Tumor; Cell Transformation, Neoplastic; Disease Models, Animal; Epidermal Growth Factor; ErbB Receptors; Estrogen Receptor alpha; Female; Gene Expression; Heterografts; Humans; Indoles; Insulin-Like Growth Factor I; Mice; Prognosis; Receptor, Cannabinoid, CB2; Receptor, IGF Type 1; Signal Transduction

2017
Gefitinib enhances radiotherapeutic effects of
    Nuclear medicine and biology, 2017, Volume: 48

    Gefitinib is an epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor which has been proven effective for cancer treatment. In this study, we sought to determine whether gefitinib could increase the in vivo tumor uptake of human

    Topics: Animals; Carcinoma, Squamous Cell; Cell Line, Tumor; Cell Transformation, Neoplastic; Epidermal Growth Factor; ErbB Receptors; Gefitinib; Gene Expression Regulation, Neoplastic; Head and Neck Neoplasms; Humans; Iodine Radioisotopes; Mice; Mice, Nude; Protein Transport; Quinazolines; Radiation Tolerance; Radiochemistry; Tissue Distribution

2017
Cyclin-dependent kinase 2 (CDK2) is a key mediator for EGF-induced cell transformation mediated through the ELK4/c-Fos signaling pathway.
    Oncogene, 2016, Mar-03, Volume: 35, Issue:9

    Cyclin-dependent kinase 2 (CDK2) is a known regulator in the cell cycle control of the G1/S and S/G2 transitions. However, the role of CDK2 in tumorigenesis is controversial. Evidence from knockout mice as well as colon cancer cell lines indicated that CDK2 is dispensable for cell proliferation. In this study, we found that ectopic CDK2 enhances Ras (G12V)-induced foci formation and knocking down CDK2 expression markedly decreases epidermal growth factor (EGF)-induced cell transformation mediated through the downregulation of c-fos expression. Interestingly, CDK2 directly phosphorylates ELK4 at Thr194 and Ser387 and regulates the ELK4 transcriptional activity, which serves as a mechanism to regulate c-fos expression. In addition, ELK4 is overexpressed in melanoma and knocking down the ELK4 or CDK2 expression significantly attenuated the malignant phenotype of melanoma cells. Taken together, our study reveals a novel function of CDK2 in EGF-induced cell transformation and the associated signal transduction pathways. This indicates that CDK2 is a useful molecular target for the chemoprevention and therapy against skin cancer.

    Topics: Animals; Cell Cycle; Cell Line; Cell Proliferation; Cell Transformation, Neoplastic; Cyclin-Dependent Kinase 2; Epidermal Growth Factor; ets-Domain Protein Elk-4; Humans; Melanoma; Mice; Phosphorylation; Proto-Oncogene Proteins c-fos; Transcriptional Activation

2016
p85α promotes nucleolin transcription and subsequently enhances EGFR mRNA stability and EGF-induced malignant cellular transformation.
    Oncotarget, 2016, Mar-29, Volume: 7, Issue:13

    p85α is a regulatory subunit of phosphatidylinositol 3-kinase (PI3K) that is a key lipid enzyme for generating phosphatidylinositol 3, 4, 5-trisphosphate, and subsequently activates signaling that ultimately regulates cell cycle progression, cell growth, cytoskeletal changes, and cell migration. In addition to form a complex with the p110 catalytic subunit, p85α also exists as a monomeric form due to that there is a greater abundance of p85α than p110 in many cell types. Our previous studies have demonstrated that monomeric p85α exerts a pro-apoptotic role in UV response through induction of TNF-α gene expression in PI3K-independent manner. In current studies, we identified a novel biological function of p85α as a positive regulator of epidermal growth factor receptor (EGFR) expression and cell malignant transformation via nucleolin-dependent mechanism. Our results showed that p85α was crucial for EGFR and nucleolin expression and subsequently resulted in an increase of malignant cellular transformation by using both specific knockdown and deletion of p85α in its normal expressed cells. Mechanistic studies revealed that p85α upregulated EGFR protein expression mainly through stabilizing its mRNA, whereas nucleolin (NCL) was able to bind to egfr mRNA and increase its mRNA stability. Consistently, overexpression of NCL in p85α-/- cells restored EGFR mRNA stabilization, protein expression and cell malignant transformation. Moreover, we discovered that p85α upregulated NCL gene transcription via enhancing C-Jun activation. Collectively, our studies demonstrate a novel function of p85α as a positive regulator of EGFR mRNA stability and cell malignant transformation, providing a significant insight into the understanding of biomedical nature of p85α protein in mammalian cells and further supporting that p85α might be a potential target for cancer prevention and therapy.

    Topics: Animals; Cell Transformation, Neoplastic; Cells, Cultured; Class Ia Phosphatidylinositol 3-Kinase; Embryo, Mammalian; Epidermal Growth Factor; ErbB Receptors; Fibroblasts; Gene Expression Regulation; Immunoblotting; Mice, Knockout; Nucleolin; Phosphoproteins; Reverse Transcriptase Polymerase Chain Reaction; RNA Interference; RNA Stability; RNA-Binding Proteins; Transcription, Genetic

2016
PDZ-binding kinase/T-LAK cell-originated protein kinase is a target of the fucoidan from brown alga Fucus evanescens in the prevention of EGF-induced neoplastic cell transformation and colon cancer growth.
    Oncotarget, 2016, Apr-05, Volume: 7, Issue:14

    The fucoidan with high anticancer activity was isolated from brown alga Fucus evanescens. The compound effectively prevented EGF-induced neoplastic cell transformation through inhibition of TOPK/ERK1/2/MSK 1 signaling axis. In vitro studies showed that the fucoidan attenuated mitogen-activated protein kinases downstream signaling in a colon cancer cells with different expression level of TOPK, resulting in growth inhibition. The fucoidan exerts its effects by directly interacting with TOPK kinase in vitro and ex vivo and inhibits its kinase activity. In xenograft animal model, oral administration of the fucoidan suppressed HCT 116 colon tumor growth. The phosphorylation of TOPK downstream signaling molecules in tumor tissues was also inhibited by the fucoidan. Taken together, our findings support the cancer preventive efficacy of the fucoidan through its targeting of TOPK for the prevention of neoplastic cell transformation and progression of colon carcinomas in vitro and ex vivo.

    Topics: Animals; Cell Line, Tumor; Cell Transformation, Neoplastic; Colonic Neoplasms; Epidermal Growth Factor; Humans; Mice; Mice, Nude; Mitogen-Activated Protein Kinase Kinases; Phosphorylation; Polysaccharides; Signal Transduction; Xenograft Model Antitumor Assays

2016
Guanidine Alkaloids from the Marine Sponge Monanchora pulchra Show Cytotoxic Properties and Prevent EGF-Induced Neoplastic Transformation in Vitro.
    Marine drugs, 2016, Jul-15, Volume: 14, Issue:7

    Guanidine alkaloids from sponges Monanchora spp. represent diverse bioactive compounds, however, the mechanisms underlying bioactivity are very poorly understood. Here, we report results of studies on cytotoxic action, the ability to inhibit EGF-induced neoplastic transformation, and the effects on MAPK/AP-1 signaling of eight rare guanidine alkaloids, recently isolated from the marine sponge Monanchora pulchra, namely: monanchocidin A (1), monanchocidin B (2), monanchomycalin C (3), ptilomycalin A (4), monanchomycalin B (5), normonanchocidin D (6), urupocidin A (7), and pulchranin A (8). All of the compounds induced cell cycle arrest (apart from 8) and programmed death of cancer cells. Ptilomycalin A-like compounds 1-6 activated JNK1/2 and ERK1/2, following AP-1 activation and caused p53-independent programmed cell death. Compound 7 induced p53-independent cell death without activation of AP-1 or caspase-3/7, and the observed JNK1/2 activation did not contribute to the cytotoxic effect of the compound. Alkaloid 8 induced JNK1/2 (but not ERK1/2) activation leading to p53-independent cell death and strong suppression of AP-1 activity. Alkaloids 1-4, 7, and 8 were able to inhibit the EGF-induced neoplastic transformation of JB6 P⁺ Cl41 cells. Our results suggest that investigated guanidine marine alkaloids hold potential to eliminate human cancer cells and prevent cancer cell formation and spreading.

    Topics: Alkaloids; Animals; Cell Cycle Checkpoints; Cell Death; Cell Line; Cell Line, Tumor; Cell Transformation, Neoplastic; Epidermal Growth Factor; Guanidine; Guanidines; HeLa Cells; Humans; MAP Kinase Signaling System; Mice; Porifera; Transcription Factor AP-1; Tumor Suppressor Protein p53

2016
XIAP RING domain mediates miR-4295 expression and subsequently inhibiting p63α protein translation and promoting transformation of bladder epithelial cells.
    Oncotarget, 2016, Aug-30, Volume: 7, Issue:35

    The X-linked inhibitor of apoptosis protein (XIAP) contains three N-terminal BIR domains that mediate anti-apoptosis and one C-terminal RING finger domain whose function(s) are not fully defined. Here we show that the RING domain of XIAP strongly inhibits the expression of p63α, a known tumor suppressor. XIAP knockdown in urothelial cells or RING deletion in knockin mice markedly upregulates p63α expression. This RING-mediated p63α downregulation is critical for the malignant transformation of normal urothelial cells following EGF treatment. We further show that the RING domain promotes Sp1-mediated transcription of miR-4295 which targets the 3'UTR of p63α mRNA and consequently inhibits p63α translation. Our results reveal a previously unknown function of the RING of XIAP in promoting miR-4295 transcription, thereby reducing p63α translation and enhancing urothelial transformation. Our data offer novel insights into the multifunctional effects of the XIAP RING domain on urothelial tumorigenesis and the potential for targeting this frequently overexpressed protein as a therapeutic alternative.

    Topics: Animals; Apoptosis; Carcinogenesis; Cell Adhesion; Cell Proliferation; Cell Transformation, Neoplastic; Epidermal Growth Factor; Epithelial Cells; Gene Expression Regulation, Neoplastic; Humans; Methionine; Mice; Mice, Transgenic; MicroRNAs; Protein Biosynthesis; Protein Domains; Sp1 Transcription Factor; Transcription Factors; Tumor Suppressor Proteins; Up-Regulation; Urinary Bladder; Urinary Bladder Neoplasms; X-Linked Inhibitor of Apoptosis Protein

2016
Eupatilin inhibits EGF-induced JB6 cell transformation by targeting PI3K.
    International journal of oncology, 2016, Volume: 49, Issue:3

    Phosphatidylinositol 3-kinases (PI3Ks) are lipid kinases that play fundamental roles in regulation of multiple signaling pathways, including cell proliferation, survival and cell cycle. Increasing evidence has shown that abnormal activation of PI3K pathway contributes to tumorigenesis and progression of various malignant tumors. Therefore, it is an attractive target of chemoprevention and chemotherapy. Eupatilin, a natural flavone compound extracted from Artemisia vulgaris, has antitumor and anti-inflammation efficacy. However, the direct target(s) of eupatilin in cancer chemoprevention are still elusive. In the present study, we reported eupatilin suppressed JB6 cell proliferation and its EGF-induced colony formation. Eupatilin attenuated phosphorylation of PI3K downstream signaling molecules. Downregulation of cyclin D1 expression and arresting in G1 phase were induced through eupatilin treatment. Furthermore, we found it could bind to the p110α, a catalytic subunit of PI3K, by computational docking methods. Pull down assay outcomes also verified the binding of eupatilin with PI3K. Taken together, our results suggest that epatilin is a potential chemopreventive agent in inhibition of skin cell transformation by targeting PI3K.

    Topics: Animals; Cell Line; Cell Proliferation; Cell Transformation, Neoplastic; Class I Phosphatidylinositol 3-Kinases; Epidermal Growth Factor; Flavonoids; Gene Expression Regulation; Mice; Molecular Docking Simulation; Phosphatidylinositol 3-Kinases; Phosphorylation; Signal Transduction

2016
Prolonged overexpression of Wnt10b induces epidermal keratinocyte transformation through activating EGF pathway.
    Histochemistry and cell biology, 2015, Volume: 144, Issue:3

    Wnt10b is a signaling protein regulating skin development and homeostasis, and the expression of Wnt10b is restricted to epidermal keratinocytes in embryonic and postnatal skin. Recent studies indicate an elevated expression of Wnt10b in skin tumors. However, how Wnt10b regulates skin tumorigenesis remains largely unknown. Here we report that continuous expression of Wnt10b mediates transformation of epidermal keratinocytes through activating genes involved in EGF/MAPK signaling pathways. We first established a prolonged Wnt10b overexpression system in JB6P- cells to represent the elevated Wnt10b expression level in skin keratinocytes. Through expression assays and observations under phase-contrast microscopy, prolonged expression of Wnt10b activated Wnt/β-catenin pathway and induced morphological changes of cells showing longer protrusions and multilayer growth, indicating early-stage cell transformation. Wnt10b also increased cellular proliferation and migration according to BrdU incorporation and cell mobility assays. Furthermore, multi-doses of AdWnt10b treatment to JB6P- cells induced colony formation, stronger invasive ability in transwell system, and anchorage-independent growth in agar gel. In molecular level, AdWnt10b treatment induced increased transcriptional expressions of Egf, downstream Mapk pathway factors, and MMPs. Administration of Wnt antagonist DKK1 blocked the tumor promotion process induced by Wnt10b. Taken together, these findings clearly demonstrate that Wnt10b promotes epidermal keratinocyte transformation through induced Egf pathway.

    Topics: Antimetabolites, Antineoplastic; Bromodeoxyuridine; Cell Proliferation; Cell Transformation, Neoplastic; Epidermal Cells; Epidermal Growth Factor; Epidermis; Humans; Intercellular Signaling Peptides and Proteins; Keratinocytes; Matrix Metalloproteinases; Mitogen-Activated Protein Kinases; Proto-Oncogene Proteins; Signal Transduction; Skin Neoplasms; Tumor Stem Cell Assay; Wnt Proteins

2015
Cripto-1 as a novel therapeutic target for triple negative breast cancer.
    Oncotarget, 2015, May-20, Volume: 6, Issue:14

    Triple-negative breast cancer (TNBC) presents the poorest prognosis among the breast cancer subtypes and no current standard therapy. Here, we performed an in-depth molecular analysis of a mouse model that establishes spontaneous lung metastasis from JygMC(A) cells. These primary tumors resembled the triple-negative breast cancer (TNBC) both phenotypically and molecularly. Morphologically, primary tumors presented both epithelial and spindle-like cells but displayed only adenocarcinoma-like features in lung parenchyma. The use of laser-capture microdissection combined with Nanostring mRNA and microRNA analysis revealed overexpression of either epithelial and miRNA-200 family or mesenchymal markers in adenocarcinoma and mesenchymal regions, respectively. Cripto-1, an embryonic stem cell marker, was present in spindle-like areas and its promoter showed activity in primary tumors. Cripto-1 knockout by the CRISPR-Cas9 system inhibited tumor growth and pulmonary metastasis. Our findings show characterization of a novel mouse model that mimics the TNBC and reveal Cripto-1 as a TNBC target hence may offer alternative treatment strategies for TNBC.

    Topics: Animals; Cell Line, Tumor; Cell Transformation, Neoplastic; Epidermal Growth Factor; Epithelial-Mesenchymal Transition; Female; Fluorescent Antibody Technique; Gene Knockout Techniques; Immunohistochemistry; In Situ Nick-End Labeling; Laser Capture Microdissection; Mammary Neoplasms, Experimental; Membrane Glycoproteins; Mice; Mice, Inbred BALB C; Mice, Nude; Neoplasm Proteins; Oligonucleotide Array Sequence Analysis; Polymerase Chain Reaction; Triple Negative Breast Neoplasms

2015
TNFR1 Regulates Ovarian Cancer Cell Tumorigenicity Through PIK3CB-p110Beta.
    Current molecular medicine, 2015, Volume: 15, Issue:5

    Studies have shown that TNFR1 is a key factor in the tumor microenvironment that is dependent on the TNF-α-initiated cascade for tumorigenesis. In this present study, we found that TNFR1 is over-expressed in ovarian cancer, which is relevant to both clinical survival and disease free status. Knockdown of TNFR1 dramatically attenuates malignant phenotypes, including proliferation and colony growth in soft agar, as well as glycolysis in ovarian cancer cells. Unexpectedly, knocking down TNFR1 blocks EGF-induced p-AKT and p-p70S6K expression and EGF-induced cell transformation through PIK3-p110beta rather than p110alpha expression. Taken together, our data provide evidence that TNFR1 plays a critical role in ovarian cancer and show that the EGF induced signaling pathway is independent of the TNF-α triggering cascade signal. Therefore, TNFR1 may serve as a prognostic molecule in ovarian cancer.

    Topics: Animals; Cell Line, Tumor; Cell Transformation, Neoplastic; Class I Phosphatidylinositol 3-Kinases; Disease Models, Animal; Epidermal Growth Factor; Female; Gene Expression; Gene Knockdown Techniques; Glucose; Glycolysis; Heterografts; Humans; Immunohistochemistry; Lactic Acid; Mice; Ovarian Neoplasms; Phenotype; Phosphatidylinositol 3-Kinases; Prognosis; Receptors, Tumor Necrosis Factor, Type I; Signal Transduction; Tumor Burden

2015
The Role of the Pleckstrin Homology Domain-containing Protein CKIP-1 in Activation of p21-activated Kinase 1 (PAK1).
    The Journal of biological chemistry, 2015, Aug-21, Volume: 290, Issue:34

    Upon growth factor stimulation, PAK1 is recruited to the plasma membrane and activated by a mechanism that requires its phosphorylation at Ser-223 by the protein kinase CK2. However, the upstream signaling molecules that regulate this phosphorylation event are not clearly defined. Here, we demonstrate a major role of the CK2α-interacting protein CKIP-1 in activation of PAK1. CK2α, CKIP-1, and PAK1 are translocated to membrane ruffles in response to the epidermal growth factor (EGF), where CKIP-1 mediates the interaction between CK2α and PAK1 in a PI3K-dependent manner. Consistently, PAK1 mediates phosphorylation and modulation of the activity of p41-Arc, one of its plasma membrane substrate, in a fashion that requires PI3K and CKIP-1. Moreover, CKIP-1 knockdown or PI3K inhibition suppresses PAK1-mediated cell migration and invasion, demonstrating the physiological significance of the PI3K-CKIP-1-CK2-PAK1 signaling pathway. Taken together, these findings identify a novel mechanism for the activation of PAK1 at the plasma membrane, which is critical for cell migration and invasion.

    Topics: Actin-Related Protein 2-3 Complex; Casein Kinase II; Cell Line; Cell Line, Tumor; Cell Transformation, Neoplastic; Epidermal Growth Factor; Epithelial Cells; Gene Expression Regulation, Neoplastic; Humans; Intracellular Signaling Peptides and Proteins; Male; Membrane Microdomains; p21-Activated Kinases; Phosphatidylinositol 3-Kinases; Phosphoinositide-3 Kinase Inhibitors; Phosphorylation; Prostate; Protein Kinase Inhibitors; Protein Transport; Recombinant Proteins; RNA, Small Interfering; Signal Transduction

2015
Aschantin targeting on the kinase domain of mammalian target of rapamycin suppresses epidermal growth factor-induced neoplastic cell transformation.
    Carcinogenesis, 2015, Volume: 36, Issue:10

    Mammalian target of rapamycin (mTOR), a serine/threonine protein kinase, forms two different complexes, complex 1 and 2, and plays a key role in the regulation of Akt signaling-mediated cell proliferation and transformation. This study reveals aschantin, a natural compound abundantly found in Magnolia flos, as a novel mTOR kinase inhibitor. Aschantin directly targeted the active pocket of mTOR kinase domain by competing with adenosine triphosphate (ATP), but not PI3K and PDK1. Aschantin inhibited epidermal growth factor (EGF)-induced full activation of Akt by phosphorylation at Ser473/Thr308, resulting in inhibition of the mTORC2/Akt and Akt/mTORC1/p70S6K signaling pathways and activation of GSK3β by abrogation of Akt-mediated GSK3β phosphorylation at Ser9. The activated GSK3β inhibited cell proliferation by c-Jun phosphorylation at Ser243, which facilitated destabilization and degradation of c-Jun through the ubiquitination-mediated proteasomal degradation pathway. Notably, aschantin treatment decreased c-Jun stability through inhibition of the mTORC2-Akt signaling pathway, which suppressed EGF-induced anchorage-independent cell transformation in non-malignant JB6 Cl41 and HaCaT cells and colony growth of LNCaP and MIAPaCa-2 cancer cells in soft agar. Altogether, the results show that aschantin targets mTOR kinase and destabilizes c-Jun, which implicate aschantin as a potential chemopreventive or therapeutic agent.

    Topics: Animals; Benzodioxoles; Cell Line, Tumor; Cell Proliferation; Cell Transformation, Neoplastic; Epidermal Growth Factor; Gene Expression Regulation, Neoplastic; Glycogen Synthase Kinase 3; Glycogen Synthase Kinase 3 beta; Humans; Lignans; Mechanistic Target of Rapamycin Complex 1; Mechanistic Target of Rapamycin Complex 2; Mice; Multiprotein Complexes; Phosphorylation; Proto-Oncogene Proteins c-akt; Signal Transduction; TOR Serine-Threonine Kinases

2015
Dipeptidyl peptidase 4 promotes epithelial cell transformation and breast tumourigenesis via induction of PIN1 gene expression.
    British journal of pharmacology, 2015, Volume: 172, Issue:21

    Dipeptidyl peptidase 4 (DPP4) is an aminopeptidase that is widely expressed in different cell types. Recent studies suggested that DPP4 plays an important role in tumour progression in several human malignancies. Here we have examined the mechanisms by which up-regulation of DPP4 expression causes epithelial transformation and mammary tumourigenesis.. Expression of DPP4 and the peptidylprolyl cis/trans isomerase, NIMA-interacting 1 (PIN1), and the cytotoxic effects of combined treatment with sitagliptin and juglone were investigated by immunohistochemistry, immunoblotting, real-time PCR, TUNEL and soft agar assays, using MCF7 cells. The effects of sitagliptin on tumour development in vivo were studied in the syngeneic 4T1 metastatic breast cancer model.. Activity of the transcription factor E2F1 induced by EGF was enhanced by DPP4, thus increasing PIN1 expression. Furthermore, DPP4 enhanced MEK/ERK and JNK/c-Jun signalling induced by EGF, inducing AP-1 activity and epithelial cell transformation. In contrast, DPP4 silencing or DPP4 inhibition in MCF7 cells inhibited PIN1 expression via E2F1 activity induced by EGF, decreasing colony formation and inducing DNA fragmentation. In the syngeneic 4T1 metastatic breast cancer model, DPP4 overexpression increased tumour development, whereas treatment with sitagliptin and/or juglone suppressed it. Consistent with these observations, DPP4 levels were positively correlated with PIN1 expression in human breast cancer.. DPP4 promoted EGF-induced epithelial cell transformation and mammary tumourigenesis via induction of PIN1 expression, suggesting that sitagliptin targeting of DPP4 could be a treatment strategy in patients with breast cancer.

    Topics: Animals; Apoptosis; Breast Neoplasms; Cell Transformation, Neoplastic; Cyclin D1; Dipeptidyl Peptidase 4; Epidermal Growth Factor; Female; Humans; MCF-7 Cells; Mice; Mice, Inbred BALB C; NIMA-Interacting Peptidylprolyl Isomerase; Peptidylprolyl Isomerase; Protein Kinases; Signal Transduction; Sitagliptin Phosphate; Transcription Factor AP-1; Up-Regulation

2015
Increased Notch Signaling Enhances Radioresistance of Malignant Stromal Cells Induced by Glioma Stem/ Progenitor Cells.
    PloS one, 2015, Volume: 10, Issue:11

    Host malignant stromal cells induced by glioma stem/progenitor cells were revealed to be more radiation-resistant than the glioma stem/progenitor cells themselves after malignant transformation in nude mice. However, the mechanism underlying this phenomenon remains unclear.. Malignant stromal cells induced by glioma stem/progenitor cell 2 (GSC-induced host brain tumor cells, ihBTC2) were isolated and identified from the double color-coded orthotopic glioma nude mouse model. The survival fraction at 2 Gy (SF2) was used to evaluate the radiation resistance of ihBTC2, the human glioma stem/progenitor cell line SU3 and its radiation-resistant sub-strain SU3-5R and the rat C6 glioma cell line. The mRNA of Notch 1 and Hes1 from ihBTC2 cells were detected using qPCR before and after 4 Gy radiation. The expression of the Notch 1, pAkt and Bcl-2 proteins were investigated by Western blot. To confirm the role of the Notch pathway in the radiation resistance of ihBTC2, Notch signaling blocker gamma secretase inhibitors (GSIs) were used.. The ihBTC2 cells had malignant phenotypes, such as infinite proliferation, hyperpentaploid karyotype, tumorigenesis in nude mice and expression of protein markers of oligodendroglia cells. The SF2 of ihBTC2 cells was significantly higher than that of any other cell line (P<0.05, n = 3). The expression of Notch 1 and Hes1 mRNAs from ihBTC2 cells was significantly increased after radiation. Moreover, the Notch 1, pAkt and Bcl-2 proteins were significantly increased after radiation (P<0.05, n = 3). Inhibition of Notch signaling markedly enhanced the radiosensitivity of ihBTC2 cells.. In an orthotopic glioma model, the malignant transformation of host stromal cells was induced by glioma stem/progenitor cells. IhBTC2 cells are more radiation-resistant than the glioma stem/progenitor cells, which may be mediated by activation of the Notch signaling pathway.

    Topics: Animals; Brain Neoplasms; Cell Line, Tumor; Cell Proliferation; Cell Transformation, Neoplastic; Epidermal Growth Factor; Glioma; Humans; Karyotyping; Mice; Mice, Nude; Neoplastic Stem Cells; Phenotype; Proto-Oncogene Proteins c-akt; Proto-Oncogene Proteins c-bcl-2; Radiation Tolerance; Rats; Receptor, Notch1; Signal Transduction

2015
The P110 subunit of PI3-K is a therapeutic target of acacetin in skin cancer.
    Carcinogenesis, 2014, Volume: 35, Issue:1

    The identification of primary molecular targets of cancer-preventive phytochemicals is essential for a comprehensive understanding of their mechanism of action. In the present study, we investigated the chemopreventive effects and molecular targets of acacetin, a flavonoid found in Robinia p seudoacacia, also known as black locust. Acacetin treatment significantly suppressed epidermal growth factor (EGF)-induced cell transformation. Immunoblot analysis revealed that acacetin attenuated EGF-induced phosphorylation of Akt and p70(S6K), which are downstream effectors of phosphatidylinositol 3-kinase (PI3-K). An immunoprecipitation kinase assay of PI3-K and pull-down assay results demonstrated that acacetin substantially inhibits PI3-K activity by direct physical binding. Acacetin exhibited stronger inhibitory effects against anchorage-dependent and -independent cell growth in cells expressing higher PI3-K activity compared with those exhibiting relatively low PI3-K activity. Binding assay data combined with computational modeling suggest that acacetin binds in an adenosine triphosphate (ATP)-competitive manner with the p110α subunit of PI3-K and interacts with Val828, Glu826, Asp911, Trp760, Ile777, Ile825, Tyr813, Ile910 and Met900 residues. Acacetin was also found to significantly reduce SK-MEL-28 tumor growth and Akt phosphorylation in vivo. Taken together, these results indicate that acacetin is an ATP-competitive PI3-K inhibitor and a promising agent for melanoma chemoprevention.

    Topics: Adenosine Triphosphate; Animals; Antineoplastic Agents, Phytogenic; Cell Proliferation; Cell Transformation, Neoplastic; Class Ia Phosphatidylinositol 3-Kinase; Enzyme Inhibitors; Epidermal Growth Factor; Flavones; Mice; Mice, Nude; Molecular Docking Simulation; Phosphatidylinositol 3-Kinases; Phosphoinositide-3 Kinase Inhibitors; Phosphorylation; Proto-Oncogene Proteins c-akt; Skin Neoplasms; Tumor Cells, Cultured; Xenograft Model Antitumor Assays

2014
Prolyl-isomerase Pin1 impairs trastuzumab sensitivity by up-regulating fatty acid synthase expression.
    Anticancer research, 2014, Volume: 34, Issue:3

    Clinical trials have shown efficacy of the anti-HER2 monoclonal antibody trastuzumab in metastatic breast cancer patients. The aim of the present study was to elucidate the mechanisms by which up-regulation of fatty acid synthase (FAS) expression confers resistance to trastuzumab in HER2-positive breast cancers.. The expression of FAS as well as the cytotoxic effects of combinatorial treatment of trastuzumab and juglone was investigated by immunoblotting, BrdU incorporation, TUNEL assay, and soft agar assay.. Pin1 enhanced EGF-induced SREBP1c promoter activity, resulting in the induction of FAS expression in BT474 cells. In contrast, juglone, a potent Pin1 inhibitor, significantly enhanced trastuzumab-induced FAS down-regulation and cell death in BT474 cells. Furthermore, trastuzumab, when used in combination with gene silencing or chemical inhibition of Pin1, increased cleaved poly(ADP-ribose) polymerase and DNA fragmentation to increase trastuzumab sensitivity.. Pin1-mediated FAS overexpression is a major regulator of trastuzumab-resistant breast cancer growth and survival.

    Topics: Animals; Antibodies, Monoclonal, Humanized; Antineoplastic Agents; Apoptosis; Blotting, Western; Breast Neoplasms; Cell Adhesion; Cell Movement; Cell Proliferation; Cell Transformation, Neoplastic; Epidermal Growth Factor; Fatty Acid Synthases; Female; Gene Expression Regulation, Neoplastic; Humans; Mice; Mice, Inbred BALB C; Mice, Nude; NIMA-Interacting Peptidylprolyl Isomerase; Peptidylprolyl Isomerase; Promoter Regions, Genetic; RNA, Small Interfering; Sterol Regulatory Element Binding Protein 1; Trastuzumab; Tumor Cells, Cultured; Xenograft Model Antitumor Assays

2014
Epidermal growth factor/heat shock protein 27 pathway regulates vasculogenic mimicry activity of breast cancer stem/progenitor cells.
    Biochimie, 2014, Volume: 104

    Tumor vascularization, which is mainly contributed by angiogenesis and vascularization, is necessary for tumor maintenance and progression. Vasculogenic mimicry (VM), vascular-like channels which are lack of the involvement of endothelial cells, has been observed in aggressive cancers and also involves in tumor vascularization. Breast cancer stem/progenitor cells (BCSCs) have been identified as a subpopulation of breast cancer cells with markers of CD24(-)CD44(+), high aldehyde dehydrogenase activity (ALDH(+)) or could be enriched by mammosphere cultivation. These cells have been proven to be associated with tumor vascularization. Here we investigated the molecular mechanisms in VM activity of BCSCs. By periodic acid-Schiff or hematoxylin-eosin stain, we found that there were VM structures in two xenografted human breast cancer tissues established from CD24(-)CD44(+) or ALDH(+) cells. Only ALDH(+) or mammosphere-forming BCSCs could form tube structures on matrigel-coated surface as similar as microvascular endothelial cells. Inhibition of the phosphorylation of epidermal growth factor receptor (EGFR) by gefitinib or knockdown of EGFR by lentiviral shRNA abolished the in vitro VM activity of BCSCs. By quercetin treatment, a plant flavonoid compound which is known to suppress heat shock proteins, or siRNA-mediated gene silencing, both Hsp27 expression and VM capability of BCSCs were suppressed. Forced expression of phosphor-mimic form of Hsp27 in ALDH(+) BCSCs could overcome the inhibitory effect of gefitinib. In conclusion, our data demonstrate that VM activity of BCSCs is mediated by EGF/Hsp27 signaling and targeting this pathway may benefit to breast cancer therapy.

    Topics: Animals; Breast Neoplasms; Cell Line, Tumor; Cell Transformation, Neoplastic; Epidermal Growth Factor; Female; HSP27 Heat-Shock Proteins; Humans; Mice; Neoplastic Stem Cells; Neovascularization, Pathologic; Phosphorylation; Signal Transduction

2014
The MEKK1 PHD ubiquitinates TAB1 to activate MAPKs in response to cytokines.
    The EMBO journal, 2014, Nov-03, Volume: 33, Issue:21

    Unlike the other MAP3Ks, MEKK1 (encoded by Map3k1) contains a PHD motif. To understand the role of this motif, we have created a knockin mutant of mouse Map3k1 (Map3k1(m) (PHD)) with an inactive PHD motif. Map3k1(m) (PHD) ES cells demonstrate that the MEKK1 PHD controls p38 and JNK activation during TGF-β, EGF and microtubule disruption signalling, but does not affect MAPK responses to hyperosmotic stress. Protein microarray profiling identified the adaptor TAB1 as a PHD substrate, and TGF-β- or EGF-stimulated Map3k1(m) (PHD) ES cells exhibit defective non-canonical ubiquitination of MEKK1 and TAB1. The MEKK1 PHD binds and mediates the transfer of Lys63-linked poly-Ub, using the conjugating enzyme UBE2N, onto TAB1 to regulate TAK1 and MAPK activation by TGF-β and EGF. Both the MEKK1 PHD and TAB1 are critical for ES-cell differentiation and tumourigenesis. Map3k1(m) (PHD) (/+) mice exhibit aberrant cardiac tissue, B-cell development, testis and T-cell signalling.

    Topics: Adaptor Proteins, Signal Transducing; Amino Acid Motifs; Animals; Cell Differentiation; Cell Transformation, Neoplastic; Embryonic Stem Cells; Epidermal Growth Factor; MAP Kinase Kinase 4; MAP Kinase Kinase Kinase 1; MAP Kinase Signaling System; Mice; Mice, Knockout; p38 Mitogen-Activated Protein Kinases; Polyubiquitin; Protein Binding; Transforming Growth Factor beta; Ubiquitination

2014
Crucial role of c-Jun phosphorylation at Ser63/73 mediated by PHLPP protein degradation in the cheliensisin a inhibition of cell transformation.
    Cancer prevention research (Philadelphia, Pa.), 2014, Volume: 7, Issue:12

    Cheliensisin A (Chel A), as a novel styryl-lactone isolated from Goniothalamus cheliensis Hu, has been demonstrated to have an inhibition of EGF-induced Cl41 cell transformation via stabilizing p53 protein in a Chk1-dependent manner, suggesting its chemopreventive activity in our previous studies. However, its underlying molecular mechanisms have not been fully characterized yet. In the current study, we found that Chel A treatment could increase c-Jun protein phosphorylation and activation, whereas the inhibition of c-Jun phosphorylation, by ectopic expression of a dominant-negative mutant of c-Jun, TAM67, reversed the Chel A inhibition of EGF-induced cell transformation and impaired Chel A induction of p53 protein and apoptosis. Moreover, our results indicated that Chel A treatment led to a PHLPP downregulation by promoting PHLPP protein degradation. We also found that PHLPP could interact with and bind to c-Jun protein, whereas ectopic PHLPP expression blocked c-Jun activation, p53 protein and apoptotic induction by Chel A, and further reversed the Chel A inhibition of EGF-induced cell transformation. With the findings, we have demonstrated that Chel A treatment promotes a PHLPP protein degradation, which can bind to c-Jun and mediates c-Jun phosphorylation, and further leading to p53 protein induction, apoptotic responses, subsequently resulting in cell transformation inhibition and chemopreventive activity of Chel A.

    Topics: Animals; Blotting, Western; Cell Proliferation; Cell Transformation, Neoplastic; Cells, Cultured; Epidermal Cells; Epidermal Growth Factor; Epidermis; Epoxy Compounds; Flow Cytometry; Immunoprecipitation; Mice; Nuclear Proteins; Phosphoprotein Phosphatases; Phosphorylation; Proteolysis; Proto-Oncogene Proteins c-jun; Pyrones; Real-Time Polymerase Chain Reaction; Reverse Transcriptase Polymerase Chain Reaction; RNA, Messenger; Serine; Signal Transduction; Tumor Suppressor Protein p53

2014
MiRNA expression may account for chronic but not for acute regulation of mRNA expression in human thyroid tumor models.
    PloS one, 2014, Volume: 9, Issue:11

    For thyroid tumorigenesis, two main human in vitro models are available: primary cultures of human thyrocytes treated with TSH or EGF/serum as models for autonomous adenomas (AA) or papillary thyroid carcinomas (PTC) respectively, and human thyroid tumor derived cell lines. Previous works of our group have assessed properties of those models, with a special emphasis on mRNA regulations. It is often assumed that miRNA may be one of the primary events inducing these mRNA regulations.. The purpose of this study was to investigate the representativity of those models to study microRNA regulations and their relation with mRNA expression. To achieve this aim, the miRNA expressions profiles of primary cultures treated with TSH or EGF/serum and of 6 thyroid cancer cell lines were compared to the expression profiles of 35 tumor tissues obtained by microarrays.. Our data on primary cultures have shown that the TSH or EGF/serum treatment did not greatly modify the microRNA expression profiles, which is contrary to what is observed for mRNA expression profiles, although they still evolved differently according to the treatment. The analysis of miRNA and mRNA expressions profiles in the cell lines has shown that they have evolved into a common, dedifferentiated phenotype, closer to ATC than to the tumors they are derived from.. Long-terms TSH or EGF/serum treatments do not mimic AA or PTC respectively in terms of miRNA expression as they do for mRNA, suggesting that the regulations of mRNA expression induced by these physiological agents occur independently of miRNA. The general patterns of miRNA expression in the cell lines suggest that they represent a useful model for undifferentiated thyroid cancer. Mirna probably do not mediate the rapid changes in gene expression in rapid cell biology regulation.

    Topics: Carcinoma, Papillary; Cell Line, Tumor; Cell Transformation, Neoplastic; Epidermal Growth Factor; Gene Expression Regulation, Neoplastic; Humans; MicroRNAs; Oligonucleotide Array Sequence Analysis; RNA, Messenger; Thyroid Gland; Thyroid Neoplasms; Thyrotropin

2014
Transformation of polarized epithelial cells by apical mistrafficking of epiregulin.
    Proceedings of the National Academy of Sciences of the United States of America, 2013, May-28, Volume: 110, Issue:22

    Establishment and maintenance of apico-basolateral trafficking pathways are critical to epithelial homeostasis. Loss of polarity and trafficking fidelity are thought to occur as a consequence of transformation; however, here we report that selective mistrafficking of the epidermal growth factor receptor (EGFR) ligand epiregulin (EREG) from the basolateral to the apical cell surface drives transformation. Normally, EREG is preferentially delivered to the basolateral surface of polarized Madin-Darby canine kidney cells. EREG basolateral trafficking is regulated by a conserved tyrosine-based basolateral sorting motif in its cytoplasmic domain (YXXΦ: Y(156)ERV). Both Y156 and V159 are required for basolateral sorting of EREG, because Y156A and V159G substitutions redirect EREG to the apical cell surface. We also show that basolateral sorting of EREG is adaptor protein 1B-independent. Apical mistrafficking of EREG has a distinctive phenotype. In contrast to transient EGFR tyrosine phosphorylation after basolateral EREG stimulation, apical EREG leads to prolonged EGFR tyrosine phosphorylation, which may be related, at least in part, to a lack of negative regulatory Y1045 phosphorylation and subsequent ubiquitylation. Notably, Madin-Darby canine kidney cells stably expressing apically mistrafficked EREG form significantly larger, hyperproliferative, poorly differentiated, and locally invasive tumors in nude mice compared with WT EREG-expressing cells.

    Topics: Animals; Cell Polarity; Cell Transformation, Neoplastic; Dogs; Electric Impedance; Electrophoresis, Polyacrylamide Gel; Epidermal Growth Factor; Epiregulin; Epithelial Cells; Fluorescent Antibody Technique; Immunoblotting; Immunoprecipitation; Madin Darby Canine Kidney Cells; Mice; Microscopy, Confocal; Phosphorylation; Protein Transport; Signal Transduction

2013
Cheliensisin A inhibits EGF-induced cell transformation with stabilization of p53 protein via a hydrogen peroxide/Chk1-dependent axis.
    Cancer prevention research (Philadelphia, Pa.), 2013, Volume: 6, Issue:9

    Cheliensisin A (Chel A), a novel styryl-lactone isolated from Goniothalamus cheliensis Hu, has been shown to induce apoptosis in human promyelocytic leukemia HL-60 cells with Bcl-2 downregulation. Yet, the potential chemopreventive effect of Chel A has not been explored. Here, we showed that Chel A treatment with various concentrations (0.5, 1.0, 2.0, and 4.0 μmol/L) for 3 weeks could dramatically inhibit EGF-induced cell transformation in Cl41 cells (IC50 ∼2.0 μmol/L). Also, coincubation of Cl41 cells with Chel A (2.0 and 4.0 μmol/L) for 48 hours could induce cell apoptosis in a caspase-3-dependent manner. Mechanically, Chel A treatment could result in increased p53 phosphorylation at Ser15 and elevated p53 total protein expression. Moreover, we found that p53 induction by Chel A was regulated at the protein degradation level, but not at either the transcription or the mRNA level. Further studies showed that p53 stabilization by Chel A was mediated via induction of phosphorylation and activation of Chk1 protein at Ser345. This notion was substantiated by the results that transfection of dominant negative mutant of Chk1 (GFP-Chk1 D130A) significantly attenuated the p53 protein expression, cell apoptosis, and inhibition of cell transformation by Chel A. Finally, increased hydrogen peroxide was found to mediate Chk1 phosphorylation at Ser345, p53 protein induction, cell apoptotic induction, and transformation inhibition following Chel A treatment. Taken together, our studies identify Chel A as a chemopreventive agent with the understanding of the molecular mechanisms involved.

    Topics: Animals; Apoptosis; Blotting, Western; Cell Proliferation; Cell Transformation, Neoplastic; Cells, Cultured; Checkpoint Kinase 1; Epidermal Cells; Epidermal Growth Factor; Epidermis; Epoxy Compounds; Flow Cytometry; Humans; Hydrogen Peroxide; Mice; Oxidants; Phosphorylation; Protein Kinases; Protein Stability; Proteolysis; Pyrones; Reactive Oxygen Species; Real-Time Polymerase Chain Reaction; Reverse Transcriptase Polymerase Chain Reaction; RNA, Messenger; Signal Transduction; Tumor Suppressor Protein p53

2013
Protein kinase CK2 phosphorylates and activates p21-activated kinase 1.
    Molecular biology of the cell, 2013, Volume: 24, Issue:18

    Activation of the p21-activated kinase 1 (PAK1) is achieved through a conformational change that converts an inactive PAK1 dimer to an active monomer. In this paper, we show that this change is necessary but not sufficient to activate PAK1 and that it is, rather, required for CK2-dependent PAK1(S223) phosphorylation that converts a monomeric PAK1 into a catalytically active form. This phosphorylation appears to be essential for autophosphorylation at specific residues and overall activity of PAK1. A phosphomimetic mutation (S223E) bypasses the requirement for GTPases in PAK1 activation, whereas the constitutive activity of the PAK1 mutant (PAK1(H83,86L)), postulated to mimic GTPase-induced structural changes, is abolished by inhibition of S223 phosphorylation. Thus, S223 is likely accessible to CK2 upon conformational changes of PAK1 induced by GTPase-dependent and GTPase-independent stimuli, suggesting that S223 phosphorylation may play a key role in the final step of the PAK1 activation process. The physiological significance of this phosphorylation is reinforced by the observations that CK2 is responsible for epidermal growth factor-induced PAK1 activation and that inhibition of S223 phosphorylation abrogates PAK1-mediated malignant transformation of prostate epithelial cells. Taken together, these findings identify CK2 as an upstream activating kinase of PAK1, providing a novel mechanism for PAK1 activation.

    Topics: Animals; Biocatalysis; Blotting, Western; Casein Kinase II; Cell Line, Tumor; Cell Transformation, Neoplastic; Enzyme Activation; Epidermal Growth Factor; Humans; Male; Mass Spectrometry; Mice; Models, Biological; Mutation; p21-Activated Kinases; Phosphorylation; Phosphoserine; Prostate; Reproducibility of Results

2013
EMT-induced stemness and tumorigenicity are fueled by the EGFR/Ras pathway.
    PloS one, 2013, Volume: 8, Issue:8

    Recent studies have revealed that differentiated epithelial cells would acquire stem cell-like and tumorigenic properties following an Epithelial-Mesenchymal Transition (EMT). However, the signaling pathways that participate in this novel mechanism of tumorigenesis have not been fully characterized. In Runx3 (-/-) p53 (-/-) murine gastric epithelial (GIF-14) cells, EMT-induced plasticity is reflected in the expression of the embryonal proto-oncogene Hmga2 and Lgr5, an exclusive gastrointestinal stem cell marker. Here, we report the concurrent activation of an EGFR/Ras gene expression signature during TGF-β1-induced EMT in GIF-14 cells. Amongst the altered genes was the induction of Egfr, which corresponded with a delayed sensitization to EGF treatment in GIF-14. Co-treatment with TGF-β1 and EGF or the expression of exogenous KRas led to increased Hmga2 or Lgr5 expression, sphere initiation and colony formation in soft agar assay. Interestingly, the gain in cellular plasticity/tumorigenicity was not accompanied by increased EMT. This uncoupling of EMT and the induction of plasticity reveals an involvement of distinct signaling cues, whereby the EGFR/Ras pathway specifically promotes stemness and tumorigenicity in EMT-altered GIF-14 cells. These data show that the EGFR/Ras pathway requisite for the sustenance of gastric stem cells in vivo and in vitro is involved in the genesis and promotion of EMT-induced tumor-initiating cells.

    Topics: Animals; Cell Line; Cell Transformation, Neoplastic; Cluster Analysis; Epidermal Growth Factor; Epithelial-Mesenchymal Transition; ErbB Receptors; Gene Expression Regulation; Mice; Models, Biological; Proto-Oncogene Proteins p21(ras); Signal Transduction; Spheroids, Cellular; Stem Cells; Transcriptome; Transforming Growth Factor beta1; Tumor Cells, Cultured

2013
Claudin-3 overexpression increases the malignant potential of colorectal cancer cells: roles of ERK1/2 and PI3K-Akt as modulators of EGFR signaling.
    PloS one, 2013, Volume: 8, Issue:9

    The altered expressions of claudin proteins have been reported during the tumorigenesis of colorectal cancer. However, the molecular mechanisms that regulate these events in this cancer type are poorly understood. Here, we report that epidermal growth factor (EGF) increases the expression of claudin-3 in human colorectal adenocarcinoma HT-29 cells. This increase was related to increased cell migration and the formation of anchorage-dependent and anchorage-independent colonies. We further showed that the ERK1/2 and PI3K-Akt pathways were involved in the regulation of these effects because specific pharmacological inhibition blocked these events. Genetic manipulation of claudin-1 and claudin-3 in HT-29 cells showed that the overexpression of claudin-1 resulted in decreased cell migration; however, migration was not altered in cells that overexpressed claudin-3. Furthermore, the overexpression of claudin-3, but not that of claudin-1, increased the tight junction-related paracellular flux of macromolecules. Additionally, an increased formation of anchorage-dependent and anchorage-independent colonies were observed in cells that overexpressed claudin-3, while no such changes were observed when claudin-1 was overexpressed. Finally, claudin-3 silencing alone despite induce increase proliferation, and the formation of anchoragedependent and -independent colonies, it was able to prevent the EGF-induced increased malignant potential. In conclusion, our results show a novel role for claudin-3 overexpression in promoting the malignant potential of colorectal cancer cells, which is potentially regulated by the EGF-activated ERK1/2 and PI3K-Akt pathways.

    Topics: Cell Line, Tumor; Cell Movement; Cell Proliferation; Cell Transformation, Neoplastic; Claudin-1; Claudin-3; Colorectal Neoplasms; Epidermal Growth Factor; ErbB Receptors; Gene Expression; Gene Expression Regulation, Neoplastic; Gene Silencing; Humans; Mitogen-Activated Protein Kinase 1; Mitogen-Activated Protein Kinase 3; Phosphatidylinositol 3-Kinases; Protein Transport; Proto-Oncogene Proteins c-akt; Signal Transduction; Tumor Stem Cell Assay

2013
Identification and functional characterization of p130Cas as a substrate of protein tyrosine phosphatase nonreceptor 14.
    Oncogene, 2013, Apr-18, Volume: 32, Issue:16

    Protein tyrosine phosphatase nonreceptor type 14 (PTPN14) is frequently mutated in a variety of human cancers. However, the cell signaling pathways regulated by PTPN14 largely remain to be elucidated. Here, we identify a list of potential substrates of PTPN14 using a phospho-proteomic approach. We show that p130 Crk-associated substrate (p130Cas) is a direct substrate of PTPN14 and that PTPN14 specifically regulates p130Cas phosphorylation at tyrosine residue 128 (Y128) in colorectal cancer (CRC) cells. We engineered CRC cells homozygous for a p130Cas Y128F knock-in mutant and found that these cells exhibit significantly reduced migration and colony formation, impaired anchorage-independent growth, slower xenograft tumor growth in nude mice and have decreased phosphorylation of AKT. Furthermore, we demonstrate that SRC phosphorylates p130Cas Y128 and that CRC cell lines harboring high levels of pY128Cas are more sensitive to SRC family kinase inhibitor Dasatinib. These findings suggest that p130Cas Y128 phosphorylation may be exploited as a predictive marker for Dasatinib response in cancer patients. In aggregate, our studies reveal a novel signaling pathway that has an important role in colorectal tumorigenesis.

    Topics: Amino Acid Sequence; Animals; Cell Transformation, Neoplastic; Colorectal Neoplasms; Crk-Associated Substrate Protein; Dasatinib; Epidermal Growth Factor; Female; Gene Knock-In Techniques; HCT116 Cells; HEK293 Cells; HT29 Cells; Humans; Mice; Mice, Nude; Molecular Sequence Data; Phosphorylation; Protein Kinase Inhibitors; Protein Tyrosine Phosphatases, Non-Receptor; Pyrimidines; Signal Transduction; Thiazoles; Transplantation, Heterologous

2013
Alterations of EGFR, p53 and PTEN that mimic changes found in basal-like breast cancer promote transformation of human mammary epithelial cells.
    Cancer biology & therapy, 2013, Volume: 14, Issue:3

    Breast cancer can be classified into different molecular subtypes with varying clinical and pathological characteristics. The basal-like breast cancer subtype represents one of the most aggressive and lethal types of breast cancer, and due to poor mechanistic understanding, it lacks targeted therapy. Many basal-like breast cancer patient samples display alterations of established drivers of cancer development, including elevated expression of EGFR, p53 inactivating mutations and loss of expression of the tumor suppressor PTEN; however, their contribution to human basal-like breast cancer pathogenesis remains ill-defined. Using non-transformed human mammary epithelial cells, we set out to determine whether altering EGFR, p53 and PTEN in different combinations could contribute to basal-like breast cancer progression through transformation of cells. Altering PTEN in combination with either p53 or EGFR in contrast to any of the single alterations caused increased growth of transformed colonies in soft agar. Concomitantly modifying all three genes led to the highest rate of cellular proliferation and the greatest degree of anchorage-independent colony formation. Results from our effort to engineer a model of BBC expressing alterations of EGFR, p53 and PTEN suggest that these changes are cooperative and likely play a causal role in basal-like breast cancer pathogenesis. Consideration should be given to targeting EGFR and restoring p53 and PTEN signaling simultaneously as a strategy for treatment of this subtype of breast cancer.

    Topics: Animals; Breast Neoplasms; Cell Line, Tumor; Cell Proliferation; Cell Transformation, Neoplastic; Epidermal Growth Factor; ErbB Receptors; Female; Gene Expression; Gene Expression Regulation, Neoplastic; Gene Knockout Techniques; Humans; Mice; Models, Biological; Neoplasms, Basal Cell; PTEN Phosphohydrolase; Signal Transduction; Transplantation, Heterologous; Tumor Stem Cell Assay; Tumor Suppressor Protein p53

2013
Pin1 is required for ultraviolet A-stimulated cyclooxygenase-2 induction in mouse epidermal cells.
    Cancer letters, 2013, Jul-10, Volume: 335, Issue:1

    Ultraviolet A (UVA) radiation (320-400 nm) is considered a major cause of human skin photoaging and skin cancer. Overexpression of cyclooxygenase-2 (COX-2) leads to prostanoid formation in skin tissue, disturbs the balance between proliferation and apoptosis, and subsequently promotes tumorigenesis. The peptidyl-prolyl isomerase Pin1 is known to be overexpressed in most cancer cell types and plays an important role in oncogenesis. Here, we studied whether exposure of JB6 Cl41 mouse epidermal cells to UVA affects COX-2 expression and the possible involvement of Pin1 activation. UVA increased COX-2 protein expression and prostaglandin E2 production in an energy-dependent manner. Pre-exposure of JB6 Cl41 cells to UVA potentiated epidermal growth factor-induced anchorage-independent growth; this effect was significantly suppressed by inhibition of COX-2. UVA-stimulated COX-2 expression was significantly decreased by inhibition of Pin1. The increased COX-2 gene transcription in response to UVA was preceded by activation of the transcription factors nuclear factor-κB (NF-κB), cAMP response element-binding protein (CREB), CCAAT/enhancer-binding proteins α and β (C/EBPα and C/EBPβ) and c-Jun/activator protein-1 (AP-1). Pin1 inhibitor treatment suppressed the activation of NF-κB, CREB, and C/EBP by UVA irradiation. Conversely, JB6 C141 cells overexpressing Pin1 showed increased basal COX-2 expression and NF-κB, CREB, C/EBP, and AP-1 activities. These results suggest that UVA-induced COX-2 expression is mediated by Pin1 activation and this is associated with malignant transformation of epidermal cells.

    Topics: Animals; CCAAT-Enhancer-Binding Proteins; Cell Line; Cell Transformation, Neoplastic; Cyclic AMP Response Element-Binding Protein; Cyclooxygenase 2; Cyclooxygenase 2 Inhibitors; Dinoprostone; Enzyme Activation; Enzyme Induction; Epidermal Growth Factor; Epidermis; Epithelial Cells; Meloxicam; Mice; Naphthoquinones; NF-kappa B; NIMA-Interacting Peptidylprolyl Isomerase; Peptidylprolyl Isomerase; Signal Transduction; Thiazines; Thiazoles; Transcription Factor AP-1; Ultraviolet Rays

2013
EGFR signaling upregulates expression of microsomal prostaglandin E synthase-1 in cancer cells leading to enhanced tumorigenicity.
    Oncogene, 2012, Jul-19, Volume: 31, Issue:29

    In this report we describe the contribution of prostaglandin E(2) (PGE(2)) derived from the inducible microsomal PGE-synthase type-1 (mPGES-1) to the epidermal growth factor receptor (EGFR) oncogenic drive in tumor epithelial cells and in tumor-bearing mice. EGFR stimulation upregulated expression of mPGES-1 in HT-29, A431 and A549 cancer cells. Egr-1, a transcription factor induced by EGF, mediated this response. The Egr-1 rise provoked the overexpression of mPGES-1 messenger and protein, and enhanced PGE(2) formation. These changes were suppressed either by silencing Egr-1, or by upstream blockade of EGFR or ERK1/2 signals. Further, in a clonogenic assay on tumor cells, EGF induced a florid tumorigenic phenotype, which regressed when mPGES-1 was silenced or knocked down. EGF-induced mPGES-1 overexpression in epithelial cell reduced E-cadherin expression, whereas enhancing that of vimentin, suggesting an incipient mesenchymal phenotype. Additionally, inhibiting the EGFR in mice bearing the A431 tumor, the mPGES-1 expression and the tumor growth, exhibited a parallel decline. In conclusion, these findings provide novel evidence that a tight cooperation between the EGF/EGFR and mPGES-1 leads to a significant tumorigenic gain in epithelial cells, and provide clues for controlling the vicious association.

    Topics: Animals; Carcinoma, Squamous Cell; Cell Line, Tumor; Cell Transformation, Neoplastic; Early Growth Response Protein 1; Enzyme Activation; Epidermal Growth Factor; ErbB Receptors; Female; Gene Silencing; Humans; Intramolecular Oxidoreductases; Mice; Mitogen-Activated Protein Kinase 1; Mitogen-Activated Protein Kinase 3; Prostaglandin-E Synthases; Signal Transduction; Up-Regulation

2012
Analysis of the role of p38 MAP kinase in epidermal growth factor-induced JB6 Cl41 cell transformation by cDNA array.
    Gene, 2012, Apr-10, Volume: 497, Issue:1

    To further explore the mechanism of p38 MAP kinase in regulation of JB6 Cl41 cell transformation. cDNA array was employed to scan the differential expression genes between DN-p38 cells and CMV-neo JB6 Cl41 cells after EGF stimuli. We found that up-expression genes including oncogenes and tumor suppressor genes, p53-associated protein, transcription repressors, apoptosis-associated genes, and growth arrest and DNA damage-inducible protein 153 were detected in DN-p38 cells, but low expression in CMV-neo JB6 Cl41 cells after EGF treatment. Meanwhile, some proto-oncogenes, such as c-Myc, and signal transducer and activator of transcription 1 (STAT1) were lowly expressed in EGF-stimulated DN-p38 cells, but had relatively high expression level in CMV-neo JB6 Cl41 cells under the same stimuli. Four of the differential expression genes were further confirmed by quantitative RT-PCR analysis. Our results indicate that p38 MAP kinase is involved in EGF-induced JB6 Cl41 cell transformation through effecting on more genes expression levels including transcription factors, proto-oncogene, apoptosis-related genes and growth arrest genes.

    Topics: Cell Line; Cell Transformation, Neoplastic; Epidermal Growth Factor; Gene Expression; Gene Expression Profiling; Humans; Oligonucleotide Array Sequence Analysis; p38 Mitogen-Activated Protein Kinases; Proto-Oncogene Mas; Transfection

2012
Ezrin overexpression by transformed human ovarian surface epithelial cells, ovarian cleft cells, and serous ovarian adenocarcinoma cells.
    Reproductive sciences (Thousand Oaks, Calif.), 2012, Volume: 19, Issue:8

    We have shown that ezrin expression correlates with ovarian epithelial cancer (OVCA) cell proliferation and metastatic behavior. In this study, we evaluated ezrin expression in transformed ovarian superficial epithelial cells (OSE) in ovarian clefts and in culture.. Immunohistochemistry and Western blotting for immunoreactive ezrin (ir-ezrin) in normal ovarian tissue, cultured OSE, and ovarian epithelial cancer cells.. While ir-ezrin was not demonstrable in normal cuboidal surface cells or interior ovarian organelles, cells lining the ovarian clefts strongly expressed ir-ezrin. Long-term culture of OSE increased ezrin expression and cytological abnormalities. Administration of estradiol and insulin at levels reported in inclusions dramatically induced OSE ir-ezrin expression to OVCA levels and membrane specializations; ruffling, pseudopodia and filopodia. Moreover epidermal growth factor (EGF) drastically increased ezrin translocation in OSE cells in a time-dependent manner.. Ezrin expression by OSE increases during transformation. Ezrin expression is responsive to estradiol and growth factors previously shown to be present in ovarian inclusions. These findings suggest that the microenvironment in ovarian inclusions and clefts contributes to the development of OVCA. Our findings elaborate on the mechanism of the ovarian origin of OVCA.

    Topics: Blotting, Western; Carcinoma, Ovarian Epithelial; Cell Line, Tumor; Cell Transformation, Neoplastic; Cells, Cultured; Cystadenocarcinoma, Serous; Cytoskeletal Proteins; Epidermal Growth Factor; Epithelial Cells; Estradiol; Female; Gene Expression; Humans; Immunohistochemistry; Insulin; Neoplasms, Glandular and Epithelial; Ovarian Neoplasms; Ovary

2012
Macropinocytosis of the PDGF β-receptor promotes fibroblast transformation by H-RasG12V.
    Molecular biology of the cell, 2012, Volume: 23, Issue:13

    Receptor tyrosine kinase (RTK) signaling is frequently increased in tumor cells, sometimes as a result of decreased receptor down-regulation. The extent to which the endocytic trafficking routes can contribute to such RTK hyperactivation is unclear. Here, we show for the first time that fibroblast transformation by H-RasG12V induces the internalization of platelet-derived growth factor β-receptor (PDGFRβ) by macropinocytosis, enhancing its signaling activity and increasing anchorage-independent proliferation. H-RasG12V transformation and PDGFRβ activation were synergistic in stimulating phosphatidylinositol (PI) 3-kinase activity, leading to receptor macropinocytosis. PDGFRβ macropinocytosis was both necessary and sufficient for enhanced receptor activation. Blocking macropinocytosis by inhibition of PI 3-kinase prevented the increase in receptor activity in transformed cells. Conversely, increasing macropinocytosis by Rabankyrin-5 overexpression was sufficient to enhance PDGFRβ activation in nontransformed cells. Simultaneous stimulation with PDGF-BB and epidermal growth factor promoted macropinocytosis of both receptors and increased their activation in nontransformed cells. We propose that H-Ras transformation promotes tumor progression by enhancing growth factor receptor signaling as a result of increased receptor macropinocytosis.

    Topics: Becaplermin; Cell Adhesion; Cell Proliferation; Cell Transformation, Neoplastic; Cells, Cultured; Epidermal Growth Factor; ErbB Receptors; Fibroblasts; Humans; Mutation, Missense; Phosphorylation; Pinocytosis; Protein Processing, Post-Translational; Protein Transport; Proto-Oncogene Proteins c-sis; Proto-Oncogene Proteins p21(ras); Receptor, Platelet-Derived Growth Factor beta; Signal Transduction; Transport Vesicles

2012
Topical polyethylene glycol as a novel chemopreventive agent for oral cancer via targeting of epidermal growth factor response.
    PloS one, 2012, Volume: 7, Issue:6

    Head and neck squamous cell carcinoma (HNSCC) is a major cause of morbidity and mortality underscoring the need for safe and effective chemopreventive strategies. Targeting epidermal growth factor receptor (EGFR) is attractive in that it is an early critical event in HNSCC pathogenesis. However, current agents lack efficacy or have unacceptable toxicity. Several groups have demonstrated that the over-the-counter medication, polyethylene glycol (PEG) has remarkable chemopreventive efficacy against colon carcinogenesis. Importantly, we reported that this effect is mediated through EGFR internalization/degradation. In the current study, we investigated the chemopreventive efficacy of this agent against HNSCC, using both the well validated animal model 4-NQO (4-nitroquinoline 1-oxide) rat model and cell culture with the human HNSCC cell line SCC-25. We demonstrated that daily topical application of 10% PEG-8000 in the oral cavity (tongue and cavity wall) post 4NQO initiation resulted in a significant reduction in tumor burden (both, tumor size and tumors/tumor bearing rat) without any evidence of toxicity. Immunohistochemical studies depicted decreased proliferation (number of Ki67-positive cells) and reduced expression of EGFR and its downstream effectors cyclin D1 in the tongue mucosa of 4NQO-rats treated with PEG. We showed that EGFR was also markedly downregulated in SCC-25 cells by PEG-8000 with a concomitant induction of G1-S phase cell-cycle arrest, which was potentially mediated through upregulated p21(cip1/waf1). In conclusion, we demonstrate, for the first time, that PEG has promising efficacy and safety as a chemopreventive efficacy against oral carcinogenesis.

    Topics: 4-Nitroquinoline-1-oxide; Administration, Oral; Administration, Topical; Animals; Antineoplastic Agents; Blotting, Western; Cell Cycle; Cell Line, Tumor; Cell Proliferation; Cell Transformation, Neoplastic; Chemoprevention; Cyclin D1; Cyclin-Dependent Kinase Inhibitor p21; Disease Progression; Down-Regulation; Epidermal Growth Factor; Epithelial Cells; ErbB Receptors; Humans; Male; Molecular Targeted Therapy; Mouth Mucosa; Mouth Neoplasms; Polyethylene Glycols; Rats; Rats, Inbred F344

2012
Tumor necrosis factor receptor-associated factor family protein 2 is a key mediator of the epidermal growth factor-induced ribosomal S6 kinase 2/cAMP-responsive element-binding protein/Fos protein signaling pathway.
    The Journal of biological chemistry, 2012, Jul-27, Volume: 287, Issue:31

    TRAF2 has an important function in mediating the TNF-R signaling pathway toward activation of NF-κB and JNKs. Here we reveal a novel function of TRAF2 in the epidermal growth factor (EGF) signaling pathway. Knockdown of TRAF2 blocked EGF-induced AP-1 activity and anchorage- independent cell transformation. Notably, we showed that EGF induces ribosomal S6 kinase 2 (RSK2) ubiquitination, and knocking down TRAF2 suppresses ubiquitination of RSK2 induced by EGF. We also found that TRAF2 affects RSK2 activity through RSK2 ubiquitination. RSK2 plays a critical role in AP-1 activity mediated through CREB and c-Fos, which regulates anchorage-independent cell transformation. In addition, TRAF2 is overexpressed in colon cancer and required for colon cancer development, suggesting that TRAF2 might be a potential molecular target for cancer prevention and treatment.

    Topics: Animals; Cell Line, Tumor; Cell Proliferation; Cell Transformation, Neoplastic; Colorectal Neoplasms; Cyclic AMP Response Element-Binding Protein; Epidermal Growth Factor; Female; Gene Knockdown Techniques; Humans; Mice; Mice, Nude; Neoplasm Transplantation; Polyubiquitin; Protein Binding; Protein Processing, Post-Translational; Proto-Oncogene Proteins c-fos; Ribosomal Protein S6 Kinases, 90-kDa; Signal Transduction; TNF Receptor-Associated Factor 2; Transcription Factor AP-1; Transcription, Genetic; Tumor Burden; Ubiquitination

2012
Althaea rosea Cavanil and Plantago major L. suppress neoplastic cell transformation through the inhibition of epidermal growth factor receptor kinase.
    Molecular medicine reports, 2012, Volume: 6, Issue:4

    For thousands of years in Asia, Althaea rosea Cavanil (ARC) and Plantago major L. (PML) have been used as powerful non-toxic therapeutic agents that inhibit inflammation. However, the anticancer mechanisms and molecular targets of ARC and PML are poorly understood, particularly in epidermal growth factor (EGF)-induced neoplastic cell transformation. The aim of this study was to evaluate the chemopreventive effects and mechanisms of the methanol extracts from ARC (MARC) and PML (MPML) in EGF-induced neoplastic cell transformation of JB6 P+ mouse epidermal cells using an MTS assay, anchorage-independent cell transformation assay and western blotting. Our results showed that MARC and MPML significantly suppressed neoplastic cell transformation by inhibiting the kinase activity of the EGF receptor (EGFR). The activation of EGFR by EGF was suppressed by MARC and MPML treatment in EGFR(+/+) cells, but not in EGFR(-/-) cells. In addition, MARC and MPML inhibited EGF-induced cell proliferation in EGFR-expressing murine embryonic fibroblasts (EGFR(+/+)). These results strongly indicate that EGFR targeting by MARC and MPML may be a good strategy for chemopreventive or chemotherapeutic applications.

    Topics: Althaea; Animals; Cell Line; Cell Transformation, Neoplastic; Epidermal Growth Factor; ErbB Receptors; Fibroblasts; Methanol; Mice; Phosphorylation; Plant Extracts; Plantago

2012
Identification of mammalian target of rapamycin as a direct target of fenretinide both in vitro and in vivo.
    Carcinogenesis, 2012, Volume: 33, Issue:9

    N-(4-hydroxyphenyl) retinamide (4HPR, fenretinide) is a synthetic retinoid that has been tested in clinical trials as a cancer therapeutic and chemopreventive agent. Although 4HPR has been shown to be cytotoxic to many kinds of cancer cells, the underlying molecular mechanisms are only partially understood. Until now, no direct cancer-related molecular target has been reported to be involved in the antitumor activities of 4HPR. Herein, we found that 4HPR inhibited mammalian target of rapamycin (mTOR) kinase activity by directly binding with mTOR, which suppressed the activities of both the mTORC1 and the mTORC2 complexes. The predicted binding mode of 4HPR with mTOR was based on a homology computer model, which showed that 4HPR could bind in the ATP-binding pocket of the mTOR protein through hydrogen bonds and hydrophobic interactions. In vitro studies also showed that 4HPR attenuated mTOR downstream signaling in a panel of non-small-cell lung cancer cells, resulting in growth inhibition. Moreover, knockdown of mTOR in cancer cells decreased their sensitivity to 4HPR. Results of an in vivo study demonstrated that i.p. injection of 4HPR in A549 lung tumor-bearing mice effectively suppressed cancer growth. The expression of mTOR downstream signaling molecules in tumor tissues was also decreased after 4HPR treatment. Taken together, our results are the first to identify mTOR as a direct antitumor target of 4HPR both in vitro and in vivo, providing a valuable rationale for guiding the clinical uses of 4HPR.

    Topics: Animals; Antineoplastic Agents; Cell Line, Tumor; Cell Transformation, Neoplastic; Epidermal Growth Factor; Fenretinide; Humans; Mechanistic Target of Rapamycin Complex 1; Mice; Multiprotein Complexes; Proteins; Signal Transduction; TOR Serine-Threonine Kinases; Transcription Factors; Xenograft Model Antitumor Assays

2012
Rac1 activation driven by 14-3-3ζ dimerization promotes prostate cancer cell-matrix interactions, motility and transendothelial migration.
    PloS one, 2012, Volume: 7, Issue:7

    14-3-3 proteins are ubiquitously expressed dimeric adaptor proteins that have emerged as key mediators of many cell signaling pathways in multiple cell types. Its effects are mainly mediated by binding to selective phosphoserine/threonine proteins. The importance of 14-3-3 proteins in cancer have only started to become apparent and its exact role in cancer progression as well as the mechanisms by which 14-3-3 proteins mediate cancer cell function remain unknown. While protein 14-3-3σ is widely accepted as a tumor suppressor, 14-3-3ζ, β and γ isoforms have been shown to have tumor promoting effects. Despite the importance of 14-3-3 family in mediating various cell processes, the exact role and mechanism of 14-3-3ζ remain unexplored. In the current study, we investigated the role of protein 14-3-3ζ in prostate cancer cell motility and transendothelial migration using biochemical, molecular biology and electric cell-substrate impedance sensing approaches as well as cell based functional assays. Our study indicated that expression with wild-type protein 14-3-3ζ significantly enhanced Rac activity in PC3 cells. In contrast, expression of dimer-resistant mutant of protein 14-3-3ζ (DM-14-3-3) inhibited Rac activity and associated phosphorylation of p21 activated kinase-1 and 2. Expression with wild-type 14-3-3ζ or constitutively active Rac1 enhanced extracellular matrix recognition, lamellipodia formation, cell migration and trans-endothelial migration by PC3 cells. In contrast, expression with DM 14-3-3ζ or DN-Rac1 in PC3 cells significantly inhibited these cell functions. Our results demonstrate for the first time that 14-3-3ζ enhances prostate cancer cell-matrix interactions, motility and transendothelial migration in vitro via activation of Rac1-GTPase and is an important target for therapeutic interventions for prostate cancer.

    Topics: 14-3-3 Proteins; Animals; Cell Line, Tumor; Cell Transformation, Neoplastic; Enzyme Activation; Epidermal Growth Factor; Extracellular Matrix; Humans; Male; Mice; p21-Activated Kinases; Prostatic Neoplasms; Protein Binding; Protein Multimerization; Pseudopodia; rac1 GTP-Binding Protein; Signal Transduction; Transendothelial and Transepithelial Migration

2012
EGF receptor targeted tumor imaging with biotin-PEG-EGF linked to (99m)Tc-HYNIC labeled avidin and streptavidin.
    Nuclear medicine and biology, 2012, Volume: 39, Issue:8

    As direct radiolabeled peptides suffer limitations for in vivo imaging, we investigated the usefulness of radioloabeled avidin and streptavidin as cores to link peptide ligands for targeted tumor imaging.. Human epidermal growth factor (EGF) was site specifically conjugated with a single PEG-biotin molecule and linked to (99m)Tc-HYNIC labeled avidin-FITC (Av) or streptavidin-Cy5.5 (Sav). Receptor targeting was verified in vitro, and in vivo pharmacokinetic and biodistribution profiles were studied in normal mice. Scintigraphic imaging was performed in MDA-MB-468 breast tumor xenografted nude mice.. Whereas both (99m)Tc-Av-EGF and (99m)Tc-Sav-EGF retained receptor-specific binding in vitro, the two probes substantially diverged in pharmacokinetic and biodistribution behavior in vivo. (99m)Tc-Av-EGF was rapidly eliminated from the circulation with a T1/2 of 4.3 min, and showed intense hepatic accumulation but poor tumor uptake (0.6%ID/gm at 4 h). (99m)Tc-Sav-EGF displayed favorable in vivo profiles of longer circulation (T1/2β, 51.5 min) and lower nonspecific uptake that resulted in higher tumor uptake (3.8 %ID/gm) and clear tumor visualization at 15 h.. (99m)Tc-HYNIC labeled streptavidin linked with growth factor peptides may be useful as a protein-ligand complex for targeted imaging of tumor receptors.

    Topics: Animals; Avidin; Biotin; Breast Neoplasms; Cell Line, Tumor; Cell Transformation, Neoplastic; Diagnostic Imaging; Epidermal Growth Factor; ErbB Receptors; Humans; Mice; Models, Molecular; Organotechnetium Compounds; Polyethylene Glycols; Protein Conformation; Radionuclide Imaging; Streptavidin

2012
Mycalamide A shows cytotoxic properties and prevents EGF-induced neoplastic transformation through inhibition of nuclear factors.
    Marine drugs, 2012, Volume: 10, Issue:6

    Mycalamide A, a marine natural compound previously isolated from sponges, is known as a protein synthesis inhibitor with potent antitumor activity. However, the ability of this compound to prevent malignant transformation of cells has never been examined before. Here, for the first time, we report the isolation of mycalamide A from ascidian Polysincraton sp. as well as investigation of its cancer preventive properties. In murine JB6 Cl41 P(+) cells, mycalamide A inhibited epidermal growth factor (EGF)-induced neoplastic transformation, and induced apoptosis at subnanomolar or nanomolar concentrations. The compound inhibited transcriptional activity of the oncogenic nuclear factors AP-1 and NF-κB, a potential mechanism of its cancer preventive properties. Induction of phosphorylation of the kinases MAPK p38, JNK, and ERK was also observed at high concentrations of mycalamide A. The drug shows promising potential for both cancer-prevention and cytotoxic therapy and should be further developed.

    Topics: Animals; Apoptosis; Cell Line, Tumor; Cell Nucleus; Cell Transformation, Neoplastic; Epidermal Growth Factor; HeLa Cells; Humans; MAP Kinase Kinase 4; MAP Kinase Signaling System; Mice; NF-kappa B; p38 Mitogen-Activated Protein Kinases; Phosphorylation; Pyrans; Transcription Factor AP-1; Transcription, Genetic; Tumor Suppressor Protein p53; Urochordata

2012
PKM2 phosphorylates histone H3 and promotes gene transcription and tumorigenesis.
    Cell, 2012, Aug-17, Volume: 150, Issue:4

    Tumor-specific pyruvate kinase M2 (PKM2) is essential for the Warburg effect. In addition to its well-established role in aerobic glycolysis, PKM2 directly regulates gene transcription. However, the mechanism underlying this nonmetabolic function of PKM2 remains elusive. We show here that PKM2 directly binds to histone H3 and phosphorylates histone H3 at T11 upon EGF receptor activation. This phosphorylation is required for the dissociation of HDAC3 from the CCND1 and MYC promoter regions and subsequent acetylation of histone H3 at K9. PKM2-dependent histone H3 modifications are instrumental in EGF-induced expression of cyclin D1 and c-Myc, tumor cell proliferation, cell-cycle progression, and brain tumorigenesis. In addition, levels of histone H3 T11 phosphorylation correlate with nuclear PKM2 expression levels, glioma malignancy grades, and prognosis. These findings highlight the role of PKM2 as a protein kinase in its nonmetabolic functions of histone modification, which is essential for its epigenetic regulation of gene expression and tumorigenesis.

    Topics: Animals; Astrocytoma; Carrier Proteins; Cell Line; Cell Line, Tumor; Cell Transformation, Neoplastic; Cyclin D1; Epidermal Growth Factor; Epigenesis, Genetic; Female; Gene Expression Regulation, Neoplastic; Glioblastoma; Histones; Humans; Membrane Proteins; Mice; Mice, Nude; Neoplasm Transplantation; Proto-Oncogene Proteins c-myc; Thyroid Hormone-Binding Proteins; Thyroid Hormones; Transcription, Genetic; Transplantation, Heterologous

2012
RSK2 as a key regulator in human skin cancer.
    Carcinogenesis, 2012, Volume: 33, Issue:12

    Our previous report demonstrated that RSK2 plays an important role in cell proliferation and transformation induced by tumor promoters such as epidermal growth factor mediated through the N-terminal kinase domain of RSK2 in JB6 Cl41 mouse skin epidermal cells in vitro. However, no direct evidence has been reported regarding the relationship of RSK2 activity and human skin cancer. To elucidate the relationship of RSK2 activity and human skin cancer, we examined the effect of knocking down RSK2 expression on epidermal growth factor-induced anchorage-independent transformation in the premalignant HaCaT human skin keratinocyte cell line and on soft agar colony growth of SK-MEL-28 malignant melanoma cells. We found that the phosphorylated protein levels of RSK2 were enhanced in cancer tissues compared with normal tissues in a human skin cancer tissue array. We found that UVB stimulation induced increased in not only the total and phosphorylated protein levels of ERKs and RSK2 but also the nuclear localization and gene expression of RSK2. RSK2 knockdown inhibited proliferation and anchorage-independent transformation of HaCaT cells and soft agar colony growth of malignant melanoma cells. Moreover, RSK2(-/-) mouse embryonic fibroblast (MEF) showed enhanced sub-G(1) accumulation induced by UVB stimulation compared with RSK2(+/+) MEFs, indicating that RSK2 might play an important role in tolerance against stress associated with ultraviolet. Importantly, activated RSK2 protein levels were highly abundant in human skin cancer tissues compared with matched skin normal tissues. Taken together, our results demonstrated that RSK2 plays a key role in neoplastic transformation of human skin cells and in skin cancer growth.

    Topics: Cell Line, Tumor; Cell Transformation, Neoplastic; Epidermal Growth Factor; Extracellular Signal-Regulated MAP Kinases; Humans; Ribosomal Protein S6 Kinases, 90-kDa; Signal Transduction; Skin Neoplasms; Tissue Array Analysis; Ultraviolet Rays

2012
EGFR-induced and PKCε monoubiquitylation-dependent NF-κB activation upregulates PKM2 expression and promotes tumorigenesis.
    Molecular cell, 2012, Dec-14, Volume: 48, Issue:5

    Many types of human tumor cells have overexpressed pyruvate kinase M2 (PKM2). However, the mechanism underlying this increased PKM2 expression remains to be defined. We demonstrate here that EGFR activation induces PLCγ1-dependent PKCε monoubiquitylation at Lys321 mediated by RINCK1 ubiquitin ligase. Monoubiquitylated PKCε interacts with a ubiquitin-binding domain in NEMO zinc finger and recruits the cytosolic IKK complex to the plasma membrane, where PKCε phosphorylates IKKβ at Ser177 and activates IKKβ. Activated RelA interacts with HIF1α, which is required for RelA to bind the PKM promoter. PKCε- and NF-κB-dependent PKM2 upregulation is required for EGFR-promoted glycolysis and tumorigenesis. In addition, PKM2 expression correlates with EGFR and IKKβ activity in human glioblastoma specimens and with grade of glioma malignancy. These findings highlight the distinct regulation of NF-κB by EGF, in contrast to TNF-α, and the importance of the metabolic cooperation between the EGFR and NF-κB pathways in PKM2 upregulation and tumorigenesis.

    Topics: Animals; Brain Neoplasms; Carrier Proteins; Cell Line, Tumor; Cell Transformation, Neoplastic; Enzyme Activation; Epidermal Growth Factor; ErbB Receptors; Female; Gene Expression Regulation, Enzymologic; Gene Expression Regulation, Neoplastic; Genes, Reporter; Glioblastoma; Glucose; Glycolysis; HEK293 Cells; Heterogeneous-Nuclear Ribonucleoproteins; Humans; Hypoxia-Inducible Factor 1, alpha Subunit; I-kappa B Kinase; Lactic Acid; Membrane Proteins; Mice; Mice, Nude; Mutagenesis, Site-Directed; Mutation; Neoplasm Grading; Neoplasm Transplantation; NF-kappa B; Phospholipase C gamma; Phosphorylation; Polypyrimidine Tract-Binding Protein; Prognosis; Promoter Regions, Genetic; Protein Kinase C-epsilon; RNA Interference; Serine; Signal Transduction; Thyroid Hormone-Binding Proteins; Thyroid Hormones; Transcription Factor RelA; Transfection; Ubiquitination; Up-Regulation

2012
p130Cas over-expression impairs mammary branching morphogenesis in response to estrogen and EGF.
    PloS one, 2012, Volume: 7, Issue:12

    p130Cas adaptor protein regulates basic processes such as cell cycle control, survival and migration. p130Cas over-expression has been related to mammary gland transformation, however the in vivo consequences of p130Cas over-expression during mammary gland morphogenesis are not known. In ex vivo mammary explants from MMTV-p130Cas transgenic mice, we show that p130Cas impairs the functional interplay between Epidermal Growth Factor Receptor (EGFR) and Estrogen Receptor (ER) during mammary gland development. Indeed, we demonstrate that p130Cas over-expression upon the concomitant stimulation with EGF and estrogen (E2) severely impairs mammary morphogenesis giving rise to enlarged multicellular spherical structures with altered architecture and absence of the central lumen. These filled acinar structures are characterized by increased cell survival and proliferation and by a strong activation of Erk1/2 MAPKs and Akt. Interestingly, antagonizing the ER activity is sufficient to re-establish branching morphogenesis and normal Erk1/2 MAPK activity. Overall, these results indicate that high levels of p130Cas expression profoundly affect mammary morphogenesis by altering epithelial architecture, survival and unbalancing Erk1/2 MAPKs activation in response to growth factors and hormones. These results suggest that alteration of morphogenetic pathways due to p130Cas over-expression might prime mammary epithelium to tumorigenesis.

    Topics: Animals; Cell Transformation, Neoplastic; Crk-Associated Substrate Protein; Epidermal Growth Factor; Epithelium; ErbB Receptors; Estrogens; Female; Gene Expression Regulation; Mammary Glands, Animal; Mice; Mice, Transgenic; Mitogen-Activated Protein Kinase Kinases; Morphogenesis; Organ Culture Techniques; Receptors, Estrogen

2012
EGF/TGFβ1 co-stimulation of oral squamous cell carcinoma cells causes an epithelial-mesenchymal transition cell phenotype expressing laminin 332.
    Journal of oral pathology & medicine : official publication of the International Association of Oral Pathologists and the American Academy of Oral Pathology, 2011, Volume: 40, Issue:1

    Epithelial-mesenchymal transition (EMT) is suggested to be crucial for the development of an invasive and metastatic carcinoma cell phenotype. Therefore, the definition of this phenotype is of great clinical interest. We recently evidenced vimentin positive cells in oral squamous cell carcinoma (OSCC) invasive front expressing laminin γ2 chain mRNA implicating an EMT origin of these cells. To further elucidate the nature of these cells, we have investigated the relation between EMT criteria and laminin-332 expression in a cell culture model of transforming growth factor beta-1 (TGFβ1)/epithelial growth factor (EGF) long time co-stimulation. We demonstrate that in contrast to TGFβ1 or EGF alone, co-stimulation induces phenotype transition in OSCC cells which fulfils the criteria of EMT in terms of vimentin up-regulation and E-cadherin down-regulation on protein level as well as cell scattering. Furthermore, cells displayed a strongly enhanced invasiveness and adhesion to type I-IV collagens. Phenotype transition is accompanied by an enhanced expression of laminin-332, especially of its γ2 chain. We further analyse the expression of extracellular matrix related genes by RT-PCR profiling. With respect to strongly enhanced proteins, data confirm the EMT phenotype of co-stimulated OSCC cells and expression of laminin-332. Furthermore, alpha catenin, collagen type 16, the integrin α7 and β1 chains, and MMP11 are suggested as candidates with potential role in EMT in OSCC. In summary we are able to show that EMT in OSCC is mediated by multiple growth factors and is accompanied by laminin γ2 chain up-regulation evidencing the existence of an intermediate Vim(+) /Ln332(+) EMT phenotype as seen in situ.

    Topics: Carcinoma, Squamous Cell; Cell Adhesion Molecules; Cell Differentiation; Cell Line, Tumor; Cell Transformation, Neoplastic; Cells, Cultured; Epidermal Growth Factor; Epithelial Cells; Extracellular Matrix Proteins; Humans; Kalinin; Laminin; Mouth Neoplasms; Neoplasm Invasiveness; Transforming Growth Factor beta1; Vimentin

2011
Eriodictyol inhibits RSK2-ATF1 signaling and suppresses EGF-induced neoplastic cell transformation.
    The Journal of biological chemistry, 2011, Jan-21, Volume: 286, Issue:3

    RSK2 is a widely expressed serine/threonine kinase, and its activation enhances cell proliferation. Here, we report that ATF1 is a novel substrate of RSK2 and that RSK2-ATF1 signaling plays an important role in EGF-induced neoplastic cell transformation. RSK2 phosphorylated ATF1 at Ser-63 and enhanced ATF1 transcriptional activity. Docking experiments using the crystal structure of the RSK2 N-terminal kinase domain combined with in vitro pulldown assays demonstrated that eriodictyol, a flavanone found in fruits, bound with the N-terminal kinase domain of RSK2 to inhibit RSK2 N-terminal kinase activity. In cells, eriodictyol inhibited phosphorylation of ATF1 but had no effect on the phosphorylation of RSK, MEK1/2, ERK1/2, p38 or JNKs, indicating that eriodictyol specifically suppresses RSK2 signaling. Furthermore, eriodictyol inhibited RSK2-mediated ATF1 transactivation and tumor promoter-induced transformation of JB6 Cl41 cells. Eriodictyol or knockdown of RSK2 or ATF1 also suppressed Ras-mediated focus formation. Overall, these results indicate that RSK2-ATF1 signaling plays an important role in neoplastic cell transformation and that eriodictyol is a novel natural compound for suppressing RSK2 kinase activity.

    Topics: Activating Transcription Factor 1; Animals; Cell Line; Cell Transformation, Neoplastic; Epidermal Growth Factor; Flavanones; Mice; Mice, Knockout; Mitogen-Activated Protein Kinase Kinases; Phosphorylation; Protein Structure, Tertiary; ras Proteins; Ribosomal Protein S6 Kinases, 90-kDa; Signal Transduction; Transcriptional Activation

2011
Epidermal growth factor receptor (EGFR) is overexpressed in high-grade dysplasia and adenocarcinoma of the esophagus and may represent a biomarker of histological progression in Barrett's esophagus (BE).
    The American journal of gastroenterology, 2011, Volume: 106, Issue:1

    The assessment of cancer risk in patients with Barrett's esophagus (BE) is currently fraught with difficulty. The current gold standard method of assessing cancer risk is histological assessment, with the appearance of high-grade dysplasia (HGD) as the key event monitored. Sampling error during endoscopy limits the usefulness of this approach, and there has been much recent interest in supplementing histological assessment with molecular markers, which may aid in patient stratification.. No molecular marker has been yet validated to accurately correlate with esophageal histological progression. Here, we assessed the suitability of several membranous proteins as biomarkers by correlating their abundance with histological progression. In all, 107 patient samples, from 100 patients, were arranged on a tissue microarray (TMA) and represented the various stages of histological progression in BE. This TMA was probed with antibodies for eight receptor proteins (mostly membranous).. Epidermal growth factor receptor (EGFR) staining was found to be the most promising biomarker identified with clear increases in staining accompanying histological progression. Further, immunohistochemistry was performed using the full-tissue sections from BE, HGD, and adenocarcinoma tissues, which confirmed the stepwise increase in EGFR abundance. Using a robust H-score analysis, EGFR abundance was shown to increase 13-fold in the adenocarcinoma tissues compared to the BE tissues. EGFR was "overexpressed" in 35% of HGD specimens and 80% of adenocarcinoma specimens when using the H-score of the BE patients (plus 3 s.d.) as the threshold to define overexpression. EGFR staining was also noted to be higher in BE tissues adjacent to HGD/adenocarcinoma. Western blotting, although showing more EGFR protein in the adenocarcinomas compared to the BE tissue, was highly variable. EGFR overexpression was accompanied by aneuploidy (gain) of chromosome 7, plus amplification of the EGFR locus. Finally, the bile acid deoxycholic acid (DCA) (at neutral and acidic pH) and acid alone was capable of upregulating EGFR mRNA in vitro, and in the case of neutral pH DCA, this was NF-κB dependent.. EGFR is overexpressed during the histological progression in BE tissues and hence may be useful as a biomarker of histological progression. Furthermore, as EGFR is a membranous protein expressed on the luminal surface of the esophageal mucosa, it may also be a useful target for biopsy guidance during endoscopy.

    Topics: Adenocarcinoma; Aged; Barrett Esophagus; Biomarkers, Tumor; Biopsy, Needle; Blotting, Western; Cell Transformation, Neoplastic; Cohort Studies; Disease Progression; Epidermal Growth Factor; Esophageal Neoplasms; Female; Gene Expression Regulation, Neoplastic; Humans; Immunohistochemistry; In Situ Hybridization, Fluorescence; Male; Middle Aged; Neoplasm Staging; Polymerase Chain Reaction; Precancerous Conditions; Predictive Value of Tests; Prognosis; RNA, Messenger; Up-Regulation

2011
P21-activated protein kinase (PAK2)-mediated c-Jun phosphorylation at 5 threonine sites promotes cell transformation.
    Carcinogenesis, 2011, Volume: 32, Issue:5

    The oncoprotein c-Jun is one of the components of the activator protein-1 (AP-1) transcription factor complex. AP-1 regulates the expression of many genes and is involved in a variety of biological functions such as cell transformation, proliferation, differentiation and apoptosis. AP-1 activates a variety of tumor-related genes and therefore promotes tumorigenesis and malignant transformation. Here, we found that epidermal growth factor (EGF) induces phosphorylation of c-Jun by P21-activated kinase (PAK) 2. Our data showed that PAK2 binds and phosphorylates c-Jun at five threonine sites (Thr2, Thr8, Thr89, Thr93 and Thr286) in vitro and ex vivo. Knockdown of PAK2 in JB6 Cl41 (P+) cells had no effect on c-Jun phosphorylation at Ser63 or Ser73 but resulted in decreases in EGF-induced anchorage-independent cell transformation, proliferation and AP-1 activity. Mutation at all five c-Jun threonine sites phosphorylated by PAK2 decreased the transforming ability of JB6 cells. Knockdown of PAK2 in SK-MEL-5 melanoma cells also decreased colony formation, proliferation and AP-1 activity. These results indicated that PAK2/c-Jun signaling plays an important role in EGF-induced cell proliferation and transformation.

    Topics: Animals; Blotting, Western; Cell Proliferation; Cell Transformation, Neoplastic; Cells, Cultured; Epidermal Cells; Epidermal Growth Factor; Epidermis; Humans; Immunoenzyme Techniques; Immunoprecipitation; Melanoma; Mice; Mutation; p21-Activated Kinases; Phosphorylation; Proto-Oncogene Proteins c-jun; RNA, Small Interfering; Skin; Skin Neoplasms; Threonine; Tissue Array Analysis; Transcription Factor AP-1

2011
Phosphorylation of H2AX at Ser139 and a new phosphorylation site Ser16 by RSK2 decreases H2AX ubiquitination and inhibits cell transformation.
    Cancer research, 2011, Jan-15, Volume: 71, Issue:2

    Histone H2AX is a histone H2A variant that is ubiquitously expressed throughout the genome. It plays a key role in the cellular response to DNA damage and has been designated as the histone guardian of the genome. Histone H2AX deficiency decreases genomic stability and increases tumor susceptibility of normal cells and tissues. However, the role of histone H2AX phosphorylation in malignant transformation and cancer development is not totally clear. Herein, we found that ribosomal S6 kinase 2 (RSK2) directly phosphorylates histone H2AX at Ser139 and also at a newly discovered site, Ser16. Epidermal growth factor (EGF)-induced phosphorylation of histone H2AX at both sites was decreased in RSK2 knockout cells. Phosphorylated RSK2 and histone H2AX colocalized in the nucleus following EGF treatment, and the phosphorylation of histone H2AX by RSK2 enhanced the stability of histone H2AX and prevented cell transformation induced by EGF. RSK2 and DNA-PK, but not ATM or ATR, are required for EGF-induced phosphorylation of H2AX at Ser139; however, only RSK2 is required for phosphorylation of H2AX at Ser16. Phosphorylation of histone H3 was suppressed in cells expressing wild-type H2AX compared with H2AX knockout (H2AX-/-) cells. EGF-associated AP-1 transactivation activity was dramatically lower in H2AX-/- cells overexpressing wild-type H2AX than H2AX-/- cells expressing mutant H2AX-AA. Thus, the RSK2/H2AX signaling pathway negatively regulates the RSK2/histone H3 pathway and therefore maintains normal cell proliferation.

    Topics: Ataxia Telangiectasia Mutated Proteins; Cell Adhesion; Cell Cycle Proteins; Cell Growth Processes; Cell Transformation, Neoplastic; DNA-Activated Protein Kinase; DNA-Binding Proteins; Epidermal Growth Factor; HeLa Cells; Histones; Humans; Phosphorylation; Protein Serine-Threonine Kinases; Ribosomal Protein S6 Kinases, 90-kDa; Serine; Tumor Suppressor Proteins; Ubiquitination

2011
Loss of BRCA1 leads to an increase in epidermal growth factor receptor expression in mammary epithelial cells, and epidermal growth factor receptor inhibition prevents estrogen receptor-negative cancers in BRCA1-mutant mice.
    Breast cancer research : BCR, 2011, Mar-11, Volume: 13, Issue:2

    Women who carry a BRCA1 mutation typically develop "triple-negative" breast cancers (TNBC), defined by the absence of estrogen receptor (ER), progesterone receptor and Her2/neu. In contrast to ER-positive tumors, TNBCs frequently express high levels of epidermal growth factor receptor (EGFR). Previously, we found a disproportionate fraction of progenitor cells in BRCA1 mutation carriers with EGFR overexpression. Here we examine the role of EGFR in mammary epithelial cells (MECs) in the emergence of BRCA1-related tumors and as a potential target for the prevention of TNBC.. Cultures of MECs were used to examine EGFR protein levels and promoter activity in response to BRCA1 suppression with inhibitory RNA. EGFR was assessed by immunoblot and immunofluorescence analysis, real-time reverse transcriptase-polymerase chain reaction assay (RT-PCR) and flow cytometry. Binding of epidermal growth factor (EGF) to subpopulations of MECs was examined by Scatchard analysis. The responsiveness of MECs to the EGFR inhibitor erlotinib was assessed in vitro in three-dimensional cultures and in vivo. Mouse mammary tumor virus-Cre recombinase (MMTV-Cre) BRCA1flox/flox p53⁺/⁻ mice were treated daily with erlotinib or vehicle control, and breast cancer-free survival was analyzed using the Kaplan-Meier method.. Inhibition of BRCA1 in MECs led to upregulation of EGFR with an inverse correlation of BRCA1 with cellular EGFR protein levels (r² = 0.87) and to an increase in cell surface-expressed EGFR. EGFR upregulation in response to BRCA1 suppression was mediated by transcriptional and posttranslational mechanisms. Aldehyde dehydrogenase 1 (ALDH1)-positive MECs expressed higher levels of EGFR than ALDH1-negative MECs and were expanded two- to threefold in the BRCA1-inhibited MEC population. All MECs were exquisitely sensitive to EGFR inhibition with erlotinib in vitro. EGFR inhibition in MMTV-Cre BRCA1flox/flox p53⁺/⁻ female mice starting at age 3 months increased disease-free survival from 256 days in the controls to 365 days in the erlotinib-treated cohort.. We propose that even partial loss of BRCA1 leads to an overall increase in EGFR expression in MECs and to an expansion of the highly EGFR-expressing, ALDH1-positive fraction. Increased EGFR expression may confer a growth advantage to MECs with loss of BRCA1 at the earliest stages of transformation. Employing EGFR inhibition with erlotinib specifically at this premalignant stage was effective in decreasing the incidence of ER-negative breast tumors in this mouse model.

    Topics: Aldehyde Dehydrogenase 1 Family; Animals; BRCA1 Protein; Breast; Cell Line, Tumor; Cell Transformation, Neoplastic; Cells, Cultured; Epidermal Growth Factor; Epithelial Cells; ErbB Receptors; Erlotinib Hydrochloride; Female; Genes, BRCA1; Genes, p53; Humans; Isoenzymes; Mammary Glands, Animal; Mammary Neoplasms, Experimental; Mammary Tumor Virus, Mouse; Mice; Mice, Inbred C57BL; Mice, Knockout; Protein Kinase Inhibitors; Quinazolines; Receptors, Estrogen; Retinal Dehydrogenase; RNA Interference

2011
5'-Nitro-indirubinoxime inhibits epidermal growth factor- and phorbol ester-induced AP-1 activity and cell transformation through inhibition of phosphorylation of Pin1.
    Molecular carcinogenesis, 2011, Volume: 50, Issue:12

    5'-Nitro-indirubinoxime (5'-NIO), a derivative of indirubin, exhibits anti-cancer activity in a variety of human cancer cells. However, the underlying molecular mechanisms and molecular target(s) of the chemopreventive activities of 5'-NIO remain unknown. Here, we report that 5'-NIO inhibited the epidermal growth factor (EGF) or 12-O-tetradecanoylphorbol-13-acetate (TPA)-induced neoplastic cell transformation of JB6 Cl41 mouse skin epidermal cells without any cytotoxic effects. Western blot analysis revealed that 5'-NIO inhibited activities of Raf-1 (S338), MEK1/2, ERK1/2, JNK, and c-Jun induced by EGF or TPA, respectively, whereas it did not affect autophosphorylation of epidermal growth factor receptor (EGFR) induced by EGF or TPA. In addition, 5'-NIO exerted strong inhibitory effects on the EGF- or TPA-induced c-fos or c-jun transcriptional activity, and thereby inhibited the associated activator protein-1 (AP-1) transactivation activity induced by EGF or TPA. Importantly, 5'-NIO inhibited Pin1 phosphorylation at serine 16 induced by EGF or TPA, respectively, resulted in the inhibition of interaction between Pin1 and Raf-1. Immunoprecipitation/immunoblot analysis revealed that 5'-NIO bound with Pin1. Together, these findings suggest that 5'-NIO might act as an anticarcinogene in EGF- or TPA-induced carcinogenesis through the inhibition of interaction between Pin1 and Raf-1. © 2011 Wiley Periodicals, Inc.

    Topics: Animals; Anticarcinogenic Agents; Cell Line; Cell Transformation, Neoplastic; Epidermal Growth Factor; Indoles; JNK Mitogen-Activated Protein Kinases; MAP Kinase Signaling System; Mice; Mitogen-Activated Protein Kinases; NIMA-Interacting Peptidylprolyl Isomerase; Oximes; Peptidylprolyl Isomerase; Phosphorylation; Proto-Oncogene Proteins c-fos; Proto-Oncogene Proteins c-raf; Tetradecanoylphorbol Acetate; Transcription Factor AP-1; Transcription, Genetic; Transcriptional Activation

2011
Phosphorylation of histone H2B serine 32 is linked to cell transformation.
    The Journal of biological chemistry, 2011, Jul-29, Volume: 286, Issue:30

    Various types of post-translational modifications of the histone tails have been revealed, but a few modifications have been found within the histone core sequences. Histone core post-translational modifications have the potential to modulate nucleosome structure and DNA accessibility. Here, we studied the histone H2B core domain and found that phosphorylation of H2B serine 32 occurs in normal cycling and mitogen-stimulated cells. Notably, this phosphorylation is elevated in skin cancer cell lines and tissues compared with normal counterparts. The JB6 Cl41 mouse skin epidermal cell line is a well established model for tumor promoter-induced cell transformation and was used to study the function of H2B during EGF-induced carcinogenesis. Remarkably, cells overexpressing a nonphosphorylatable H2BS32A mutant exhibited suppressed growth and EGF-induced cell transformation, possibly because of decreased activation of activator protein-1, compared with control cells overexpressing wild type H2B. We identified ribosomal S6 kinase 2 (RSK2) as the kinase responsible for H2BS32 phosphorylation. Serum-starved JB6 cells contain very little endogenous H2BS32 phosphorylation, and EGF treatment induced this phosphorylation. The phosphorylation was attenuated in RSK2 knock-out MEFs and RSK2 knockdown JB6 cells. Taken together, our results demonstrate a novel role for H2B phosphorylation in cell transformation and show that H2BS32 phosphorylation is critical for controlling activator protein-1 activity, which is a major driver in cell transformation.

    Topics: Amino Acid Substitution; Animals; Cell Transformation, Neoplastic; Epidermal Growth Factor; Epidermis; HEK293 Cells; Histones; Humans; Mice; Mice, Knockout; Mutation, Missense; Phosphorylation; Ribosomal Protein S6 Kinases, 90-kDa; Serine; Transcription Factor AP-1

2011
A molecular screening approach to identify and characterize inhibitors of glioblastoma stem cells.
    Molecular cancer therapeutics, 2011, Volume: 10, Issue:10

    Glioblastoma (GBM) is among the most lethal of all cancers. GBM consist of a heterogeneous population of tumor cells among which a tumor-initiating and treatment-resistant subpopulation, here termed GBM stem cells, have been identified as primary therapeutic targets. Here, we describe a high-throughput small molecule screening approach that enables the identification and characterization of chemical compounds that are effective against GBM stem cells. The paradigm uses a tissue culture model to enrich for GBM stem cells derived from human GBM resections and combines a phenotype-based screen with gene target-specific screens for compound identification. We used 31,624 small molecules from 7 chemical libraries that we characterized and ranked based on their effect on a panel of GBM stem cell-enriched cultures and their effect on the expression of a module of genes whose expression negatively correlates with clinical outcome: MELK, ASPM, TOP2A, and FOXM1b. Of the 11 compounds meeting criteria for exerting differential effects across cell types used, 4 compounds showed selectivity by inhibiting multiple GBM stem cells-enriched cultures compared with nonenriched cultures: emetine, n-arachidonoyl dopamine, n-oleoyldopamine (OLDA), and n-palmitoyl dopamine. ChemBridge compounds #5560509 and #5256360 inhibited the expression of the 4 mitotic module genes. OLDA, emetine, and compounds #5560509 and #5256360 were chosen for more detailed study and inhibited GBM stem cells in self-renewal assays in vitro and in a xenograft model in vivo. These studies show that our screening strategy provides potential candidates and a blueprint for lead compound identification in larger scale screens or screens involving other cancer types.

    Topics: Animals; Antineoplastic Agents; Brain Neoplasms; Cell Growth Processes; Cell Line, Tumor; Cell Transformation, Neoplastic; Culture Media, Serum-Free; Emetine; Epidermal Growth Factor; Fibroblast Growth Factors; Gene Expression; Glioblastoma; High-Throughput Screening Assays; Humans; Mice; Mice, Inbred NOD; Mice, SCID; Neoplastic Stem Cells; Xenograft Model Antitumor Assays

2011
Mammary tumorigenesis induced by fibroblast growth factor receptor 1 requires activation of the epidermal growth factor receptor.
    Journal of cell science, 2011, Sep-15, Volume: 124, Issue:Pt 18

    Fibroblast growth factor receptor 1 (FGFR1) is an oncoprotein with known involvement in mammary tumorigenesis. To understand how FGFR1 signaling promotes mammary tumorigenesis, an inducible FGFR1 (iFGFR1) system was created previously. Previous studies have demonstrated that upon iFGFR1 activation in vivo, the epidermal growth factor (EGF) ligands amphiregulin (AREG) and epiregulin (EREG) are upregulated. Both AREG and EREG interact with the EGF receptor (EGFR). Here, we investigated whether the FGFR1-induced increase in AREG and EREG expression might coordinately increase EGFR signaling to promote mammary tumorigenesis. Treatment of mouse mammary epithelial cells with either AREG or EREG conferred a greater migratory potential, increased cellular proliferation and increased extracellular regulated kinase 1/2 (ERK1/2) activation. These effects could be blocked with the EGFR-specific inhibitor erlotinib, suggesting that they are EGFR-dependent. In transgenic mice with iFGFR1 under the control of the mouse mammary tumor virus (MMTV) promoter, iFGFR1 activation also led to increased mammary epithelial cell proliferation that was inhibited with erlotinib. Taken together, these data suggest that AREG and EREG mediate tumorigenic phenotypes by activating EGFR signaling, and that the oncogenic potential of FGFR1 requires EGFR activation to promote mammary tumorigenesis.

    Topics: Amphiregulin; Animals; Breast Neoplasms; Cell Growth Processes; Cell Line; Cell Movement; Cell Transformation, Neoplastic; EGF Family of Proteins; Epidermal Growth Factor; Epiregulin; ErbB Receptors; Erlotinib Hydrochloride; Female; Gene Expression Regulation, Neoplastic; Glycoproteins; Humans; Intercellular Signaling Peptides and Proteins; Mammary Glands, Human; MAP Kinase Signaling System; Mice; Mice, Transgenic; Quinazolines; Receptor, Fibroblast Growth Factor, Type 1

2011
Transformed epithelial cells and fibroblasts/myofibroblasts interaction in breast tumor: a mathematical model and experiments.
    Journal of mathematical biology, 2010, Volume: 61, Issue:3

    It is well known that tumor and its microenvironment, or stroma, interact with each other and that this interaction plays a critical role in tumor initiation, growth, and metastasis. This interaction consists of complex relations between tumor cells, stromal cells such as fibroblasts, epithelial cells and immunocytes, the vascular system, the extracellular matrix, and cytokines secreted by the cells. Understanding these relationships may lead to new therapeutic approaches to cancer. In the present paper, we consider tumor-stroma crosstalk in a simple in vitro situation which involves interaction between tumor epithelial cells from breast cancer and a microenvironment consisting of just fibroblasts. The two populations of cells are separated by a semi-permeable membrane that allows only cytokines to cross over. We develop a mathematical model that includes two critical growth factors: TGF-beta, produced by the tumor cells, and EGF, secreted by the fibroblasts. The TGF-beta modifies the microenvironment by transforming fibroblasts into myofibroblasts. Myofibroblasts secrete higher concentrations of EGF than fibroblasts, thereby, increasing the proliferation of tumor cells. Thus already in this simple setup one sees a mutual interaction between tumor cells and their microenvironment. We conducted experiments which show good agreement with the model's simulations, hence confirming the model's ability to predict aspects of tumor cell behavior in response to signaling from fibroblasts.

    Topics: Breast Neoplasms; Cell Communication; Cell Transformation, Neoplastic; Computer Simulation; Epidermal Growth Factor; Epithelial Cells; Female; Fibroblasts; Humans; Models, Immunological; Transforming Growth Factor beta

2010
The prolyl isomerase Pin1 enhances HER-2 expression and cellular transformation via its interaction with mitogen-activated protein kinase/extracellular signal-regulated kinase kinase 1.
    Molecular cancer therapeutics, 2010, Volume: 9, Issue:3

    The HER-2 oncogene, a member of the erythroblastosis oncogene B (ERBB)-like oncogene family, has been shown to be amplified in many types of cancer, including breast cancer. However, the molecular mechanism of HER-2 overexpression is not completely understood. The phosphorylation of proteins on the serine or threonine residues that immediately precede proline (pSer/Thr-Pro) is specifically catalyzed by the prolyl isomerase Pin1 and is a key signaling mechanism in cell proliferation and transformation. Here, we found that Pin1 interacts with mitogen-activated protein kinase/extracellular signal-regulated kinase kinase (MEK) protein kinase 1, resulting in the induction of HER-2 expression. Pin1(-/-) mouse embryonic fibroblasts exhibited a decrease in epidermal growth factor (EGF)-induced MEK1/2 phosphorylation compared with Pin1(+/+) mouse embryonic fibroblast. In addition, a knockdown of Pin1 resulted in the inhibition of MEK1/2 phosphorylation induced by EGF in MCF-7 cells. Furthermore, PD98059, a specific inhibitor of MEK1/2, and Juglone, a potent Pin1 inhibitor, markedly suppressed the expression of activator protein-2alpha and the HER-2 promoter activity induced by EGF or 12-O-tetradecanoylphorbol-13-acetate in MCF-7 cells. Importantly, these inhibitors inhibited the neoplastic cell transformation induced by EGF in Pin1-overexpressing JB6 Cl41 cells, which showed enhanced cellular formation compared with the control cells. Therefore, Juglone and PD98059 inhibited the colony formation of MCF-7 breast cancer cells in soft agar. These results indicate that Pin1 amplifies EGF signaling in breast cancer cells through its interaction with MEK1 and then enhances HER-2 expression, suggesting that Pin1 plays an important role in the overexpression of HER-2 through Pin1-MEK1-activator protein-2alpha signaling in breast cancer.

    Topics: Animals; Breast Neoplasms; Cell Transformation, Neoplastic; Cells, Cultured; Epidermal Growth Factor; Female; Gene Expression Regulation, Neoplastic; Humans; MAP Kinase Kinase 1; MAP Kinase Kinase 2; Mice; Mice, Knockout; Mitogen-Activated Protein Kinase 3; NIMA-Interacting Peptidylprolyl Isomerase; Peptidylprolyl Isomerase; Phosphorylation; Protein Binding; Receptor, ErbB-2; Up-Regulation

2010
The interplay between Eps8 and IRSp53 contributes to Src-mediated transformation.
    Oncogene, 2010, Jul-08, Volume: 29, Issue:27

    As an oncoprotein, Eps8 participates in v-Src-induced cellular transformation. To delineate the underlying mechanism, we conducted a yeast two-hybrid screening and identified IRSp53S, a protein critical in cell mobilization, as one of the Eps8-binding partners from a human brain cDNA library. The association was mediated by the multiple proline-rich regions of Eps8 and the C-terminal SH3-WWB containing domains of IRSp53S. In this study, we observed that Eps8 modulated the expression of IRSp53 in v-Src-transformed cells (IV5), raising the question of whether Eps8/IRSp53 interaction was crucial in carcinogenesis. To address this issue, we generated IV5-expressing irsp53 siRNA cells. Attenuation of IRSp53 reduced cell proliferation of IV5 in culture dish and tumor formation in mice, which could be partly rescued by ectopically expressed human IRSp53S. In addition, IRSp53 knockdown impaired activity of phosphatidylinositol 3-kinase (as reflected by Pi-Ser473 AKT) and Stat3 (as reflected by Pi-Tyr705 Stat3), and reduced cyclin D1 expression that culminated to impede G(1)-phase cell-cycle progression. Ectopically expressed human IRSp53S, but not its Eps8-binding defective mutants (that is, Delta363 and PPPDA), rescued these defects and partly restored cell proliferation. Remarkably, through activation of Src, EGF increased the formation of Eps8/IRSp53 complex and Stat3 activation in HeLa cells. With these results, we show for the first time that IRSp53, through its interaction with Eps8, not only affects cell migration but also dictates cellular growth in cancer cells.

    Topics: Adaptor Proteins, Signal Transducing; Animals; Cell Cycle; Cell Proliferation; Cell Transformation, Neoplastic; Cyclin D1; Enzyme Activation; Epidermal Growth Factor; Gene Expression Regulation, Neoplastic; HeLa Cells; Humans; Intracellular Signaling Peptides and Proteins; Mice; Nerve Tissue Proteins; Oncogene Protein pp60(v-src); Protein Binding; Protein Structure, Tertiary; Proto-Oncogene Proteins c-akt; RNA, Small Interfering; Sequence Deletion; STAT3 Transcription Factor; Two-Hybrid System Techniques

2010
ADAM17 regulates epidermal growth factor receptor expression through the activation of Notch1 in non-small cell lung cancer.
    Cancer research, 2010, Jul-01, Volume: 70, Issue:13

    Epidermal growth factor receptor (EGFR) overexpression and activation are hallmarks of non-small cell lung carcinoma (NSCLC). Although EGFR-targeted therapies are used, the prognosis of NSCLC remains poor. ADAM17 induces activation of the EGFR through ligand cleavage. However, we show that inhibition or knockdown of ADAM17 markedly reduces tumorigenesis and survival to a large part independently from EGFR ligand shedding in NSCLC cells. These findings strongly indicate additional oncogenic mechanisms regulated by ADAM17. We identified Notch1 signaling as an ADAM17-controlled pathway and a critical regulator of anchorage-independent growth by using both Notch1 shRNA and ectopic expression of the active intracellular Notch1 fragment. Strikingly, Notch1 knockdown led to a strong reduction of EGFR expression in all analyzed cell lines. Proliferation, survival, and colony formation of Notch1-deficient cells were insensitive to EGF stimulation. Moreover, targeting Notch1 or ADAM17 resulted in substantial cell death, whereas EGFR inhibition predominantly induced cell cycle arrest. Immunohistochemical analysis of primary human tissue revealed a significant correlation between ADAM17, Notch1 signaling, and high EGFR expression levels. In conclusion, this article describes a novel molecular circuitry in NSCLC, incorporating ADAM17 as a regulator of EGFR expression through the activation of Notch1. Due to their central role in tumorigenesis and survival of NSCLC cells, both ADAM17 and Notch1 constitute promising targets for the treatment of NSCLC.

    Topics: ADAM Proteins; ADAM17 Protein; Animals; Basic Helix-Loop-Helix Transcription Factors; Carcinoma, Non-Small-Cell Lung; Cell Growth Processes; Cell Line, Tumor; Cell Survival; Cell Transformation, Neoplastic; Epidermal Growth Factor; ErbB Receptors; Homeodomain Proteins; Humans; Lung Neoplasms; Mice; Receptor, Notch1; Signal Transduction; Transcription Factor HES-1; Transplantation, Heterologous

2010
Phosphorylation and activation of cell division cycle associated 5 by mitogen-activated protein kinase play a crucial role in human lung carcinogenesis.
    Cancer research, 2010, Jul-01, Volume: 70, Issue:13

    We analyzed the gene expression profiles of clinical lung carcinomas using a cDNA microarray containing 27,648 genes or expressed sequence tags, and identified CDCA5 (cell division cycle associated 5) to be upregulated in the majority of lung cancers. Tumor tissue microarray analysis of 262 non-small cell lung cancer patients revealed that CDCA5 positivity was an independent prognostic factor for lung cancer patients. Suppression of CDCA5 expression with siRNAs inhibited the growth of lung cancer cells; concordantly, induction of exogenous expression of CDCA5 conferred growth-promoting activity in mammalian cells. We also found that extracellular signal-regulated kinase (ERK) kinase phosphorylated CDCA5 at Ser79 and Ser209 in vivo. Exogenous expression of phospho-mimicking CDCA5 protein whose Ser209 residue was replaced with glutamine acid further enhanced the growth of cancer cells. In addition, functional inhibition of the interaction between CDCA5 and ERK kinase by a cell-permeable peptide corresponding to a 20-amino-acid sequence part of CDCA5, which included the Ser209 phosphorylation site by ERK, significantly reduced phosphorylation of CDCA5 and resulted in growth suppression of lung cancer cells. Our data suggest that transactivation of CDCA5 and its phosphorylation at Ser209 by ERK play an important role in lung cancer proliferation, and that the selective suppression of the ERK-CDCA5 pathway could be a promising strategy for cancer therapy.

    Topics: Adaptor Proteins, Signal Transducing; Amino Acid Sequence; Animals; Carcinoma, Non-Small-Cell Lung; Carcinoma, Small Cell; Cell Cycle Proteins; Cell Growth Processes; Cell Line, Tumor; Cell Transformation, Neoplastic; Chlorocebus aethiops; COS Cells; Epidermal Growth Factor; Extracellular Signal-Regulated MAP Kinases; Gene Expression Profiling; Gene Knockdown Techniques; Humans; Lung Neoplasms; MAP Kinase Signaling System; Molecular Sequence Data; Peptides; Phosphorylation; RNA, Small Interfering; Transcriptional Activation

2010
Antisense inhibition of amphiregulin expression reduces EGFR phosphorylation in transformed human breast epithelial cells.
    Anticancer research, 2010, Volume: 30, Issue:6

    The activation of epidermal growth factor receptor (EGFR) by its ligands constitutes an important step in the metastatic process but the clinical response to its inhibition in breast cancer patients has so far been very low. In this work, we investigated the role of the EGFR ligand amphiregulin (AR) in modulating EGFR activation. For this, transformed epithelial mammary tumor cells NS2T2A1 were used in which AR or EGFR expression was down-regulated by antisense cDNA technique. This down-regulation was associated with a significant inhibition of matrix metalloproteinase-9 production as well as cell proliferation, but this inhibition was only minimally reversed by exogenously added AR or EGF. EGFR protein levels were not affected but EGFR-tyrosine phosphorylation in response to EGF was markedly reduced. Thus, the inhibition of AR expression, which impairs EGFR response to its exogenously available ligands, may represent an alternative anti-EGFR therapeutic strategy in breast cancer.

    Topics: Amphiregulin; Antisense Elements (Genetics); Breast Neoplasms; Cell Line, Tumor; Cell Proliferation; Cell Transformation, Neoplastic; EGF Family of Proteins; Epidermal Growth Factor; ErbB Receptors; Female; Glycoproteins; Humans; Intercellular Signaling Peptides and Proteins; Matrix Metalloproteinase 9; Matrix Metalloproteinase Inhibitors; Phosphorylation; Tyrosine

2010
Gene expression of oncogenes, antimicrobial peptides, and cytokines in the development of oral leukoplakia.
    Oral surgery, oral medicine, oral pathology, oral radiology, and endodontics, 2010, Volume: 110, Issue:3

    The aim of this study was to investigate the expression pattern of oncogenes, antimicrobial peptides, and genes involved in inflammation in leukoplakia of the oral cavity compared with healthy gingiva.. Biopsies of healthy gingiva (n=20) and leukoplakia (n=20), were obtained during routine surgical procedures. RNA was extracted according to standard protocols. Transcript levels of alpha-defensin (DEFA) 1/3, DEFA-4, S100-A7, deleted-in-oral-cancer (Doc) 1, interleukin (IL) 1beta, IL-6, IL-8, IL-10, tumor necrosis factor (TNF) alpha, cyclooxygenase (Cox) 2, epidermal growth factor (EGF), keratinocyte growth factor (KGF), transforming growth factor (TGF) beta1, TGF-alpha, collagen-IA1 (Col-1), and tenascin-c were analyzed by real-time reverse-transcription polymerase chain reaction. The proteins encoded by the different genes were visualized by immunostaining.. Compared with healthy gingiva (set as 1), there was an increased gene expression of DEFA-4 (179.2-fold), S100-A7 (25.4-fold), EGF (24.8-fold), TGF-beta1 (25.2-fold), and tenascin-c (34.3-fold) in oral leukoplakia. The expression of IL-1beta and Doc-1 was decreased (0.01-fold and 0.2-fold, respectively).. The combination of an increased expression of the antimicrobial peptide DEFA-4, the oncogene S100-A7, EGF, and tenascin-c, and a decreased Doc-1 expression in oral leukoplakia might characterize its potency of malignant transformation. Chronic inflammation seems not to be involved in the development of this lesion.

    Topics: alpha-Defensins; Case-Control Studies; Cell Differentiation; Cell Transformation, Neoplastic; Collagen Type I; Collagen Type I, alpha 1 Chain; Cyclooxygenase 2; Epidermal Growth Factor; Fibroblast Growth Factor 7; Gene Expression Profiling; Gingiva; Humans; Immunohistochemistry; Interleukins; Leukoplakia, Oral; Reference Values; RNA; S100 Calcium Binding Protein A7; S100 Proteins; Tenascin; Transforming Growth Factor alpha; Transforming Growth Factor beta1; Tumor Necrosis Factor-alpha; Tumor Suppressor Proteins

2010
HER-2 signaling, acquisition of growth factor independence, and regulation of biological networks associated with cell transformation.
    Cancer research, 2010, Oct-15, Volume: 70, Issue:20

    Activated oncogenes are the dominant drivers of malignant progression in human cancer, yet little is known about how the transformation from proto-oncogene to activated oncogene drives the expression of transformed phenotypes. An isogenic model of HER-2-mediated transformation of human mammary epithelial cells was used along with HER-2-amplified human breast cancers to investigate how HER-2 activation alters its properties as a signaling molecule and changes the networks of HER-2-regulated genes. Our results show that full oncogenic activation of HER-2 is the result of a transition in which activated HER-2 acquires dominant signaling properties that qualitatively alter the network of genes regulated by the activated oncogene compared with the proto-oncogene. Consequently, gene expression programs related to invasion, cell stress, and stemness become regulated by HER-2 in a manner not observed in nontransformed cells, even when HER-2 is overexpressed. Our results offer novel insights into biological processes that come under the control of HER-2 after it acquires full oncogenic potential.

    Topics: Breast Neoplasms; Cell Division; Cell Line, Tumor; Cell Transformation, Neoplastic; Epidermal Growth Factor; Female; Gene Expression Regulation, Neoplastic; Genes, Reporter; Humans; Intercellular Signaling Peptides and Proteins; Oligonucleotide Array Sequence Analysis; Phosphorylation; Proto-Oncogene Mas; Receptor, ErbB-2; Receptor, ErbB-3; Reverse Transcriptase Polymerase Chain Reaction; RNA, Messenger; Signal Transduction

2010
Myricetin directly targets JAK1 to inhibit cell transformation.
    Cancer letters, 2009, Mar-08, Volume: 275, Issue:1

    JAK1/STAT3 pathway has been suggested to play a role in cell transformation and carcinogenesis. In the present study, we found that myricetin (3, 3', 4', 5, 5', 7-hexahydroxyflavone), a typical flavonol existing in many fruits and vegetables, could directly bind to JAK1/STAT3 molecules to inhibit cell transformation in epidermal growth factor (EGF)-activated mouse JB6 P(+) cells. Colony assay revealed that myricetin had the strongest inhibitory effect on cell transformation among three flavonols including myricetin, quercetin and kaempferol. Molecular data revealed that myricetin inhibited DNA- binding and transcriptional activity of STAT3. Furthermore, myricetin inhibited the phosphorylation of STAT3 at Tyr705 and Ser727. Cellular signaling analyses revealed that EGF could induce the phosphorylation of Janus Kinase (JAK) 1, but not JAK2. Myricetin inhibited the phosphorylation of JAK1 and increased the autophosphorylation of EGF receptor (EGFR). Moreover, ex vivo and in vitro pull-down assay revealed that myricetin bound to JAK1 and STAT3, but not EGFR. Affinity data further demonstrated that myricetin had a higher affinity for JAK1 than STAT3. Thus, our data indicate that myricetin might directly target JAK1 to block cell transformation in mouse JB6 cells.

    Topics: Animals; Antioxidants; Cell Line; Cell Transformation, Neoplastic; DNA; Epidermal Growth Factor; Flavonoids; Janus Kinase 1; Kaempferols; Mice; Phosphorylation; Protein Binding; Quercetin; STAT3 Transcription Factor; Transcription, Genetic

2009
Cyclin-dependent kinase-3-mediated c-Jun phosphorylation at Ser63 and Ser73 enhances cell transformation.
    Cancer research, 2009, Jan-01, Volume: 69, Issue:1

    c-Jun is a component of the activator protein-1 (AP-1) complex, which plays a crucial role in the regulation of gene expression, cell proliferation, and cell transformation, as well as cancer development. Herein, we found that cyclin-dependent kinase (Cdk)-3, but not Cdk2 or c-Jun NH(2)-terminal kinase, is a novel kinase of c-Jun induced by stimulation with growth factors such as epidermal growth factor (EGF). Cdk3 was shown to phosphorylate c-Jun at Ser63 and Ser73 in vitro and ex vivo. EGF-induced Cdk3 activation caused c-Jun phosphorylation at Ser63 and Ser73, resulting in increased AP-1 transactivation. Ectopic expression of Cdk3 resulted in anchorage-independent cell transformation of JB6 Cl41 cells induced by EGF and foci formation stimulated by constitutively active Ras (Ras(G12V)), which was mediated by AP-1 in NIH3T3 cells. These results showed that the Cdk3/c-Jun signaling axis plays an important role in EGF-stimulated cell proliferation and cell transformation.

    Topics: Animals; Cell Adhesion; Cell Growth Processes; Cell Line; Cell Transformation, Neoplastic; Cyclin-Dependent Kinase 3; Cyclin-Dependent Kinases; Epidermal Growth Factor; Humans; Mice; NIH 3T3 Cells; Phosphorylation; Proto-Oncogene Proteins c-jun; Serine; Signal Transduction; Transcription Factor AP-1; Transfection

2009
Tpl-2 kinase downregulates the activity of p53 and enhances signaling pathways leading to activation of activator protein 1 induced by EGF.
    Carcinogenesis, 2009, Volume: 30, Issue:4

    Tumor progression locus-2 (Tpl-2) kinase is a member of the mitogen-activated protein kinase kinase kinase family that has been implicated in cellular transformation. The enhanced expression of this protein has been shown to activate both the mitogen-activated protein kinase and c-Jun N-terminal kinase pathways. However, the molecular mechanisms responsible for the oncogenic potential of Tpl-2 are still largely unknown. Here, we showed that Tpl-2 interacted with p53 both in vitro and ex vivo. The overexpression of Tpl-2 inhibited the epidermal growth factor (EGF)-induced p53 phosphorylation (Ser15) through upregulating the activity of protein phosphatase 2A, which interacted with p53 stimulated by EGF. Also, the EGF-induced p53 activity was suppressed in the Tpl-2 wild-type (WT)-transfected HEK 293 cells, but had no effect in the Tpl-2-mutant (S413A)-transfected cells. Furthermore, introduction of small interfering RNA-Tpl-2 into HEK 293 cells resulted in decreased cell viability compared with only adenovirus-p53-infected cells. In addition, the Tpl-2 WT, but not Tpl-2 mutant (S413A), showed increased EGF-induced c-fos promoter activity, followed by activator protein 1 (AP-1) transactivation activity, which was associated with the cell transformation prompted by the H-Ras-Tpl-2-AP-1 signaling axis. These results indicated that the Ser413 of Tpl-2 plays an important role in EGF-induced carcinogenesis as well as inactivation of the p53.

    Topics: Adenoviridae; Animals; Cell Transformation, Neoplastic; Cells, Cultured; Down-Regulation; Epidermal Growth Factor; Genes, ras; Humans; Immunoblotting; Kidney; MAP Kinase Kinase Kinases; Mice; Mutation; NIH 3T3 Cells; Promoter Regions, Genetic; Protein Phosphatase 2; Proto-Oncogene Proteins; Proto-Oncogene Proteins c-fos; RNA, Small Interfering; Signal Transduction; Transcription Factor AP-1; Transcriptional Activation; Tumor Suppressor Protein p53

2009
Lung carcinogenesis induced by chronic tuberculosis infection: the experimental model and genetic control.
    Oncogene, 2009, Apr-30, Volume: 28, Issue:17

    Coexistence of pulmonary tuberculosis (TB) and lung cancer in clinic poses significant challenges for the diagnostic and treatment of both diseases. Although association of chronic inflammation and cancer is well-documented, causal relationship between TB infection and lung cancer are not understood. We present experimental evidence that chronic TB infection induces cell dysplasia and squamous cell carcinoma (SCC) in a lung-specific manner. First, squamous cell aggregates consistently appeared within the lung tissue associated with chronic TB lesions, and in some cases resembled SCCs. A transplantable tumor was established after the transfer of cells isolated from TB lung lesions into syngeneic recipients. Second, the (Mycobacterium tuberculosis) MTB-infected macrophages play a pivotal role in TB-induced carcinogenesis by inducing DNA damage in their vicinity and by the production of a potent epidermal growth factor epiregulin, which may serve as a paracrine survival and growth factor responsible for squamous metaplasia and tumorigenesis. Third, lung carcinogenesis during the course of chronic TB infection was more pronounced in animals with severe lung tissue damage mediated by TB-susceptibility locus sst1. Together, our experimental findings showed a causal link between pulmonary TB and lung tumorigenesis and established a genetic model for further analysis of carcinogenic mechanisms activated by TB infection.

    Topics: Animals; Antitubercular Agents; Carcinoma, Squamous Cell; Cell Transformation, Neoplastic; Chronic Disease; Disease Models, Animal; Epidermal Growth Factor; Epiregulin; Female; Gene Expression; Genetic Predisposition to Disease; Host-Pathogen Interactions; Isoniazid; Lung; Lung Neoplasms; Macrophages; Male; Mice; Mice, Congenic; Mice, Inbred C3H; Mice, Inbred C57BL; Mice, Inbred Strains; Mycobacterium tuberculosis; Reverse Transcriptase Polymerase Chain Reaction; Time Factors; Tuberculosis, Pulmonary

2009
A regulatory mechanism for RSK2 NH(2)-terminal kinase activity.
    Cancer research, 2009, May-15, Volume: 69, Issue:10

    Our previous findings indicated that RSK2 plays a critical role in proliferation and cell transformation induced by tumor promoters, such as epidermal growth factor or 12-O-tetradecanoylphorbol-13-acetate, and that kaempferol, a natural compound found in edible plants, selectively inhibits RSK2 activity. However, the molecular mechanism for RSK2 activation is unclear. Herein, we provide evidence showing that NH(2)-terminal kinase domain (NTD) activation of RSK2 is required for the activation of the extracellular signal-regulated kinase-mediated COOH-terminal kinase domain (CTD). We also found that the NTD plays a key role in substrate phosphorylation and that kaempferol binds with the NTD but not the CTD in both the active and inactive forms. Homology modeling of the RSK2 NH(2)-terminal domain and small-molecule docking, validated by mutagenesis experiments, clearly showed that Val(82) and Lys(100) are critical amino acids for kaempferol binding and RSK2 activity. Furthermore, immunohistofluorescence and Western blot results indicated that the RSK2 protein level is markedly higher in cancer cell lines as well as cancer tissues compared with nonmalignant cell lines or normal tissues. In addition, kaempferol inhibited proliferation of malignant human cancer cell lines, including A431, SK-MEL-5 and SK-MEL-28, and HCT-116. These results indicate that targeting RSK2 with natural compounds, such as kaempferol, might be a good strategy for chemopreventive or chemotherapeutic application.

    Topics: Binding Sites; Cell Division; Cell Line, Tumor; Cell Transformation, Neoplastic; Epidermal Growth Factor; Humans; Kaempferols; Kinetics; Lysine; Neoplasms; Phosphorylation; Protein Kinases; Recombinant Proteins; Reference Values; Ribosomal Protein S6 Kinases, 90-kDa; Skin; Tetradecanoylphorbol Acetate; Tissue Array Analysis; Valine

2009
Monosomy of chromosome 10 associated with dysregulation of epidermal growth factor signaling in glioblastomas.
    JAMA, 2009, Jul-15, Volume: 302, Issue:3

    Glioblastomas--uniformly fatal brain tumors--often have both monosomy of chromosome 10 and gains of the epidermal growth factor receptor (EGFR) gene locus on chromosome 7, an association for which the mechanism is poorly understood.. To assess whether coselection of EGFR gains on 7p12 and monosomy 10 in glioblastomas promotes tumorigenic epidermal growth factor (EGF) signaling through loss of the annexin A7 (ANXA7) gene on 10q21.1-q21.2 and whether ANXA7 acts as a tumor suppressor gene by regulating EGFR in glioblastomas.. Multidimensional analysis of gene, coding sequence, promoter methylation, messenger RNA (mRNA) transcript, protein data for ANXA7 (and EGFR), and clinical patient data profiles of 543 high-grade gliomas from US medical centers and The Cancer Genome Atlas pilot project (made public 2006-2008; and unpublished, tumors collected 2001-2008). Functional analyses using LN229 and U87 glioblastoma cells.. Associations among ANXA7 gene dosage, coding sequence, promoter methylation, mRNA transcript, and protein expression. Effect of ANXA7 haploinsufficiency on EGFR signaling and patient survival. Joint effects of loss of ANXA7 and gain of EGFR expression on tumorigenesis.. Heterozygous ANXA7 gene deletion is associated with significant loss of ANXA7 mRNA transcript expression (P = 1 x 10(-15); linear regression) and a reduction (mean [SEM]) of 91.5% (2.3%) of ANXA7 protein expression compared with ANXA7 wild-type glioblastomas (P = .004; unpaired t test). ANXA7 loss of function stabilizes the EGFR protein (72%-744% increase in EGFR protein abundance) and augments EGFR transforming signaling in glioblastoma cells. ANXA7 haploinsufficiency doubles tumorigenic potential of glioblastoma cells, and combined ANXA7 knockdown and EGFR overexpression promotes tumorigenicity synergistically. The heterozygous loss of ANXA7 in approximately 75% of glioblastomas in the The Cancer Genome Atlas plus infrequency of ANXA7 mutation (approximately 6% of tumors) indicates its role as a haploinsufficiency gene. ANXA7 mRNA transcript expression, dichotomized at the median, associates with patient survival in 191 glioblastomas (log-rank P = .008; hazard ratio [HR], 0.667; 95% confidence interval [CI], 0.493-0.902; 46.9 vs 74.8 deaths/100 person-years for high vs low ANXA7 mRNA expression) and with a separate group of 180 high-grade gliomas (log-rank P = .00003; HR, 0.476; 95% CI, 0.333-0.680; 21.8 vs 50.0 deaths/100 person-years for high vs low ANXA7 mRNA expression). Deletion of the ANXA7 gene associates with poor patient survival in 189 glioblastomas (log-rank P = .042; HR, 0.686; 95% CI, 0.476-0.989; 54.0 vs 80.1 deaths/100 person-years for wild-type ANXA7 vs ANXA7 deletion).. Haploinsufficiency of the tumor suppressor ANXA7 due to monosomy of chromosome 10 provides a clinically relevant mechanism to augment EGFR signaling in glioblastomas beyond that resulting from amplification of the EGFR gene.

    Topics: Annexin A7; Brain Neoplasms; Cell Line, Tumor; Cell Transformation, Neoplastic; Chromosomes, Human, Pair 10; Chromosomes, Human, Pair 7; Epidermal Growth Factor; Epigenesis, Genetic; ErbB Receptors; Female; Gene Deletion; Gene Dosage; Gene Expression; Gene Expression Regulation, Neoplastic; Gene Knockdown Techniques; Genes, Tumor Suppressor; Glioblastoma; Humans; Loss of Heterozygosity; Male; Middle Aged; Monosomy; Mutation; PTEN Phosphohydrolase; RNA, Messenger; Signal Transduction; Survival Analysis

2009
Caspase-8 as a potential mediator of pro-tumorigenic signals.
    Cell cycle (Georgetown, Tex.), 2009, Nov-01, Volume: 8, Issue:21

    Topics: Animals; Apoptosis; Caspase 8; Cell Adhesion; Cell Movement; Cell Transformation, Neoplastic; Epidermal Growth Factor; Extracellular Signal-Regulated MAP Kinases; Humans; Signal Transduction

2009
Essential roles of PI(3)K-p110beta in cell growth, metabolism and tumorigenesis.
    Nature, 2008, Aug-07, Volume: 454, Issue:7205

    On activation by receptors, the ubiquitously expressed class IA isoforms (p110alpha and p110beta) of phosphatidylinositol-3-OH kinase (PI(3)K) generate lipid second messengers, which initiate multiple signal transduction cascades. Recent studies have demonstrated specific functions for p110alpha in growth factor and insulin signalling. To probe for distinct functions of p110beta, we constructed conditional knockout mice. Here we show that ablation of p110beta in the livers of the resulting mice leads to impaired insulin sensitivity and glucose homeostasis, while having little effect on phosphorylation of Akt, suggesting the involvement of a kinase-independent role of p110beta in insulin metabolic action. Using established mouse embryonic fibroblasts, we found that removal of p110beta also had little effect on Akt phosphorylation in response to stimulation by insulin and epidermal growth factor, but resulted in retarded cell proliferation. Reconstitution of p110beta-null cells with a wild-type or kinase-dead allele of p110beta demonstrated that p110beta possesses kinase-independent functions in regulating cell proliferation and trafficking. However, the kinase activity of p110beta was required for G-protein-coupled receptor signalling triggered by lysophosphatidic acid and had a function in oncogenic transformation. Most strikingly, in an animal model of prostate tumour formation induced by Pten loss, ablation of p110beta (also known as Pik3cb), but not that of p110alpha (also known as Pik3ca), impeded tumorigenesis with a concomitant diminution of Akt phosphorylation. Taken together, our findings demonstrate both kinase-dependent and kinase-independent functions for p110beta, and strongly indicate the kinase-dependent functions of p110beta as a promising target in cancer therapy.

    Topics: Animals; Cell Proliferation; Cell Transformation, Neoplastic; Epidermal Growth Factor; Fibroblasts; Glucose; Glucose Intolerance; Homeostasis; Humans; Insulin; Insulin Resistance; Liver; Male; Mice; Mice, Inbred C57BL; Phosphatidylinositol 3-Kinases; Phosphorylation; Prostatic Neoplasms; Proto-Oncogene Proteins c-akt; PTEN Phosphohydrolase; Signal Transduction

2008
Receptor activator of NF-kappaB Ligand (RANKL) expression is associated with epithelial to mesenchymal transition in human prostate cancer cells.
    Cell research, 2008, Volume: 18, Issue:8

    Epithelial-mesenchymal transition (EMT) in cancer describes the phenotypic and behavioral changes of cancer cells from indolent to virulent forms with increased migratory, invasive and metastatic potential. EMT can be induced by soluble proteins like transforming growth factor beta1 (TGFbeta1) and transcription factors including Snail and Slug. We utilized the ARCaP(E)/ARCaP(M) prostate cancer progression model and LNCaP clones stably overexpressing Snail to identify novel markers associated with EMT. Compared to ARCaP(E) cells, the highly tumorigenic mesenchymal ARCaP(M) and ARCaP(M1) variant cells displayed a higher incidence of bone metastasis after intracardiac administration in SCID mice. ARCaP(M) and ARCaP(M1) expressed mesenchymal stromal markers of vimentin and N-cadherin in addition to elevated levels of Receptor Activator of NF-kappaB Ligand (RANKL). We observed that both epidermal growth factor (EGF) plus TGFbeta1 treatment and Snail overexpression induced EMT in ARCaP(E) and LNCaP cells, and EMT was associated with increased expression of RANKL protein. Finally, we determined that the RANKL protein was functionally active, promoting osteoclastogenesis in vitro. Our results indicate that RANKL is a novel marker for EMT during prostate cancer progression. RANKL may function as a link between EMT, bone turnover, and prostate cancer skeletal metastasis.

    Topics: Animals; Biomarkers, Tumor; Bone Remodeling; Cadherins; Carcinoma; Cell Dedifferentiation; Cell Differentiation; Cell Line, Tumor; Cell Transformation, Neoplastic; Epidermal Growth Factor; Epithelial Cells; Humans; Male; Mesoderm; Mice; Mice, SCID; Neoplasm Metastasis; Neoplasm Transplantation; Osteoclasts; Prostatic Neoplasms; RANK Ligand; Snail Family Transcription Factors; Transcription Factors; Transforming Growth Factor beta1; Vimentin

2008
Discovery of drug-resistant and drug-sensitizing mutations in the oncogenic PI3K isoform p110 alpha.
    Cancer cell, 2008, Aug-12, Volume: 14, Issue:2

    p110 alpha (PIK3CA) is the most frequently mutated kinase in human cancer, and numerous drugs targeting this kinase are currently in preclinical development or early-stage clinical trials. Clinical resistance to protein kinase inhibitors frequently results from point mutations that block drug binding; similar mutations in p110 alpha are likely, but currently none have been reported. Using a S. cerevisiae screen against a structurally diverse panel of PI3K inhibitors, we have identified a potential hotspot for resistance mutations (I800), a drug-sensitizing mutation (L814C), and a surprising lack of resistance mutations at the "gatekeeper" residue. Our analysis further reveals that clinical resistance to these drugs may be attenuated by using multitargeted inhibitors that simultaneously inhibit additional PI3K pathway members.

    Topics: Animals; Cell Line, Tumor; Cell Transformation, Neoplastic; Class I Phosphatidylinositol 3-Kinases; Drug Resistance, Neoplasm; Epidermal Growth Factor; Humans; Isoenzymes; Mice; Mutagenesis; Mutant Proteins; Mutation; Oncogene Proteins; Phosphatidylinositol 3-Kinases; Phosphorylation; Protein Kinase Inhibitors; Proto-Oncogene Proteins c-akt; Saccharomyces cerevisiae

2008
The histone H2B-specific ubiquitin ligase RNF20/hBRE1 acts as a putative tumor suppressor through selective regulation of gene expression.
    Genes & development, 2008, Oct-01, Volume: 22, Issue:19

    Histone monoubiquitylation is implicated in critical regulatory processes. We explored the roles of histone H2B ubiquitylation in human cells by reducing the expression of hBRE1/RNF20, the major H2B-specific E3 ubiquitin ligase. While H2B ubiquitylation is broadly associated with transcribed genes, only a subset of genes was transcriptionally affected by RNF20 depletion and abrogation of H2B ubiquitylation. Gene expression dependent on RNF20 includes histones H2A and H2B and the p53 tumor suppressor. In contrast, RNF20 suppresses the expression of several proto-oncogenes, which reside preferentially in closed chromatin and are modestly transcribed despite bearing marks usually associated with high transcription rates. Remarkably, RNF20 depletion augmented the transcriptional effects of epidermal growth factor (EGF), increased cell migration, and elicited transformation and tumorigenesis. Furthermore, frequent RNF20 promoter hypermethylation was observed in tumors. RNF20 may thus be a putative tumor suppressor, acting through selective regulation of a distinct subset of genes.

    Topics: Animals; Breast Neoplasms; Cell Movement; Cell Transformation, Neoplastic; Chromatin; DNA Methylation; DNA, Neoplasm; Epidermal Growth Factor; Female; Gene Expression Regulation; HeLa Cells; Histones; Humans; Mice; Mice, Nude; Promoter Regions, Genetic; RNA Polymerase II; RNA, Small Interfering; Suppression, Genetic; Tumor Suppressor Protein p53; Tumor Suppressor Proteins; Ubiquitin-Protein Ligases; Ubiquitination

2008
CRIPTO3, a presumed pseudogene, is expressed in cancer.
    Biochemical and biophysical research communications, 2008, Dec-05, Volume: 377, Issue:1

    Cripto is a cell surface protein highly expressed in certain solid tumors, and overexpression of Cripto protein is oncogenic. Cripto-1 protein is encoded by CRIPTO1 gene. CRIPTO3, a presumed pseudogene, has an open reading frame with six amino acid differences from Cripto-1. We show that CRIPTO3 mRNA is the CRIPTO message expressed in many cancer samples. A CRIPTO3 SAGE tag was found in several cancer SAGE libraries, while the CRIPTO1 tag was found in ES cell libraries. In vitro experiments indicate both Cripto-1 and Cripto-3 proteins are functional in the Nodal-dependent signal pathway. Our data indicate that CRIPTO3 is an expressed gene, particularly in certain cancers, and suggest a potentially novel mechanism of oncogenesis through activation of a retrogene.

    Topics: Amino Acid Sequence; Cell Line, Tumor; Cell Transformation, Neoplastic; Epidermal Growth Factor; Gene Expression Regulation, Neoplastic; Genes, Neoplasm; GPI-Linked Proteins; Humans; Intercellular Signaling Peptides and Proteins; Membrane Glycoproteins; Molecular Sequence Data; Neoplasm Proteins; Neoplasms; Pseudogenes; Transcription, Genetic

2008
Synergistic effect between EGF and TGF-beta1 in inducing oncogenic properties of intestinal epithelial cells.
    Oncogene, 2008, Apr-17, Volume: 27, Issue:18

    Transforming growth factor (TGF)-beta1 has a biphasic effect on rat intestinal epithelial (RIE) cells. By itself, TGF-beta1 functions as a tumor suppressor by inhibiting the growth, migration and invasion of RIE cells. We show in this study that in conjunction with epidermal growth factor (EGF), TGF-beta1 helped to augment migration, invasion and anchorage-independent growth (AIG) compared to that by EGF alone. EGF plus TGF-beta1 induced a dramatic morphological change characteristic of epithelial-mesenchymal transition (EMT). The mechanism for this enhanced effect of TGF-beta1 and EGF on oncogenic properties was explored by analysis of EGF- and TGF-beta1-mediated signaling pathways and complementary DNA arrays. TGF-beta1 augmented EGF-mediated signaling of mitogen-activated protein kinase (MAPK) and AKT by enhancing and prolonging the activation of the former and prolonging the activation of the latter. Inhibition of MAPK, but not phosphoinositide-3 kinase (PI3K), abolished TGF-beta1 plus EGF-induced EMT and downregulation of E-cadherin at mRNA and protein levels. By contrast, cell migration and invasion were sensitive to inhibition of either MAPK or PI3 kinase. TGF-beta1 plus EGF-induced AIG was significantly more resistant to inhibition of PI3K and MAPK compared to that induced by EGF alone. EGF and TGF-beta1 synergistically induced the expression of a series of proteases including matrix metalloproteinase (MMP) 1 (collagenase), MMP3, MMP9, MMP10, MMP14 and cathepsin. Among them, the expression of MMP1, MMP3, MMP9 and MMP10 was MAPK dependent. Inhibition of the MMPs or cathepsin significantly blocked EGF plus TGF-beta1-induced invasion, but had no effect on colony formation. Phospholipase C (PLC) and Cox2 induced by EGF plus TGF-beta1 also played a significant role in invasion, whereas PLC was also important for colony formation. Our study reveals specific signaling functions and induction of genes differentially required for enhanced effect of EGF- and TGF-beta1-induced oncogenic properties, and helps to explain the tumor-promoting effect of TGF-beta1 in human cancer with elevated expression or activation of TGF-beta1 and receptor protein tyrosine kinases.

    Topics: Animals; Cathepsins; Cell Movement; Cell Proliferation; Cell Transformation, Neoplastic; Drug Synergism; Epidermal Growth Factor; Epithelial Cells; Extracellular Signal-Regulated MAP Kinases; Gene Expression Profiling; Humans; Intestinal Mucosa; Matrix Metalloproteinase Inhibitors; Matrix Metalloproteinases; Oligonucleotide Array Sequence Analysis; Phosphatidylinositol 3-Kinases; Proto-Oncogene Proteins c-akt; Rats; Signal Transduction; Transforming Growth Factor beta1; Tumor Suppressor Proteins; Type C Phospholipases

2008
Fyn is a novel target of (-)-epigallocatechin gallate in the inhibition of JB6 Cl41 cell transformation.
    Molecular carcinogenesis, 2008, Volume: 47, Issue:3

    The cancer preventive action of (-)-epigallocatechin gallate (EGCG), found in green tea, is strongly supported by epidemiology and laboratory research data. However, the mechanism by which EGCG inhibits carcinogenesis and cell transformation is not clear. In this study, we report that EGCG suppressed epidermal growth factor (EGF)-induced cell transformation in JB6 cells. We also found that EGCG inhibited EGF-induced Fyn kinase activity and phosphorylation in vitro and in vivo. Fyn was implicated in the process because EGF-induced JB6 cell transformation was inhibited by small interfering RNA (siRNA)-Fyn-JB6 cells. With an in vitro protein-binding assay, we found that EGCG directly bound with the GST-Fyn-SH2 domain but not the GST-Fyn-SH3 domain. The K(d) value for EGCG binding to the Fyn SH2 domain was 0.367 +/- 0.122 microM and B(max) was 1.35 +/- 0.128 nmol/mg. Compared with control JB6 Cl41 cells, EGF-induced phosphorylation of p38 MAP kinase (p38 MAPK) (Thr180/Tyr182), ATF-2 (Thr71) and signal transducer and activator of transcription 1 (STAT1) (Thr727) was decreased in siRNA-Fyn-JB6 cells. EGCG could inhibit the phosphorylation of p38 MAPK, ATF-2, and STAT1. The DNA binding ability of AP-1, STAT1, and ATF-2 was also decreased in siRNA-Fyn-JB6 cells. Overall, these results demonstrated that EGCG interacted with Fyn and inhibited Fyn kinase activity and thereby regulated EGF-induced cell transformation. Inhibition of Fyn kinase activity is a novel and important mechanism that may be involved in EGCG-induced inhibition of cell transformation.

    Topics: Activating Transcription Factor 2; Adaptor Proteins, Signal Transducing; Animals; Antioxidants; Catechin; Cell Survival; Cell Transformation, Neoplastic; CREB-Binding Protein; Dose-Response Relationship, Drug; Epidermal Growth Factor; Glutathione Transferase; Kinetics; Mice; p38 Mitogen-Activated Protein Kinases; Phosphorylation; Protein Binding; Protein Structure, Tertiary; Recombinant Fusion Proteins; RNA, Small Interfering; STAT1 Transcription Factor; Time Factors; Transcription Factor AP-1

2008
Inhibitory effects of caffeine analogues on neoplastic transformation: structure-activity relationship.
    Carcinogenesis, 2008, Volume: 29, Issue:6

    Some xanthine analogues, including 1,3,7-trimethylxanthine (caffeine) and 1,3-dimethylxanthine (theophylline), have been shown to exert anticancer activities in both cell culture and animal models. The present study focused on the relationship of structure and activity of 50 different caffeine analogues in preventing epidermal growth factor (EGF)-induced malignant transformation of mouse epidermal JB6 promotion-sensitive (P+) Cl41 (JB6 P+) cells. Results indicated that the inhibition of cell transformation by the 1,3,7-trialkylxanthines depends on the number of carbons at the alkyl groups R1 and R3, but not R7. Notably, 1-ethyl-3-hexylxanthine (xanthine 70) was the most effective compound for inhibiting EGF-induced neoplastic transformation among the 50 xanthine analogues tested. The 50% inhibition of cell transformation (ICT(50)) value for xanthine 70 was 48- or 75-fold less than the ICT(50) value of caffeine or theophylline, respectively. Further study revealed that xanthine 70 (5-40 muM) dose dependently inhibited EGF-induced transactivation of activator protein 1 (AP-1), whereas theophylline or caffeine (up to 500 muM) had no effect on AP-1 activity. In addition, xanthine 70 (10 muM) inhibited 12-O-tetradecanoylphorbol-13-acetate- or H-Ras-induced neoplastic transformation in JB6 P+ cells by 78.2 or 62.0%, respectively. Collectively, these results indicated that the number of carbons at R1 and R3 is important for the antitumor-promoting activity of the trialkylxanthines and xanthine 70 might be a promising anticancer agent.

    Topics: Animals; Antineoplastic Agents; Caffeine; Cell Line; Cell Transformation, Neoplastic; Enzyme Activation; Epidermal Growth Factor; Mice; Structure-Activity Relationship; Transcription Factor AP-1

2008
Fucoidan from Laminaria cichorioides inhibits AP-1 transactivation and cell transformation in the mouse epidermal JB6 cells.
    Molecular carcinogenesis, 2008, Volume: 47, Issue:8

    Fucoidan, a sulfated polysaccharide extracted from brown seaweeds, has anticoagulant and antithrombotic activities. Unlike heparine, fucoidan is known to exhibit anticarcinogenic activities. However, the underlying molecular mechanisms of the chemopreventive activities of fucoidan are not understood. Here we report that fucoidan from Laminaria cichorioides inhibited the epidermal growth factor (EGF) or 12-O-tetradecanoylphorbol-13-acetate (TPA)-induced neoplastic cell transformation, but had less cytotoxic effects on JB6 mouse epidermal cells. The EGF-induced phosphorylation of extracellular signal-regulated kinases 1/2 and c-Jun N-terminal kinases, and c-Jun was inhibited by fucoidan, resulting from the inhibition of phosphorylation of epidermal growth factor receptor (EGFR). Fucoidan dose-dependently attenuated the c-fos or c-jun transcriptional activity, and thereby inhibited the associated activator protein-1 (AP-1) transactivation activity. In vitro binding assay revealed that fucoidan directly interacted with EGF, suggested that antitumor promoting effect of fucoidan might be due to preventing the binding of EGF to its cell surface receptor (EGFR). These findings are the first to reveal a molecular basis for the anticarcinogenic action of fucoidan and may partially account for the reported chemopreventive effects of brown seaweeds.

    Topics: Animals; Cell Adhesion; Cell Line; Cell Transformation, Neoplastic; Epidermal Cells; Epidermal Growth Factor; Laminaria; MAP Kinase Signaling System; Mice; Phosphorylation; Polysaccharides; Tetradecanoylphorbol Acetate; Transcription Factor AP-1; Transcriptional Activation

2008
Inhibitory effects of fucoidan on activation of epidermal growth factor receptor and cell transformation in JB6 Cl41 cells.
    Food and chemical toxicology : an international journal published for the British Industrial Biological Research Association, 2008, Volume: 46, Issue:5

    Algal fucoidan is a marine sulfated polysaccharide with a wide variety of biological activities including anti-thrombotic, anti-inflammatory, and anti-tumor activities. In this study, we tested the hypothesis that fucoidan may suppress neoplastic cell transformation by inhibiting the phosphorylation of epidermal growth factor receptor (EGFR) in mouse epidermal JB6 Cl41 cells. Our results provided the first evidence that fucoidan from Laminaria guryanovae exerted a potent inhibitory effect on EGF-induced phosphorylation of EGFR. Consistent with its inhibitory action on phosphorylation of EGFR, fucoidan clearly suppressed the phosphorylation of extracellular signal-regulated kinase or c-jun N-terminal kinases induced by EGF. Moreover, EGF-induced the c-fos and c-jun transcriptional activities were inhibited by fucoidan, resulting to suppressing of activator protein-1 (AP-1) activity and cell transformation induced by EGF. Taken together, these results indicate that fucoidan might exert chemopreventive effects through the inhibition of phosphorylation of the EGFR.

    Topics: Animals; Antineoplastic Agents; Biotransformation; Blotting, Western; Cell Line, Tumor; Cell Survival; Cell Transformation, Neoplastic; Chromatography, Ion Exchange; Epidermal Growth Factor; ErbB Receptors; Genes, fos; Genes, jun; Genes, Reporter; Indicators and Reagents; Laminaria; Mice; Phosphorylation; Polysaccharides; Signal Transduction; Tetrazolium Salts; Thiazoles; Transcription Factor AP-1

2008
Mitogen- and stress-activated kinase 1-mediated histone H3 phosphorylation is crucial for cell transformation.
    Cancer research, 2008, Apr-01, Volume: 68, Issue:7

    Mitogen- and stress-activated kinase 1 (MSK1) belongs to a family of dual protein kinases that are activated by either extracellular signal-regulated kinase or p38 mitogen-activated protein kinases in response to stress or mitogenic extracellular stimuli. The physiologic role of MSK1 in malignant transformation and cancer development is not well understood. Here, we report that MSK1 is involved in 12-O-tetradecanoylphorbol-13-acetate (TPA)-induced or epidermal growth factor (EGF)-induced neoplastic transformation of JB6 Cl41 cells. H89, a potent inhibitor of MSK1, strongly suppressed TPA-induced or EGF-induced cell transformation. When cells overexpressing wild-type MSK1 were treated with TPA or EGF, colony formation increased substantially compared with untreated cells or cells that did not overexpress MSK1. In contrast, MSK1 COOH terminal or NH(2) terminal dead dominant negative mutants dramatically suppressed cell transformation. Introduction of small interfering RNA-MSK1 into JB6 Cl41 cells resulted in suppressed TPA-induced or EGF-induced cell transformation. In addition, cell proliferation was inhibited in MSK1 knockdown cells compared with MSK1 wild-type cells. In wild-type MSK1-overexpressing cells, activator protein (AP-1) activation increased after TPA or EGF stimulation, whereas AP-1 activation decreased in both MSK1 dominant-negative mutants and in MSK1 knockdown cells. Moreover, TPA-induced or EGF-induced phosphorylation of histone H3 at Ser(10) was increased in wild-type cells but the induced phosphorylation was abolished in MSK1 dominant-negative mutant or MSK1 knockdown cells. Thus, MSK1 is required for tumor promoter-induced cell transformation through its phosphorylation of histone H3 at Ser(10) and AP-1 activation.

    Topics: Animals; Cell Growth Processes; Cell Transformation, Neoplastic; Enzyme Activation; Epidermal Growth Factor; G1 Phase; Histones; Isoquinolines; Mice; Phosphorylation; Protein Kinase Inhibitors; Ribosomal Protein S6 Kinases, 90-kDa; RNA, Small Interfering; Skin; Sulfonamides; Tetradecanoylphorbol Acetate; Transcription Factor AP-1

2008
Transformation of human urothelial cells (UROtsa) by as and cd induces the expression of keratin 6a.
    Environmental health perspectives, 2008, Volume: 116, Issue:4

    Cadmium and arsenite can directly and malignantly transform the UROtsa cell line. The tumor heterotransplants produced from these transformed cells have histologic features consistent with human bladder cancer. Previous microarray analysis of total RNA from the parental and transformed cells suggested that keratin 6a was overexpressed as a result of cell transformation.. Our goals were to verify overexpression of keratin 6a in Cd(2+)- and As(3+)-transformed UROtsa cells, the corresponding tumor heterotransplants, and human bladder cancer biopsy specimens and to assess what factors may be involved in keratin 6a overexpression.. Expression was assessed with real-time polymerase chain reaction, Western blot analysis, and immunohistochemistry. We used the effect of addition and deletion of potential growth factors in the cell culture growth medium to assess possible pathways used in keratin 6a overexpression.. Cd(2+)- and As(3+)-transformed cells grown in serum-containing growth medium, as well as the derived tumor heterotransplants, overexpressed keratin 6a mRNA and protein compared with UROtsa cells grown in serum-containing growth medium. Immunostaining of keratin 6a in tumor heterotransplants showed focal staining of the tumor cells that was localized to the cytoplasm. Focal immunostaining of keratin 6a was also found in some but not all archival patient specimens of high-grade bladder cancer, confirming translation of the results to human bladder cancer. Studies on growth factor deletion and addition indicated that the level of keratin 6a expression was regulated by the presence of both insulin and epidermal growth factor (EGF). In contrast, growth factors had no effect on the elevated levels of keratin 6a expression found in transformed UROtsa cells.. Our present studies suggest that keratin 6a expression may be a biomarker for malignant urothelial cells that possess an activated EGF and or insulin growth factor pathway.

    Topics: Animals; Arsenites; Biomarkers, Tumor; Blotting, Western; Cadmium; Cell Line; Cell Transformation, Neoplastic; Culture Media; Epidermal Growth Factor; Humans; Immunohistochemistry; Insulin; Keratin-6; Mice; Mice, Nude; Neoplasm Transplantation; Polymerase Chain Reaction; RNA, Messenger; Transplantation, Heterologous; Urinary Bladder Neoplasms; Urothelium

2008
Structure-activity relationship of flavonoids for inhibition of epidermal growth factor-induced transformation of JB6 Cl 41 cells.
    Molecular carcinogenesis, 2007, Volume: 46, Issue:6

    We found that quercetin, myricetin, quercetagetin, fisetin, (-)-epigallocatechin gallate (EGCG), and theaflavins, among 24 flavonoids examined, markedly inhibited epidermal growth factor (EGF)-induced cell transformation of mouse epidermal JB6 Cl 41 cells. The six flavonoids suppressed the EGF-induced activation of activator protein 1 (AP-1). In addition, myricetin, quercetagetin, EGCG, and theaflavins directly inhibited EGF-induced phosphatidylinositol 3-kinase (PI3K) activation. The important structural features of flavonoids for cell transformation-inhibitory activity are 3'- and 4'-OH on the B-ring, 3-OH on the C-ring, C2=C3 double bond in the C-ring, and the phenylchromone (C6-C5-C6) skeleton in the flavonols, and the galloyl group in EGCG and theaflavins. Our results provide new insight into possible mechanisms of the anti-carcinogenic effects of flavonoids, and could help to provide a basis for the design of novel cancer chemopreventive agents.

    Topics: Animals; Apoptosis; Cell Adhesion; Cell Line, Tumor; Cell Transformation, Neoplastic; Dose-Response Relationship, Drug; Drug Screening Assays, Antitumor; Epidermal Growth Factor; Flavonoids; Mice; Models, Chemical; Protein Binding; Signal Transduction; Structure-Activity Relationship; Transcription Factor AP-1

2007
A multiscale computational approach to dissect early events in the Erb family receptor mediated activation, differential signaling, and relevance to oncogenic transformations.
    Annals of biomedical engineering, 2007, Volume: 35, Issue:6

    We describe a hierarchical multiscale computational approach based on molecular dynamics simulations, free energy-based molecular docking simulations, deterministic network-based kinetic modeling, and hybrid discrete/continuum stochastic dynamics protocols to study the dimer-mediated receptor activation characteristics of the Erb family receptors, specifically the epidermal growth factor receptor (EGFR). Through these modeling approaches, we are able to extend the prior modeling of EGF-mediated signal transduction by considering specific EGFR tyrosine kinase (EGFRTK) docking interactions mediated by differential binding and phosphorylation of different C-terminal peptide tyrosines on the RTK tail. By modeling signal flows through branching pathways of the EGFRTK resolved on a molecular basis, we are able to transcribe the effects of molecular alterations in the receptor (e.g., mutant forms of the receptor) to differing kinetic behavior and downstream signaling response. Our molecular dynamics simulations show that the drug sensitizing mutation (L834R) of EGFR stabilizes the active conformation to make the system constitutively active. Docking simulations show preferential characteristics (for wildtype vs. mutant receptors) in inhibitor binding as well as preferential enhancement of phosphorylation of particular substrate tyrosines over others. We find that in comparison to the wildtype system, the L834R mutant RTK preferentially binds the inhibitor erlotinib, as well as preferentially phosphorylates the substrate tyrosine Y1068 but not Y1173. We predict that these molecular level changes result in preferential activation of the Akt signaling pathway in comparison to the Erk signaling pathway for cells with normal EGFR expression. For cells with EGFR over expression, the mutant over activates both Erk and Akt pathways, in comparison to wildtype. These results are consistent with qualitative experimental measurements reported in the literature. We discuss these consequences in light of how the network topology and signaling characteristics of altered (mutant) cell lines are shaped differently in relationship to native cell lines.

    Topics: Animals; Cell Transformation, Neoplastic; Computer Simulation; Epidermal Growth Factor; ErbB Receptors; Gene Expression Regulation, Neoplastic; Humans; Models, Biological; Protein Interaction Mapping; Signal Transduction

2007
Dactylone inhibits epidermal growth factor-induced transformation and phenotype expression of human cancer cells and induces G1-S arrest and apoptosis.
    Cancer research, 2007, Jun-15, Volume: 67, Issue:12

    The marine natural chamigrane-type sesquiterpenoid, dactylone, is closely related to secondary metabolites of some edible species of red algae. In the present study, the effect of dactylone was tested on the mouse skin epidermal JB6 P+ Cl41 cell line and its stable transfectants as well as on several human tumor cell lines, including lung (H460), colon (HCT-116), and skin melanomas (SK-MEL-5 and SK-MEL-28). This natural product was effective at nontoxic doses as a cancer-preventive agent, which exerted its actions, at least in part, through the inhibition of cyclin D3 and Cdk4 expression and retinoblastoma tumor suppressor protein (Rb) phosphorylation. The inhibition of these cell cycle components was followed by cell cycle arrest at the G1-S transition with subsequent p53-independent apoptosis. Therefore, these data showed that application of dactylone and related compounds may lead to decreased malignant cell transformation and/or decreased tumor cell proliferation.

    Topics: Animals; Antineoplastic Agents; Apoptosis; Blotting, Western; Cell Cycle; Cell Line, Tumor; Cell Transformation, Neoplastic; Epidermal Growth Factor; Flow Cytometry; G1 Phase; Humans; Mice; Neoplasms; Phenotype; S Phase; Sesquiterpenes; Transfection

2007
PRR5, a novel component of mTOR complex 2, regulates platelet-derived growth factor receptor beta expression and signaling.
    The Journal of biological chemistry, 2007, Aug-31, Volume: 282, Issue:35

    The protein kinase mammalian target of rapamycin (mTOR) plays an important role in the coordinate regulation of cellular responses to nutritional and growth factor conditions. mTOR achieves these roles through interacting with raptor and rictor to form two distinct protein complexes, mTORC1 and mTORC2. Previous studies have been focused on mTORC1 to elucidate the central roles of the complex in mediating nutritional and growth factor signals to the protein synthesis machinery. Functions of mTORC2, relative to mTORC1, have remained little understood. Here we report identification of a novel component of mTORC2 named PRR5 (PRoline-Rich protein 5), a protein encoded by a gene located on a chromosomal region frequently deleted during breast and colorectal carcinogenesis (Johnstone, C. N., Castellvi-Bel, S., Chang, L. M., Sung, R. K., Bowser, M. J., Pique, J. M., Castells, A., and Rustgi, A. K. (2005) Genomics 85, 338-351). PRR5 interacts with rictor, but not raptor, and the interaction is independent of mTOR and not disturbed under conditions that disrupt the mTOR-rictor interaction. PRR5, unlike Sin1, another component of mTORC2, is not important for the mTOR-rictor interaction and mTOR activity toward Akt phosphorylation. Despite no significant effect of PRR5 on mTORC2-mediated Akt phosphorylation, PRR5 silencing inhibits Akt and S6K1 phosphorylation and reduces cell proliferation rates, a result consistent with PRR5 roles in cell growth and tumorigenesis. The inhibition of Akt and S6K1 phosphorylation by PRR5 knock down correlates with reduction in the expression level of platelet-derived growth factor receptor beta (PDGFRbeta). PRR5 silencing impairs PDGF-stimulated phosphorylation of S6K1 and Akt but moderately reduces epidermal growth factor- and insulin-stimulated phosphorylation. These findings propose a potential role of mTORC2 in the cross-talk with the cellular machinery that regulates PDGFRbeta expression and signaling.

    Topics: Adaptor Proteins, Signal Transducing; Breast Neoplasms; Carrier Proteins; Cell Proliferation; Cell Transformation, Neoplastic; Colorectal Neoplasms; Epidermal Growth Factor; Gene Expression Regulation; Gene Silencing; HeLa Cells; Humans; Hypoglycemic Agents; Insulin; Intracellular Signaling Peptides and Proteins; Mechanistic Target of Rapamycin Complex 1; Multiprotein Complexes; Phosphorylation; Protein Kinases; Proteins; Proto-Oncogene Proteins c-akt; Rapamycin-Insensitive Companion of mTOR Protein; Receptor, Platelet-Derived Growth Factor beta; Regulatory-Associated Protein of mTOR; Ribosomal Protein S6 Kinases; Sequence Deletion; Signal Transduction; TOR Serine-Threonine Kinases; Transcription Factors

2007
Long-term EGF/serum-treated human thyrocytes mimic papillary thyroid carcinomas with regard to gene expression.
    Experimental cell research, 2007, Sep-10, Volume: 313, Issue:15

    Constitutive activation of the RAS/RAF/MAPK pathway has been found in different tumor types including papillary thyroid carcinomas (PTCs). To get more insight into genes primarily regulated in the human tumor cells, an in vitro model was developed in which primary cultures of human thyrocytes were treated for different times with epidermal growth factor and serum (EGF/serum), which stimulate the MAPK cascade. Gene expression profiles were obtained by microarrays and compared to the expression profiles of PTCs. An evolution from short-term to long-term EGF/serum-treated cells was found, i.e., a program change showing a distinction between gene expression profiles of short-term and long-term EGF/serum-treated cells. The late pattern of EGF/serum stimulated cells converges to the pattern of PTCs. Comparison of these two types of cells with cAMP activated cells, from thyroid-stimulating hormone-treated thyrocytes and autonomous adenomas, showed distinct gene expression profiles for the two pathways. For the two models, an overlap was found in a number of genes which were early induced in vitro but down-regulated later in vitro and in the in vivo tumors. Thus, long-term stimulated human primary cultures demonstrate a clear relation with the tumor in vivo and could therefore be used as models for the disease.

    Topics: Adult; Carcinoma, Papillary; Cell Transformation, Neoplastic; Cells, Cultured; Cyclic AMP; Epidermal Growth Factor; Humans; Middle Aged; Oligonucleotide Array Sequence Analysis; Serum; Thyroid Gland; Thyroid Neoplasms

2007
Myricetin is a novel natural inhibitor of neoplastic cell transformation and MEK1.
    Carcinogenesis, 2007, Volume: 28, Issue:9

    Evidence suggests that mitogen-activated protein kinase kinase (MEK) plays a role in cell transformation and tumor development and might be a significant target for chemoprevention. 3,5,4'-Trihydroxy-trans-stilbene (resveratrol), a non-flavonoid polyphenol found in various foods and beverages, including red wines, is reported to be a natural chemopreventive agent. However, the concentrations required to exert these effects might be difficult to achieve by drinking only one or two glasses of red wine a day. On the other hand, the flavonol content of red wine is approximately 30 times higher than that of resveratrol. Here we demonstrated that 3,3',4',5,5',7-hexahydroxyflavone (myricetin), one of the major flavonols in red wine, is a novel inhibitor of MEK1 activity and transformation of JB6 P+ mouse epidermal cells. Myricetin (10 microM) inhibited 12-O-tetradecanoylphorbol-13-acetate (TPA) or epidermal growth factor (EGF)-induced cell transformation by 76 or 72%, respectively, compared with respective reductions of 26 or 19% by resveratrol (20 microM). A combination of myricetin and resveratrol exerted additive but not synergistic effects on either TPA- or EGF-induced transformation. Myricetin, but not resveratrol, attenuated tumor promoter-induced activation of c-fos or activator protein-1. Myricetin strongly inhibited MEK1 kinase activity and suppressed TPA- or EGF-induced phosphorylation of extracellular signal-regulated kinase (ERK) or p90 ribosomal S6 kinase, downstream targets of MEK. Moreover, myricetin inhibited H-Ras-induced cell transformation more effectively than either PD098059, a MEK inhibitor, or resveratrol. Myricetin directly bound with glutathione S-transferase-MEK1 but did not compete with ATP. Overall, these results indicated that myricetin has potent anticancer-promoting activity and mainly targets MEK signaling, which may contribute to the chemopreventive potential of several foods including red wines.

    Topics: Animals; Antineoplastic Agents; Cell Division; Cell Transformation, Neoplastic; Cells, Cultured; Enzyme Inhibitors; Epidermal Growth Factor; Fireflies; Flavonoids; Genes, Reporter; Luciferases; MAP Kinase Kinase 1; Mice; Recombinant Proteins; Tetradecanoylphorbol Acetate

2007
The role of glycogen synthase kinase 3beta in the transformation of epidermal cells.
    Cancer research, 2007, Aug-15, Volume: 67, Issue:16

    Glycogen synthase kinase 3beta (GSK3beta) is a multifunctional serine/threonine kinase. We showed that the expression of GSK3beta was drastically down-regulated in human cutaneous squamous cell carcinomas and basal cell carcinomas. Due to its negative regulation of many oncogenic proteins, we hypothesized that GSK3beta may function as a tumor suppressor during the neoplastic transformation of epidermal cells. We tested this hypothesis using an in vitro model system, JB6 mouse epidermal cells. In response to epidermal growth factor (EGF) or 12-O-tetradecanoylphorbol-13-acetate (TPA), the promotion-sensitive JB6 P+ cells initiate neoplastic transformation, whereas the promotion-resistant JB6 P- cells do not. JB6 P- cells expressed much higher levels of GSK3beta than JB6 P+ cells; JB7 cells, the transformed derivatives of JB6, had the least amount of GSK3beta. The activity of GSK3beta is negatively regulated by its phosphorylation at Ser9. EGF and TPA induced strong Ser9 phoshorylation in JB6 P+ cells, but phosphorylation was seen at a much lesser extent in JB6 P- cells. EGF and TPA-stimulated Ser9 phosphorylation was mediated by phosphoinositide-3-kinase (PI3K)/Akt and protein kinase C (PKC) pathways. Inhibition of GSK3beta activation significantly stimulated activator protein-1 (AP-1) activity. Overexpression of wild-type (WT) and S9A mutant GSK3beta in JB6 P+ cells suppressed EGF and TPA-mediated anchorage-independent growth in soft agar and tumorigenicity in nude mice. Overexpression of a kinase-deficient (K85R) GSK3beta, in contrast, potentiated anchorage-independent growth and drastically enhanced in vivo tumorigenicity. Together, these results indicate that GSK3beta plays an important role in skin tumorigenesis.

    Topics: Animals; Carcinoma, Basal Cell; Carcinoma, Squamous Cell; Cell Transformation, Neoplastic; Enzyme Activation; Epidermal Growth Factor; Glycogen Synthase Kinase 3; Glycogen Synthase Kinase 3 beta; Humans; Male; Mice; Mice, Inbred BALB C; Oncogene Protein v-akt; Phosphatidylinositol 3-Kinases; Protein Kinase C; Signal Transduction; Skin; Skin Neoplasms; Tetradecanoylphorbol Acetate; Transcription Factor AP-1; Transfection

2007
Ribosomal S6 kinase 2 is a key regulator in tumor promoter induced cell transformation.
    Cancer research, 2007, Sep-01, Volume: 67, Issue:17

    The ribosomal S6 kinase 2 (RSK2), a member of the p90(RSK) (RSK) family of proteins, is a widely expressed serine/threonine kinase that is activated by extracellular signal-regulated kinase 1/2 and phosphoinositide-dependent kinase 1 in response to many growth factors and peptide hormones. Its activation signaling enhances cell survival. However, the roles of RSK2 in cell transformation have not yet been elucidated. Here, we found that RSK2 is a critical serine/threonine kinase for the regulation of cell transformation. When cells were stimulated with tumor promoters, such as epidermal growth factor (EGF) or 12-O-tetradecanoylphorbol-13-acetate (TPA), phosphorylation of RSK was increased within 5 min. Cell proliferation was suppressed in RSK2(-/-) mouse embryonic fibroblasts (MEFs) compared with RSK2(+/+) MEFs. Moreover, RSK2(-/-) MEFs accumulated at the G(1) phase of the cell cycle under normal cell culture conditions as well as after stimulation with EGF or TPA. In the anchorage-independent cell transformation assay (soft agar), stable expression of RSK2 in JB6 cells significantly enhanced colony formation in either the presence or absence of tumor promoters. Furthermore, knockdown of RSK2 with small interfering RNA-RSK2 suppressed constitutively active Ras (Ras(G12V))-induced foci formation in NIH3T3 cells. In addition, kaempferol, an inhibitor of RSK2, suppressed EGF-induced colony formation of JB6 Cl41 cells in soft agar, which was associated with inhibition of histone H3 phosphorylation (Ser(10)). These results showed that RSK2 is a key regulator for cell transformation induced by tumor promoters such as EGF and TPA.

    Topics: Animals; Base Sequence; Carcinogens; Cell Adhesion; Cell Proliferation; Cell Transformation, Neoplastic; Cells, Cultured; Epidermal Growth Factor; G1 Phase; Mice; Molecular Sequence Data; NIH 3T3 Cells; Ribosomal Protein S6 Kinases, 90-kDa; Sequence Homology, Nucleic Acid; Tetradecanoylphorbol Acetate; Transfection

2007
Effects of MAP kinase inhibitors on epidermal growth factor-induced neoplastic transformation of human keratinocytes.
    Molecular carcinogenesis, 2006, Volume: 45, Issue:1

    We previously reported data regarding the mechanism of neoplastic transformation in JB6 Cl41 mouse skin epidermal cells. However, experimental in vitro models for studying neoplastic transformation of human cells could provide further insight into the mechanisms of human cancer development. In this study, we have established a neoplastic transformation model with HaCaT cells, a human keratinocyte cell line, and showed the usefulness of this cell line for studying the mechanisms of neoplastic transformation. Epidermal growth factor (EGF) treatment induced a dose-dependent anchorage-independent cell transformation in HaCaT cells. Furthermore, PD98059, a mitogen-activated protein (MAP) kinase/ERK kinase (MEK) inhibitor, or SP600125, c-Jun N-terminal kinase (JNK) inhibitor, decreased cell growth, EGF-induced DNA synthesis and transformation. Unlike observations in the JB6 mouse epidermal cell model, SB203580, a stress-activated protein kinase-2/p38 alpha and beta (p38) inhibitor, increased EGF-induced transformation in HaCaT cells. These results suggest that extracellular-signal regulated kinase (ERK), JNK, or p38 are implicated in EGF-induced neoplastic transformation of human cells.

    Topics: Cell Line; Cell Transformation, Neoplastic; Enzyme Inhibitors; Epidermal Growth Factor; Humans; Keratinocytes; Mitogen-Activated Protein Kinases; Signal Transduction

2006
Molecular pathways regulating EGF-induced epithelio-mesenchymal transition in human ovarian surface epithelium.
    American journal of physiology. Cell physiology, 2006, Volume: 290, Issue:6

    The ovarian surface epithelium (OSE) is the precursor of common epithelial ovarian carcinomas. In the present study, we examined the molecular mechanisms and possible physiological basis for the propensity of OSE cells to undergo epithelio-mesenchymal transition (EMT) in response to environmental influences. We hypothesized that EMT may be a homeostatic mechanism that permits displaced OSE to assume a stromal phenotype within the ovarian cortex. We report that EGF in conjunction with hydrocortisone is the EMT-inducing factor of OSE as shown by changes to a fibroblast-like morphology and growth pattern. EGF increased cell motility, enhanced the activities of secreted pro-matrix metalloproteinase (MMP)-2 and -9, and enhanced expression and activation of Erk and integrin-linked kinase (ILK). Increased ILK expression correlated with the activation of PKB/Akt, the phosphorylation of GSK-3beta, and the increased expression of cyclin E and cdk2 kinase. EGF withdrawal resulted in a more epithelial morphology and reversal of the EGF-induced activation of signaling pathways and pro-MMP activity. In contrast, treatment of EGF-treated cells with specific inhibitors of phosphatidylinositol 3-kinase, Mek, or ILK inhibited the inhibitor-specific pathways. The inhibitors caused suppression of EGF-induced migration and pro-MMP-2/-9 activities but did not lead to any change in EGF-induced mesenchymal morphology. ILK small interfering RNA inhibited Akt phosphorylation and reduced pro-MMP-2/-9 activities but had no effect on Erk activation or cell morphology. These results indicate that the EGF-induced morphological and functional changes in OSE cells are controlled by distinct signaling mechanisms working in concert. EMT of OSE cells displaced by ovulation likely permits their survival and integration with a fibroblast-like identity within the stroma. Failure to do so may lead to the formation of epithelium-derived inclusion cysts, which are known preferential sites of malignant transformation.

    Topics: Adult; Blotting, Western; Cell Movement; Cell Transformation, Neoplastic; Enzyme Activation; Epidermal Growth Factor; Epithelium; Extracellular Signal-Regulated MAP Kinases; Female; Humans; Matrix Metalloproteinases; Mesoderm; Middle Aged; Ovary; Protein Serine-Threonine Kinases; RNA, Small Interfering; Signal Transduction

2006
Multiple oncogenic changes (K-RAS(V12), p53 knockdown, mutant EGFRs, p16 bypass, telomerase) are not sufficient to confer a full malignant phenotype on human bronchial epithelial cells.
    Cancer research, 2006, Feb-15, Volume: 66, Issue:4

    We evaluated the contribution of three genetic alterations (p53 knockdown, K-RAS(V12), and mutant EGFR) to lung tumorigenesis using human bronchial epithelial cells (HBEC) immortalized with telomerase and Cdk4-mediated p16 bypass. RNA interference p53 knockdown or oncogenic K-RAS(V12) resulted in enhanced anchorage-independent growth and increased saturation density of HBECs. The combination of p53 knockdown and K-RAS(V12) further enhanced the tumorigenic phenotype with increased growth in soft agar and an invasive phenotype in three-dimensional organotypic cultures but failed to cause HBECs to form tumors in nude mice. Growth of HBECs was highly dependent on epidermal growth factor (EGF) and completely inhibited by EGF receptor (EGFR) tyrosine kinase inhibitors, which induced G1 arrest. Introduction of EGFR mutations E746-A750 del and L858R progressed HBECs toward malignancy as measured by soft agar growth, including EGF-independent growth, but failed to induce tumor formation. Mutant EGFRs were associated with higher levels of phospho-Akt, phospho-signal transducers and activators of transcription 3 [but not phospho-extracellular signal-regulated kinase (ERK) 1/2], and increased expression of DUSP6/MKP-3 phosphatase (an inhibitor of phospho-ERK1/2). These results indicate that (a) the HBEC model system is a powerful new approach to assess the contribution of individual and combinations of genetic alterations to lung cancer pathogenesis; (b) a combination of four genetic alterations, including human telomerase reverse transcriptase overexpression, bypass of p16/RB and p53 pathways, and mutant K-RAS(V12) or mutant EGFR, is still not sufficient for HBECs to completely transform to cancer; and (c) EGFR tyrosine kinase inhibitors inhibit the growth of preneoplastic HBEC cells, suggesting their potential for chemoprevention.

    Topics: Aged; Bronchi; Cell Adhesion; Cell Growth Processes; Cell Transformation, Neoplastic; Dual Specificity Phosphatase 6; Epidermal Growth Factor; Epithelial Cells; ErbB Receptors; Erlotinib Hydrochloride; Female; Gefitinib; Genes, erbB-1; Genes, p16; Genes, p53; Genes, ras; Humans; Lung Neoplasms; Mitogen-Activated Protein Kinase Kinases; Oncogenes; Phosphorylation; Protein Tyrosine Phosphatases; Proto-Oncogene Proteins c-akt; Quinazolines; RNA Interference; STAT3 Transcription Factor; Telomerase; Transfection; Up-Regulation

2006
Csk-binding protein (Cbp) negatively regulates epidermal growth factor-induced cell transformation by controlling Src activation.
    Oncogene, 2006, Sep-07, Volume: 25, Issue:40

    Epidermal growth factor receptor (EGFR) and Src tyrosine kinase cooperate in regulating EGFR-mediated cell signaling and promoting cell transformation and tumorigenesis in pathological conditions. Activation of Src is tightly regulated by the C-terminal Src kinase (Csk). The Csk-binding protein (Cbp) is a ubiquitously expressed transmembrane protein. Its functions include suppression of T-cell receptor activation through recruiting Csk and inhibiting Src family kinase (SFK). However, a potential role of Cbp in EGF-induced cell activities has not been investigated. Here, we report that EGF-stimulation-induced Cbp tyrosine phosphorylation followed by Cbp-Csk association, in a SFK-dependent manner. Expression of wild-type (wt) Cbp remarkably suppressed EGF-induced activation of Src, ERK1/2, and Akt-1 enzymes, and NIH3T3 cell transformation, as well as colony formation of a breast cancer cell line (MDA-MB-468) in soft agar. In contrast, expression of CbpY317F or knockdown endogenous Cbp in NIH3T3 cells by RNA interference significantly enhanced EGF-induced activation of these enzymes and cell transformation. In addition, overexpression of multiple receptor tyrosine kinases (RTKs)-induced Cbp tyrosine phosphorylation. These results demonstrate that Cbp functions as a negative regulator of cell transformation and tumor cell growth through downregulation of Src activation, suggesting that Cbp might be broadly involved in RTKs-activated signaling pathways and tumorigenesis.

    Topics: Animals; Carrier Proteins; Caveolin 1; Cell Line, Tumor; Cell Transformation, Neoplastic; Corticosterone; CSK Tyrosine-Protein Kinase; Down-Regulation; Epidermal Growth Factor; Humans; Mice; NIH 3T3 Cells; Phosphorylation; Protein Binding; Protein-Tyrosine Kinases; Proto-Oncogene Proteins; Proto-Oncogene Proteins pp60(c-src); RNA Interference; Signal Transduction; src-Family Kinases; Transfection

2006
Involvement of the paxillin pathway in JB6 Cl41 cell transformation.
    Cancer research, 2006, Jun-01, Volume: 66, Issue:11

    Paxillin is a substrate of the Src tyrosine onco-kinase and is involved in cell transformation, cell spreading, migration, and cancer development mediated through the mitogen-activated protein kinase signaling cascades. Here, we showed that paxillin plays a key role in skin cell transformation induced by epidermal growth factor (EGF) or 12-O-tetradecanoylphorbol-13-acetate (TPA). To investigate the mechanism of paxillin's role in cell transformation, we established a paxillin knockdown stably transfected cell line by introducing small interfering RNA-paxillin (si-paxillin). The si-paxillin cells displayed a dramatic suppression of cell proliferation and anchorage-independent cell transformation induced by EGF or TPA compared with si-mock control cells. In si-paxillin cells, decreased activator protein-1 (AP-1)-dependent luciferase activity corresponded with suppressed AP-1 DNA binding activity. Importantly, knockdown of paxillin inhibited EGF- or TPA-induced c-Jun phosphorylation at Ser(63) and Ser(73). Furthermore, total c-Jun protein level was dramatically decreased in si-paxillin cells and was dependent on serum deprivation time. The down-regulation of c-Jun was restored in si-paxillin cells by treatment with the proteasome inhibitor lactacystin but not by the lysosome inhibitor leupeptin. These results clearly provided evidence that paxillin regulates c-Jun protein level and plays a key role in cell transformation most likely through the regulation of c-Jun stability.

    Topics: Animals; Base Sequence; Cell Growth Processes; Cell Line; Cell Transformation, Neoplastic; Down-Regulation; Epidermal Growth Factor; MAP Kinase Kinase 4; Mice; Molecular Sequence Data; Paxillin; Phosphorylation; Proto-Oncogene Proteins c-jun; RNA, Small Interfering; Serine; Skin; Skin Neoplasms; Tetradecanoylphorbol Acetate; Transfection

2006
Effects of cotransfection of antisense-EGFR and wild-type PTEN cDNA on human glioblastoma cells.
    Neuropathology : official journal of the Japanese Society of Neuropathology, 2006, Volume: 26, Issue:3

    The main molecular genetic changes identified in glioblastomas are overexpression/amplification of the epidermal growth factor receptor (EGFR) gene and mutation/ deletion of the tumor suppressor PTEN gene. These two genetic changes both play important roles in glial tumorigenesis and progression. In this study, we demonstrated that wild-type PTEN transfection inhibited the growth and transforming ability of U87MG cells by 69.3% and 73.5%, respectively. On the other hand, antisense-EGFR transfection inhibited the growth and transforming phenotype of these cells by 50.3% and 46.8%, respectively. However, cotransfection of U87MG cells with wild-type PTEN and antisense EGFR constructs could inhibit the cellular growth by 91.7%. The transforming phenotype of these cells was completely inhibited. In addition, these cotransfected cells showed a differentiated form and expressed much lower telomerase activity than cells transfected with wild-type PTEN or antisense-EGFR alone. In summary, these results suggest that cotransfection is a better approach to suppress glioma cell growth than wild-type PTEN transfer or antisense-EGFR transfection alone. This approach may prove useful as an adjunct therapy in the treatment of glioblastomas.

    Topics: Blotting, Western; Brain Neoplasms; Cell Line, Tumor; Cell Proliferation; Cell Transformation, Neoplastic; DNA, Antisense; DNA, Complementary; Epidermal Growth Factor; Glial Fibrillary Acidic Protein; Glioblastoma; Humans; Immunohistochemistry; PTEN Phosphohydrolase; Reverse Transcriptase Polymerase Chain Reaction; Telomerase; Transfection

2006
Protein kinase C zeta mediates epidermal growth factor-induced growth of head and neck tumor cells by regulating mitogen-activated protein kinase.
    Cancer research, 2006, Jun-15, Volume: 66, Issue:12

    Protein kinase C (PKC) zeta has been implicated as a mediator of epidermal growth factor (EGF) receptor (EGFR) signaling in certain cell types. Because EGFR is ubiquitously expressed in squamous cell carcinomas of the head and neck (SCCHN) and plays a key role in tumor progression, we determined whether PKCzeta is required for tumor cell proliferation and viability. Examination of total and phosphorylated PKCzeta expression in normal oral mucosa, dysplasia, and carcinoma as well as SCCHN tumor cell lines revealed a significant increase in activated PKCzeta expression from normal to malignant tissue. PKCzeta activity is required for EGF-induced extracellular signal-regulated kinase (ERK) activation in both normal human adult epidermal keratinocytes and five of seven SCCHN cell lines. SCCHN cells express constitutively activated EGFR family receptors, and inhibition of either EGFR or mitogen-activated protein kinase (MAPK) activity suppressed DNA synthesis. Consistent with this observation, inhibition of PKCzeta using either kinase-dead PKCzeta mutant or peptide inhibitor suppressed autocrine and EGF-induced DNA synthesis. Finally, PKCzeta inhibition enhanced the effects of both MAPK/ERK kinase (U0126) and broad spectrum PKC inhibitor (chelerythrine chloride) and decreased cell proliferation in SCCHN cell lines. The results indicate that (a) PKCzeta is associated with SCCHN progression, (b) PKCzeta mediates EGF-stimulated MAPK activation in keratinocytes and SCCHN cell lines, (c) PKCzeta mediates EGFR and MAPK-dependent proliferation in SCCHN cell lines; and (d) PKCzeta inhibitors function additively with other inhibitors that target similar or complementary signaling pathways.

    Topics: Alkaloids; Amino Acid Sequence; Benzophenanthridines; Butadienes; Carcinoma, Squamous Cell; Cell Growth Processes; Cell Line, Tumor; Cell Transformation, Neoplastic; DNA, Neoplasm; Enzyme Activation; Epidermal Growth Factor; ErbB Receptors; Head and Neck Neoplasms; Humans; Keratinocytes; Mitogen-Activated Protein Kinases; Molecular Sequence Data; Mouth Mucosa; Mouth Neoplasms; Nitriles; Phenanthridines; Protein Kinase C; Protein Kinase Inhibitors

2006
Expressions of sonic hedgehog, patched, smoothened and Gli-1 in human intestinal stromal tumors and their correlation with prognosis.
    World journal of gastroenterology, 2006, Sep-21, Volume: 12, Issue:35

    To investigate the role that the hedgehog (Hh) signaling pathway, which includes sonic hedgehog (Shh), Patched (Ptc), Smoothened (Smo) and Gli-1, plays in human gastrointestinal stromal tumors (GISTs).. Surgically resected specimens from patients with GISTs, leiomyomas and schwannomas were examined by immunohistochemical staining for aberrant expression of hedgehog signaling components, Shh, Ptc, Smo and Gli-1, respectively.. In GISTs, 58.1% (18 of 31), 77.4% (24 of 31), 80.6% (25 of 31) and 58.1% (18 of 31) of the specimens stained positive for Shh, Ptc, Smo and Gli-1, respectively. In leiomyomas, 92.3% (12 of 13), 92.3% (12 of 13), 69.2% (9 of 13) and 92.3% (12 of 13) stained positive for Shh, Ptc, Smo and Gli-1, respectively. In schwannomas, 83.3% (5 of 6), 83.3% (5 of 6), 83.3% (5 of 6) and 100% (6 of 6) stained positive for Shh, Ptc, Smo and Gli-1, respectively. Immunohistochemistry revealed that the expressions of Shh and Gli-1 were significantly higher in leiomyomas than in GISTs (P < 0.05, respectively). Shh expression strongly correlated with the grade of tumor risk category and with tumor size (P < 0.05, respectively). However, the expressions of Ptc and Smo did not correlate with histopathological differentiation.. These results suggest that the Hh signaling pathway may play an important role in myogenic differentiation and the malignant potential of human intestinal stromal tumors.

    Topics: Cell Transformation, Neoplastic; Epidermal Growth Factor; Gastrointestinal Stromal Tumors; Gene Expression Regulation, Neoplastic; Hedgehog Proteins; Humans; Intestinal Neoplasms; Leiomyoma; Neurilemmoma; Patched Receptors; Prognosis; Protein-Tyrosine Kinases; Receptors, Cell Surface; Receptors, G-Protein-Coupled; Signal Transduction; Smoothened Receptor; Transcription Factors; Zinc Finger Protein GLI1

2006
Ectopic expression of VAV1 reveals an unexpected role in pancreatic cancer tumorigenesis.
    Cancer cell, 2005, Volume: 7, Issue:1

    Herein, we show that the hematopoietic-specific GEF VAV1 is ectopically expressed in primary pancreatic adenocarcinomas due to demethylation of the gene promoter. Interestingly, VAV1-positive tumors had a worse survival rate compared to VAV1-negative tumors. Surprisingly, even in the presence of oncogenic KRAS, VAV1 RNAi abrogates neoplastic cellular proliferation in vitro and in vivo, thus identifying Vav1 as a growth-stimulatory protein in this disease. Vav1 acts synergistically with the EGF receptor to stimulate pancreatic tumor cell proliferation. Mechanistically, the effects of Vav1 require its GEF activity and the activation of Rac1, PAK1, and NF-kappaB and involve cyclin D1 upregulation. Thus, the discovery of prooncogenic pathways regulated by Vav1 makes it an attractive target for therapeutic intervention.

    Topics: Adenocarcinoma; Animals; Cell Cycle Proteins; Cell Line, Tumor; Cell Proliferation; Cell Transformation, Neoplastic; Cyclin D1; DNA Methylation; Epidermal Growth Factor; Humans; Male; Mice; Mice, Nude; p21-Activated Kinases; Pancreatic Neoplasms; Promoter Regions, Genetic; Protein Serine-Threonine Kinases; Proto-Oncogene Proteins; Proto-Oncogene Proteins c-vav; rac1 GTP-Binding Protein; Recombinant Fusion Proteins; Signal Transduction; Survival Rate

2005
Ligand depletion negatively controls the mitogenic activity of epidermal growth factor.
    Experimental cell research, 2005, Apr-01, Volume: 304, Issue:2

    EGF activates the ErbB1 receptor, but there appears only a limited correlation between its receptor binding affinity and mitogenic activity. This is indicated by our present observation that in cells with high ErbB1 expression, including SUM102 breast tumor cells, low affinity EGF/Notch chimeras have similarly high mitogenic activity as EGF, in spite of the fact that EGF is superior in inducing receptor tyrosine phosphorylation and p42/p44 MAP-kinase activity. However, as a result of receptor-mediated internalisation high-affinity ligands such as EGF are depleted much more rapidly from the extracellular medium than low-affinity EGF/Notch chimeras. As a consequence, the mitogenic activity of EGF on ErbB1 overexpressing cells is limited by substantial degradation of internalised ligand in the period before cells enter S-phase, a phenomenon that is not observed for low affinity mutant ligands. The mitogenic activity of EGF on ErbB1 overexpressing cells does therefore not only depend on the applied concentration but also on the total amount of ligand added, and is strongly underestimated when tested in a limited assay volume. No such dependence on the incubation volume was observed for EGF activity on cells with low ErbB1 expression levels and on cells for which EGF is growth inhibitory.

    Topics: Animals; Cell Line, Tumor; Cell Transformation, Neoplastic; Endocytosis; Epidermal Growth Factor; ErbB Receptors; Feedback, Physiological; Female; Growth Substances; Humans; Ligands; Mice; Mitosis; Mutation; Neoplasms; NIH 3T3 Cells; Protein Binding; Receptor Aggregation; Receptor, Notch1; Receptors, Cell Surface; Recombinant Fusion Proteins; S Phase; Transcription Factors

2005
Cdc42 and Ras cooperate to mediate cellular transformation by intersectin-L.
    The Journal of biological chemistry, 2005, Jun-17, Volume: 280, Issue:24

    Cdc42, a Ras-related GTP-binding protein, has been implicated in the regulation of the actin cytoskeleton, membrane trafficking, cell-cycle progression, and malignant transformation. We have shown previously that a Cdc42 mutant (Cdc42(F28L)), capable of spontaneously exchanging GDP for GTP (referred to as "fast-cycling"), transformed NIH 3T3 cells because of its ability to interfere with epidermal growth factor receptor (EGFR)-Cbl interactions and EGFR down-regulation. To further examine the link between the hyperactivation of Cdc42 and its ability to alter EGFR signaling and thereby cause cellular transformation, we examined the effects of expressing different forms of the Cdc42-specific guanine nucleotide exchange factor, intersectin-L, in fibroblasts. Full-length intersectin-L exhibited little ability to stimulate nucleotide exchange on Cdc42, whereas a truncated version that contained five Src homology 3 (SH3) domains, the Dbl and pleckstrin homology domains (DH and PH domains, respectively), and a C2 domain (designated as SH3A-C2) showed modest guanine nucleotide exchange factor activity, whereas a form containing just the DH, PH, and C2 domains (DH-C2) strongly activated Cdc42. However, DH-C2 showed little ability to stimulate growth in low serum or colony formation in soft agar, whereas SH3A-C2 gave rise to a much stronger stimulation of cell growth in low serum and was highly effective in stimulating colony formation. Moreover, although SH3A-C2 strongly transformed fibroblasts, it differed from the actions of the Cdc42(F28L) mutant, as SH3A-C2 showed little ability to alter EGFR levels or the lifetime of EGF-coupled signaling through ERK. Rather, we found that SH3A-C2 exhibited strong transforming activity through its ability to mediate cooperation between Ras and Cdc42.

    Topics: Actins; Adaptor Proteins, Vesicular Transport; Agar; Animals; cdc42 GTP-Binding Protein; Cell Cycle; Cell Line; Cell Line, Transformed; Cell Transformation, Neoplastic; COS Cells; Enzyme Inhibitors; Epidermal Growth Factor; ErbB Receptors; Fibroblasts; Guanosine Diphosphate; Guanosine Triphosphate; Humans; Immunoblotting; Immunoprecipitation; JNK Mitogen-Activated Protein Kinases; MAP Kinase Kinase 4; Mice; Microscopy, Fluorescence; Mitogen-Activated Protein Kinase Kinases; Models, Biological; Mutation; NIH 3T3 Cells; Phosphatidylinositol 3-Kinases; Plasmids; Protein Binding; Protein Structure, Tertiary; ras Proteins; Signal Transduction; src Homology Domains; Time Factors; Transfection

2005
2-Arachidonoylglycerol stimulates activator protein-1-dependent transcriptional activity and enhances epidermal growth factor-induced cell transformation in JB6 P+ cells.
    The Journal of biological chemistry, 2005, Jul-22, Volume: 280, Issue:29

    2-Arachidonoylglycerol (2-AG) is the most abundant endocannabinoid, and it plays a critical role in cannabinoid receptor-mediated cell signaling. Although 2-AG was shown to induce ERK activation via the cannabinoid receptor 1 (CB1), only a nonspecific CB receptor agonist and antagonist was used in those studies. Whether cannabinoid receptor 2 (CB2) is involved in 2-AG-induced ERK activation is still unclear. Moreover, whether 2-AG is involved in mediation of AP-1 activity and cell transformation is also not known. In the present study, we show that 2-AG stimulates AP-1-dependent transcriptional activity and enhances epidermal growth factor-induced cell transformation in mouse epidermal JB6 P+ Cl41 cells. Using JB6 P+ C141 cells, stably transfected with an AP-1 luciferase reporter, we found that 10 microm 2-AG induced up to a 3-fold stimulation of AP-1 transcriptional activity. The AP-1 stimulation appeared to be mediated by ERK but not JNK or p38 kinase. PD98059, a specific inhibitor of MEK1, almost completely blocked 2-AG-induced ERK phosphorylation and AP-1 activation. Using CB1/2-/- murine embryonic fibroblasts, we present the first direct evidence that both cannabinoid receptors 1 and 2 (CB1/2) are involved in 2-AG-induced ERK activation. 2-AG could not stimulate ERK phosphorylation or Fyn kinase activity in dominant negative Fyn. In addition, the Fyn inhibitor PP2 blocked 2-AG-induced Fyn kinase activity and ERK phosphorylation and activity. Small interfering RNA Fyn also suppressed 2-AG-induced ERK phosphorylation. Interestingly, 2-AG enhanced epidermal growth factor-induced AP-1 DNA binding and cell transformation. Taken together, our data provide direct evidence suggesting that 2-AG may have a novel role in cell transformation and carcinogenesis in a signaling pathway involving CB1/2 and activation of Fyn, ERKs, and AP-1.

    Topics: Animals; Arachidonic Acids; Cell Line; Cell Transformation, Neoplastic; Drug Synergism; Endocannabinoids; Epidermal Growth Factor; Extracellular Signal-Regulated MAP Kinases; Glycerides; Mice; Proto-Oncogene Proteins; Proto-Oncogene Proteins c-fyn; Receptor, Cannabinoid, CB1; Receptor, Cannabinoid, CB2; Signal Transduction; src-Family Kinases; Transcription Factor AP-1; Transfection

2005
Uncoupling of epidermal growth factor-dependent proliferation and invasion in a model of squamous carcinoma progression.
    Oral oncology, 2005, Volume: 41, Issue:7

    Cell lines pairs were established from a primary squamous carcinoma of tongue and a lymph node metastasis and their biological behavior characterized. HN12 cells, derived from metastatic SCC, formed tumors upon subcutaneous transplantation to athymic mice, whereas HN4, derived from a primary lesion in the same individual, were non-tumorigenic in this assay. EGF stimulated proliferation of HN4 cells; in comparison, not only were metastatic HN12 cells refractory to the stimulatory effects of this growth factor but showed inhibition at higher growth factor concentrations. However, in contrast to the effects on proliferation, EGF (10 ng/ml) readily induced HN12 cells to invade in Boyden chamber assays whereas HN4 were non-invasive under these conditions. The invasive properties of HN12 cells were apparently independent of MMP-2 activity, as levels of active MMP-2 were higher in the non-invasive cells. However, EGF stimulated MMP-9 activity in invasive cells. Additionally, HN12 cells expressed constitutively high levels of active MMP-7 and MMP-3/10. The pharmacological agents LY294002, PD098059, SP600125, or SB202190 inhibited invasion of HN12 cells, suggesting requirement for phosphoinositide 3-OH kinase- and mitogen activated protein kinase-dependent pathways in the process. The data indicate that distinct biochemical differences distinguish metastatic squamous carcinoma cells from those derived from corresponding primary tumors, resulting in their contrasting biological properties.

    Topics: Animals; Carcinoma, Squamous Cell; Cell Line, Tumor; Cell Proliferation; Cell Transformation, Neoplastic; Epidermal Growth Factor; Lymphatic Metastasis; Mice; Mice, SCID; Neoplasm Invasiveness; Neoplasm Metastasis; Tongue Neoplasms

2005
Phosphorylation of histone H3 at serine 10 is indispensable for neoplastic cell transformation.
    Cancer research, 2005, Jul-01, Volume: 65, Issue:13

    Very little is known about the role of histone H3 phosphorylation in malignant transformation and cancer development. Here, we examine the function of H3 phosphorylation in cell transformation in vivo. Introduction of small interfering RNA-H3 into JB6 cells resulted in decreased epidermal growth factor (EGF)-induced cell transformation. In contrast, wild-type histone H3 (H3 WT)-overexpressing cells markedly stimulated EGF-induced cell transformation, whereas the H3 mutant S10A cells suppressed transformation. When H3 WT was overexpressed, EGF induction of c-fos and c-jun promoter activity was significantly increased compared with control cells but not in the H3 mutant S10A or S28A cells. In addition, activator protein-1 activity in H3 WT-overexpressing cells was markedly up-regulated by EGF in contrast to the H3 mutant S10A or S28A cells. These results indicate that the phosphorylation of histone H3 at Ser10 is an essential regulatory mechanism for EGF-induced neoplastic cell transformation.

    Topics: Amino Acid Sequence; Animals; Base Sequence; Cell Transformation, Neoplastic; Epidermal Growth Factor; Epithelial Cells; Gene Expression Regulation; Genes, fos; Genes, jun; Histones; Mice; Molecular Sequence Data; Phosphorylation; Promoter Regions, Genetic; Proto-Oncogene Proteins c-fos; Proto-Oncogene Proteins c-jun; RNA, Messenger; RNA, Small Interfering; Serine; Transcription Factor AP-1

2005
Inhibition of epidermal growth factor-induced cell transformation and Akt activation by caffeine.
    Molecular carcinogenesis, 2005, Volume: 44, Issue:1

    We found that caffeine significantly inhibited epidermal growth factor (EGF)- and 12-O-tetradecanoylphorbol-13-acetate (TPA)-induced cell transformation in the JB6 mouse epidermal cell line. The tumor promoter-induced cell transformation was also blocked by treatment with an adenosine A1 receptor antagonist, 8-phenyltheophylline (8-PTH). Caffeine slightly attenuated activation of EGF-induced activator protein 1 (AP-1) activation, which play important roles in cell transformation, but only at the highest concentration examined (1 mM). Interestingly, pretreatment with caffeine suppressed EGF-induced phosphorylation and activation of Akt and ribosomal p 70 S6 protein kinase (p 70 S 6 K), a target of Akt, without inhibiting phosphatidylinositol 3-kinase (PI 3 K) activation. The inhibition of Akt activation of caffeine was not a result of its adenosine receptor antagonism. Because Akt plays a key role in signal transduction pathways leading to cell proliferation and apoptosis, our results provide novel insight into possible mechanisms of the chemotherapeutic effect of caffeine.

    Topics: Animals; Caffeine; Cell Line; Cell Transformation, Neoplastic; Cyclic AMP; Enzyme Activation; Epidermal Growth Factor; Mice; Phosphatidylinositol 3-Kinases; Phosphorylation; Promoter Regions, Genetic; Protein Serine-Threonine Kinases; Proto-Oncogene Proteins; Proto-Oncogene Proteins c-akt; Purinergic P1 Receptor Antagonists; Theophylline; Transcription Factor AP-1

2005
Acquisition of androgen independence by human prostate epithelial cells during arsenic-induced malignant transformation.
    Environmental health perspectives, 2005, Volume: 113, Issue:9

    Lethal phenotypes of human prostate cancer are characterized by progression to androgen independence, although the mechanisms behind this progression remain unclear. Arsenic is a potential human prostate carcinogen that may affect tumor progression. In this study, we used a prostate cancer cell model in which an immortalized, nontumorigenic human prostate epithelial cell line (RWPE-1) had been malignantly transformed by chronic low-level arsenic to help determine whether arsenic affects prostate tumor progression. Control and CAsE-PE (chronic-arsenic-exposed human prostate epithelial) cells were continuously maintained in a complete medium [keratinocyte serum-free medium (K-SFM) with bovine pituitary extract and epidermal growth factor] or in a steroid-depleted medium (K-SFM alone). The arsenic-transformed cells showed a more rapid proliferation rate in complete medium than did control cells and also showed sustained proliferation in steroid-reduced medium. Although both control and CAsE-PE cells showed similar levels of androgen receptor (AR), androgens were less effective in stimulating cell proliferation and AR-related gene expression in CAsE-PE cells. For instance, dihydrotestosterone caused a 4.5-fold increase in prostate-specific antigen transcript in control cells but only a 1.5-fold increase in CAsE-PE cells. CAsE-PE cells also showed relatively low levels of growth stimulation by nonandrogen steroids, such as estradiol. Thus, arsenic-induced malignant transformation is associated with acquired androgen independence in human prostate cells. This acquired androgen independence was apparently not due to AR up-regulation, increased activity, or altered ligand specificity. The precise manner in which arsenic altered CAsE-PE growth and progression is undefined but may involve a bypass of AR involving direct stimulation of downstream signaling pathways.

    Topics: Androgen Antagonists; Androgens; Animals; Arsenic; Cell Line; Cell Proliferation; Cell Transformation, Neoplastic; Dihydrotestosterone; Epidermal Growth Factor; Epithelial Cells; Estradiol; Estrogen Receptor alpha; Estrogen Receptor beta; Flutamide; Gene Expression Regulation; Humans; Male; Mice; Pituitary Gland; Prostate; Prostatic Neoplasms; Receptors, Androgen; RNA, Messenger

2005
Inhibition of epidermal growth factor-induced cell transformation by tannins.
    Phytochemistry, 2005, Volume: 66, Issue:17

    The mouse epidermal JB6 cell system is a well developed model for studying tumor promotion, and the JB6 Cl 41 promotion sensitive (P+) cell line, in which transformed colonies are induced by epidermal growth factor (EGF), was used to test the anti-tumor promoting effect of seven tannins and two triterpenoids. We found that six tannins, ellagitannins (compounds 1, 2, 3 and 4) and chromone gallates (compounds 6 and 7), significantly blocked EGF-induced cell transformation in a concentration-dependent manner. The inhibition of cell transformation by the tannins was not due to growth inhibition. The ellagitannins, but not the chromone gallates, significantly attenuated EGF-induced activator protein 1 (AP-1) activation, a transcription factor. Compounds 1 and 3, among the ellagitannins analysed, inhibited the EGF-induced phosphorylation of extracellular-signal regulated protein kinases and p38 kinases, which regulate AP-1 activation. On the other hand, compounds 3 and 4 suppressed EGF-induced phosphatidylinositol 3-kinase (PI3K) activation. In addition, all tannins that blocked cell transformation markedly inhibited EGF-induced activation of Akt, a downstream effector of PI3K. Because signal-transduction pathways, including AP-1 and PI3K pathways, have been focused as prime targets for chemopreventive phytochemicals, our results suggest that inhibition by tannins of EGF-induced neoplastic transformation in JB6 cells is related to blocking of Akt activation, and also attenuation of AP-1 activation for ellagitannins.

    Topics: Animals; Cell Line, Transformed; Cell Transformation, Neoplastic; Enzyme Activation; Epidermal Growth Factor; ErbB Receptors; Extracellular Signal-Regulated MAP Kinases; Mice; p38 Mitogen-Activated Protein Kinases; Phosphorylation; Proto-Oncogene Proteins c-akt; Tannins; Tyrosine

2005
Epidermal growth factor-independent transformation of Ba/F3 cells with cancer-derived epidermal growth factor receptor mutants induces gefitinib-sensitive cell cycle progression.
    Cancer research, 2005, Oct-01, Volume: 65, Issue:19

    Epidermal growth factor receptor (EGFR) plays critical roles in many biological processes and in tumorigenesis. Here, we show that two mutated EGFRs found in lung and other malignancies, EGFR-G719S and EGFR-L858R, could transform Ba/F3 cells to interleukin-3 (IL-3)-independent growth, in a ligand-independent manner, an activity associated with the transforming function of other mutated tyrosine kinases. The mutated receptors are autophosphorylated in the absence of IL-3 without EGF stimulation, and their expression led to the constitutive activation of signal transducers and activators of transcription 5, extracellular signal-regulated kinase 1/2 (ERK1/2), ERK5, and AKT. In wild-type EGFR-expressing Ba/F3 cells, the major EGF-mediated signaling pathways were still intact. Gefitinib inhibited the growth of mutant EGFR-transformed Ba/F3 cells. Strikingly, the gefitinib sensitivity of cells expressing the L858R mutant was significantly greater than that of cells expressing the G719S mutant form, suggesting that distinct EGFR mutations may be differentially sensitive to small-molecule inhibitors. Furthermore, our data showed an antiproliferative effect of gefitinib on the EGFR-transformed Ba/F3 cells. Our results provide a model system to study the function of mutated EGFR and the differential effects of pharmacologic EGFR inhibition on the distinct mutant forms of this tyrosine kinase.

    Topics: Animals; Antineoplastic Agents; Cell Cycle; Cell Growth Processes; Cell Transformation, Neoplastic; Epidermal Growth Factor; ErbB Receptors; Gefitinib; Interleukin-3; Mice; Mutagenesis, Site-Directed; Mutation, Missense; Protein Kinase Inhibitors; Quinazolines; Signal Transduction; Transfection

2005
Cooperative interactions of HER-2 and HPV-16 oncoproteins in the malignant transformation of human mammary epithelial cells.
    Neoplasia (New York, N.Y.), 2005, Volume: 7, Issue:8

    To better understand the mechanisms of transformation by the oncogene HER-2, we transduced the human mammary epithelial (HME) cell line MCF-10A with HER-2 and developed a cell line that appeared to moderately overexpress HER-2. These MCF-10HER-2 cells were unable to grow in the absence of epidermal growth factor (EGF). However, coexpression of HER-2 with the HPV-16 oncoproteins E6 and E7 resulted in EGF-independent cells that expressed very high levels of constitutively activated HER-2. Interestingly, coexpression of E7 with HER-2 resulted in cells that were EGF-independent for growth but did not express HER-2 to high levels, and coexpression of E6 with HER-2 resulted in cells expressing higher levels of HER-2, which were still dependent on EGF for growth and survival. The MCF-10HER-2E7 and HER-2/E6E7 cells exhibited constitutive activation of a form of epidermal growth factor receptor (EGFR) that had a faster electrophoretic mobility than EGFR activated by exogenous growth factors. Exposure of cells with EGFR activation to ZD1839 (Iressa), at concentrations specific for EGFR, had little or no influence on proliferation of cells with amplified HER-2 but little or no EGFR. These results indicate that HER-2, E6, and E7 cooperate with endogenous EGFR to yield fully transformed cells.

    Topics: Breast Neoplasms; Cell Line, Transformed; Cell Proliferation; Cell Transformation, Neoplastic; Epidermal Growth Factor; Epithelial Cells; ErbB Receptors; Human papillomavirus 16; Humans; Mammary Glands, Human; Oncogene Proteins, Viral; Papillomavirus E7 Proteins; Receptor, ErbB-2; Repressor Proteins

2005
Transformation of human bronchial epithelial cells alters responsiveness to inflammatory cytokines.
    BMC cancer, 2005, Nov-04, Volume: 5

    Inflammation is commonly associated with lung tumors. Since inflammatory mediators, including members of the interleukin-6 (IL-6) cytokine family, suppress proliferation of normal epithelial cells, we hypothesized that epithelial cells must develop mechanisms to evade this inhibition during the tumorigenesis. This study compared the cytokine responses of normal epithelial cells to that of premalignant cells.. Short-term primary cultures of epithelial cells were established from bronchial brushings. Paired sets of brushings were obtained from areas of normal bronchial epithelium and from areas of metaplastic or dysplastic epithelium, or areas of frank endobronchial carcinoma. In 43 paired cultures, the signalling through the signal transducer and activator of transcription (STAT) and extracellular regulated kinase (ERK) pathways and growth regulation by IL-6, leukemia inhibitory factor (LIF), oncostatin M (OSM), interferon-gamma (IFNgamma) or epidermal growth factor (EGF) were determined. Inducible expression and function of the leukemia inhibitory factor receptor was assessed by treatment with the histone deacetylase inhibitor depsipeptide.. Normal epithelial cells respond strongly to OSM, IFNgamma and EGF, and respond moderately to IL-6, and do not exhibit a detectable response to LIF. In preneoplastic cells, the aberrant signaling that was detected most frequently was an elevated activation of ERK, a reduced or increased IL-6 and EGF response, and an increased LIF response. Some of these changes in preneoplastic cell signaling approach those observed in established lung cancer cell lines. Epigenetic control of LIF receptor expression by histone acetylation can account for the gain of LIF responsiveness. OSM and macrophage-derived cytokines suppressed proliferation of normal epithelial cells, but reduced inhibition or even stimulated proliferation was noted for preneoplastic cells. These alterations likely contribute to the supporting effects that inflammation has on lung tumor progression.. This study indicates that during the earliest stage of premalignant transformation, a modified response to cytokines and EGF is evident. Some of the altered cytokine responses in primary premalignant cells are comparable to those seen in established lung cancer cell lines.

    Topics: Cell Line, Transformed; Cell Line, Tumor; Cell Transformation, Neoplastic; Cells, Cultured; Cytokines; Densitometry; Epidermal Growth Factor; Epithelial Cells; Extracellular Signal-Regulated MAP Kinases; Fibroblasts; Histones; Humans; Inflammation; Interferon-gamma; Interleukin-6; Lung; Lung Neoplasms; Macrophages; Oncostatin M; Signal Transduction; Time Factors; Treatment Outcome

2005
ESX induces transformation and functional epithelial to mesenchymal transition in MCF-12A mammary epithelial cells.
    Oncogene, 2004, Mar-04, Volume: 23, Issue:9

    ESX is an epithelial-restricted member of a large family of transcription factors known as the Ets family. ESX expression has been shown to be correlated with Her2/neu proto-oncogene amplification in highly aggressive breast cancers and induced by Her2/neu in breast cell lines, but its role in tumorigenesis is unknown. Previously, we have shown that ESX enhances breast cell survival in colony-formation assays. In order to determine whether ESX can act as a transforming gene, we stably transfected MCF-12A human mammary epithelial cells with the ESX expression vector, pCGN2-HA-ESX. The MCF-12A cell line is immortalized, but nontransformed, and importantly, these cells fail to express endogenous ESX protein. We used pCGN2-HA-Ets-2 and pSVRas expression vectors as positive controls for transformation. Like HA-Ets-2 and V12-Ras, stable expression of ESX induced EGF-independent proliferation, serum-independent MAPK phosphorylation and growth in soft agar. Additionally, stable ESX expression conferred increased cell adhesion, motility and invasion in two-dimensional and transwell filter assays, and an epithelial to mesenchymal morphological transition. In three-dimensional cultures, parental and vector control (pCGN2) cells formed highly organized duct-like structures with evidence of cell polarity, ECM adhesion-dependent proliferation and cell survival, and lack of cellular invasion into surrounding matrix. Remarkably, the ESX stable cells formed solid, disorganized structures, with lack of cell polarity, loss of adhesion junctions and cytokeratin staining and loss of dependence on ECM adhesion for cell proliferation and survival. In addition, ESX cells invaded the surrounding matrix, indicative of a transformed and metastatic phenotype. Taken together, these data show that ESX expression alone confers a transformed and in vitro metastatic phenotype to otherwise normal MCF-12A cells.

    Topics: Breast; Cell Adhesion; Cell Differentiation; Cell Division; Cell Line; Cell Movement; Cell Transformation, Neoplastic; Contact Inhibition; DNA-Binding Proteins; Epidermal Growth Factor; Epithelial Cells; Humans; Mesoderm; Mitogen-Activated Protein Kinases; Neoplasm Invasiveness; Oncogene Protein p21(ras); Phenotype; Phosphorylation; Proto-Oncogene Mas; Proto-Oncogene Protein c-ets-2; Proto-Oncogene Proteins; Proto-Oncogene Proteins c-ets; Repressor Proteins; Trans-Activators; Transcription Factors

2004
Cell-to-cell adhesion modulates Stat3 activity in normal and breast carcinoma cells.
    Oncogene, 2004, Apr-08, Volume: 23, Issue:15

    Stat3 (signal transducer and activator of transcription-3) activity is required for transformation by a number of oncogenes, while a constitutively active form of Stat3 alone is sufficient to induce neoplastic transformation. Although in most instances Stat3 is growth-promoting, the impact of cell density on Stat3 activation status and the biological importance of Stat3 during growth arrest have not been characterized. Previous results indicated that cell density alters tyrosine phosphorylation levels of cultured cells. Since signalling through Stat3 is determined by a key phosphorylation at tyr705, we examined the effects of cell density upon Stat3 activity in normal breast epithelial cells, breast carcinoma lines and normal mouse fibroblasts. Intriguingly, the results revealed a dramatic increase in Stat3, tyr705 phosphorylation and activity with cell density, which gradually declined at later stages. This activation was dependent upon cell-cell contact, since it was eliminated if cell adhesion was disrupted through calcium chelation, while it was reinstated through cell aggregation. Furthermore, this activation was suppressed following inhibition of JAKs (Janus kinases) but not inhibition of Fer, IGF1-R, or kinases of the c-Src family. On the other hand, constitutively active Stat3 in carcinoma lines, known to harbor activated Src, was blocked by pharmacological inhibitors of Src as well as JAKs. These results point to the existence of two distinct pathways of Stat3 activation in breast carcinomas, based on Src dependence. More importantly, our results suggest that Stat3 activity is upregulated during the confluence-mediated growth arrest by a signalling mechanism that requires JAKs.

    Topics: Animals; Apoptosis; Blotting, Western; Breast Neoplasms; Calcium; Cell Adhesion; Cell Division; Cell Line, Tumor; Cell Transformation, Neoplastic; Cells, Cultured; DNA-Binding Proteins; Down-Regulation; Enzyme Activation; Epidermal Growth Factor; Fibroblasts; Humans; In Situ Nick-End Labeling; Janus Kinase 1; Luciferases; Mice; NIH 3T3 Cells; Phosphorylation; Protein-Tyrosine Kinases; Rats; Signal Transduction; src-Family Kinases; STAT3 Transcription Factor; Time Factors; Trans-Activators; Tyrosine; Up-Regulation

2004
Role of the Grb2-associated binder 1/SHP-2 interaction in cell growth and transformation.
    Cancer research, 2004, Mar-15, Volume: 64, Issue:6

    Grb2-associated binder 1 (Gab1) is a docking protein that is tyrosine phosphorylated following the activation of multiple cytokine receptors and receptor tyrosine kinases. Its function then is to recruit and activate multiple signaling molecules. In our previous work, we showed that Gab1 enhances cell growth and induces the transformed phenotype in NIH3T3 cells downstream of the epidermal growth factor (EGF) receptor. In this report, we analyze how it produces these effects. Because SHP-2 is the major binding partner of Gab1, we mutated its binding site in the Gab1 cDNA (Gab1/DeltaSHP-2). This construct was stably overexpressed in NIH3T3 cells (3T3-Gab1/DeltaSHP-2) and in the wild-type Gab1 cDNA (3T3-Gab1) or an empty expression vector (3T3-CTR). Our findings show that after EGF stimulation, Gab1/DeltaSHP-2 has a higher level of tyrosine phosphorylation at early time points than Gab1. Gab1/DeltaSHP-2 recruits more phosphatidylinositol 3'-kinase than Gab1 after EGF triggering, which accounts for a higher and more sustained AKT activation in 3T3-Gab1/DeltaSHP-2 cells relative to 3T3-Gab1 fibroblasts. Moreover, 3T3-Gab1/DeltaSHP-2 cells demonstrate a higher level of extracellular-regulated kinase 1 activation at early time points of EGF stimulation. However, there was an unexpected decrease in c-fos promoter induction in 3T3-Gab1/DeltaSHP-2 cells when compared with 3T3-Gab1 cells. Additionally, the 3T3-Gab1/DeltaSHP-2 cells show a reversion of the transformed phenotype, including fewer morphologic changes, an increase in stress fiber cytoskeletal organization, and a decrease in cell proliferation and anchorage independent growth. These results reveal that the Gab1/SHP-2 interaction is essential for cell growth and transformation but that this must occur through a novel pathway that is independent of extracellular-regulated kinase or AKT. On the basis of its role in growth and transformation, the Gab1/SHP-2 interaction may become an attractive target for the pharmacologic intervention of malignant cell growth.

    Topics: 3T3 Cells; Adaptor Proteins, Signal Transducing; Animals; Cell Adhesion; Cell Division; Cell Transformation, Neoplastic; Enzyme Activation; Epidermal Growth Factor; ErbB Receptors; Genes, fos; Humans; Intracellular Signaling Peptides and Proteins; Mice; Mitogen-Activated Protein Kinase 3; Mitogen-Activated Protein Kinases; Phosphatidylinositol 3-Kinases; Phosphoproteins; Phosphorylation; Promoter Regions, Genetic; Protein Serine-Threonine Kinases; Protein Tyrosine Phosphatase, Non-Receptor Type 11; Protein Tyrosine Phosphatases; Proto-Oncogene Proteins; Proto-Oncogene Proteins c-akt; SH2 Domain-Containing Protein Tyrosine Phosphatases; Signal Transduction; src Homology Domains; Stress Fibers; Tyrosine

2004
The molecular scaffold KSR1 regulates the proliferative and oncogenic potential of cells.
    Molecular and cellular biology, 2004, Volume: 24, Issue:10

    The specificity of signaling through mitogen-activated protein kinase pathways has been attributed to both the control of intensity and duration of signaling and the actions of protein scaffolds. Here we demonstrate that the molecular scaffold KSR1 regulates the intensity and duration of ERK activation to modulate a cell's proliferative and oncogenic potential. Deletion of KSR1 eliminates the prolonged phase of ERK activation induced by platelet-derived growth factor and blocks Ras(V12)-induced transformation. The introduction of KSR1 into KSR1(-/-) mouse embryo fibroblasts causes a concentration-dependent increase in signaling and transformation, to a maximum at 14 times the wild-type KSR1 expression levels, but inhibits these responses at higher expression levels. An increase in KSR1 expression to levels that are optimal for signaling leads to a threefold increase in proliferative capacity and is coincident with the level of KSR1 expression that maximally associates with all members of the Raf/MEK/ERK cascade. These data reveal that cells contain a reserve proliferative capacity that is accessible by the optimal expression of a noncatalytic signaling component and that altering the expression level of a molecular scaffold can modulate the actions of growth factors and oncogenes.

    Topics: Animals; Cell Division; Cell Line; Cell Transformation, Neoplastic; Epidermal Growth Factor; Genes, ras; Kinetics; MAP Kinase Signaling System; Mice; Mice, Knockout; Platelet-Derived Growth Factor; Protein Kinases; Recombinant Fusion Proteins

2004
Overexpression of the C-terminal PG-M/versican domain impairs growth of tumor cells by intervening in the interaction between epidermal growth factor receptor and beta1-integrin.
    Journal of cell science, 2004, May-01, Volume: 117, Issue:Pt 11

    Versican is highly expressed in many types of tumors. In a previous study, we found that a G3 mutant [G3DeltaEGF; a versican G3 domain lacking two epidermal growth factor (EGF)-like motifs] exerted a dominant-negative effect on versican secretion and binding. Here, we report that astrocytoma U87 cells expressing the versican G3 mutant lost the hallmark of cell transformation and tumorigenesis in vitro and in vivo. U87 cells expressing G3DeltaEGF had enhanced cell adhesion and spreading, but lost the tumor characteristic of anchorage-independent growth. When U87 cells were deprived of serum, FAK was quickly dephosphorylated, integrin/EGF-receptor (EGFR) complexes dissociated and the cells retained an appropriate level of EGFR phosphorylation. These cells quickly detached, migrated, rounded, reorganized and survived. However, after serum withdrawal from G3DeltaEGF-transfected U87 cells, sustained FAK phosphorylation and integrin-EGFR association were observed, but a greatly reduced EGFR phosphorylation. These cells remained spread and continued to grow before undergoing massive apoptosis. The addition of EGF promoted U87 cell rounding but had little effect on G3DeltaEGF-transfected cells owing to reduced EGFR phosphorylation. Our study sheds light on the question of how the matrix molecule versican modulates tumorigenesis by affecting integrin and EGFR signals.

    Topics: Apoptosis; Cell Adhesion; Cell Line, Tumor; Cell Proliferation; Cell Transformation, Neoplastic; Chondroitin Sulfate Proteoglycans; Culture Media, Serum-Free; Epidermal Growth Factor; ErbB Receptors; Focal Adhesion Kinase 1; Focal Adhesion Protein-Tyrosine Kinases; Humans; Integrin beta1; Lectins, C-Type; Phosphorylation; Protein Binding; Protein-Tyrosine Kinases; Sequence Deletion; Transfection; Tumor Stem Cell Assay; Versicans

2004
A novel role for mixed-lineage kinase-like mitogen-activated protein triple kinase alpha in neoplastic cell transformation and tumor development.
    Cancer research, 2004, Jun-01, Volume: 64, Issue:11

    Previously, no member of the mixed-lineage kinase (MLK) protein family was known to function as an oncogene. Here, we demonstrate that MLK-like mitogen-activated protein triple kinase (MLTK)-alpha, a member of the MLK family, induced neoplastic cell transformation and tumorigenesis in athymic nude mice. Introduction of small interference RNA (siRNA)-MLTK-alpha into MLTK-alpha-overexpressing cells dramatically suppressed cell transformation. Nuclear accumulation of the pHisG-MLTK-alpha fusion protein was observed after epidermal growth factor or 12-O-tetradecanoylphorbol-13-acetate treatment. Phosphorylation of downstream mitogen-activated protein kinase-targeted transcription factors including c-Myc, Elk-1, c-Jun, and activating transcription factor (ATF) 2 was also differentially enhanced in MLTK-alpha-overexpressing cells exposed to epidermal growth factor or 12-O-tetradecanoylphorbol-13-acetate stimulation compared with cells expressing mock vector or siRNA-MLTK-alpha. Very importantly, MLTK-alpha-overexpressing cells formed fibrosarcomas when injected s.c. into athymic nude mice, whereas almost no tumor formation was observed in mice that received injections of mock or siRNA-MLTK-alpha stably transfected cells. These results are the first to indicate that MLTK-alpha plays a key role in neoplastic cell transformation and cancer development.

    Topics: Animals; Base Sequence; Carcinogens; Cell Adhesion; Cell Division; Cell Transformation, Neoplastic; Cells, Cultured; Epidermal Growth Factor; MAP Kinase Kinase Kinases; MAP Kinase Signaling System; Mice; Mice, Nude; Molecular Sequence Data; Phosphorylation; RNA, Small Interfering; Skin; Skin Neoplasms; Tetradecanoylphorbol Acetate; Transcription Factors

2004
HB-EGF is a potent inducer of tumor growth and angiogenesis.
    Cancer research, 2004, Aug-01, Volume: 64, Issue:15

    Heparin-binding epidermal growth factor-like growth factor (HB-EGF) has been shown to stimulate the growth of a variety of cells in an autocrine or paracrine manner. Although HB-EGF is widely expressed in tumors compared with normal tissue, its contribution to tumorigenicity is unknown. HB-EGF can be produced as a membrane-anchored form (pro-HB-EGF) and later processed to a soluble form (s-HB-EGF), although a significant amount of pro-HB-EGF remains uncleaved on the cell surface. To understand the roles of two forms of HB-EGF in promoting tumor growth, we have studied the effects of HB-EGF expression in the process of tumorigenesis using in vitro and in vivo systems. We demonstrate here that in EJ human bladder cancer cells containing a tetracycline-regulatable s-HB-EGF or pro-HB-EGF expression system, s-HB-EGF expression increased their transformed phenotypes, including growth rate, colony-forming ability, and activation of cyclin D1 promoter, as well as induction of vascular endothelial growth factor in vitro. Moreover, s-HB-EGF or wild-type HB-EGF induced the expression and activities of the metalloproteases, MMP-9 and MMP-3, leading to enhanced cell migration. In vivo studies also demonstrated that tumor cells expressing s-HB-EGF or wild-type HB-EGF significantly enhanced tumorigenic potential in athymic nude mice and exerted an angiogenic effect, increasing the density and size of tumor blood vessels. However, cells expressing solely pro-HB-EGF did not exhibit any significant tumorigenic potential. These findings establish s-HB-EGF as a potent inducer of tumor growth and angiogenesis and suggest that therapeutic intervention aimed at the inhibition of s-HB-EGF functions may be useful in cancer treatment.

    Topics: Animals; Blotting, Northern; Cell Division; Cell Membrane; Cell Movement; Cell Transformation, Neoplastic; Colony-Forming Units Assay; Cyclin D1; Cytosol; Epidermal Growth Factor; Gene Expression Regulation; Heparin-binding EGF-like Growth Factor; Humans; Intercellular Signaling Peptides and Proteins; Matrix Metalloproteinase 3; Matrix Metalloproteinase 9; Mice; Mice, Nude; Neovascularization, Pathologic; Phenotype; Promoter Regions, Genetic; Tumor Cells, Cultured; Urinary Bladder Neoplasms; Vascular Endothelial Growth Factor A; Wound Healing

2004
Drug development for ovarian hyper-stimulation and anti-cancer treatment: blocking of gonadotropin signaling for epiregulin and amphiregulin biosynthesis.
    Biochemical pharmacology, 2004, Sep-15, Volume: 68, Issue:6

    Gonadotropins play a crucial role in ovarian homeostasis and fertilization through the activation of the cAMP cascade. However, gonadotropin hyper-stimulation may be associated with higher risk for ovarian cancer development. It has been suggested, that high gonadotropin levels in peritoneal and ovarian cystic fluids of patients suffering from benign ovarian cysts, may lead to malignancy. Moreover, we have recently discovered that gonadotropin stimulation can activate the MAPK cascade in target cells. Using DNA microarray technology and RNA from human granulosa cells, we discovered that stimulation with saturating doses of gonadotropins dramatically elevates activity of genes coding for epiregulin and amphiregulin. These gene products can bind and activate the EGF receptor and ERBB4, which are associated with the development of various cancers such as ovarian, breast endometrial and other non-gynecological malignancies. Gonadotropin receptors are expressed not only in the gonads, but also in non-gonadal tissues and in cancer cells. The discovery that gonadotropins activate certain mitogenic signal transduction pathways, may serve as a guide for novel anti-cancer therapy by (1) specific interference at the receptor level to block the gonadotropic response, or arresting the receptor expression and (2) blocking downstream mitogenic signals generated by these hormones, like attenuation of the expression of epiregulin and amphiregulin that belong to the EGF family, using anti-sense and/or SiRNA techniques targeted to suppress their expression. Moreover, since amphiregulin and epiregulin act as mediators of luteinizing hormone (LH) action in the mammalian ovulatory follicles, regulation of the expression of these factors may open new possibilities in treatment of ovarian malfunction implicated with ovarian hyper-stimulation.

    Topics: Amphiregulin; Antineoplastic Agents; Cell Transformation, Neoplastic; Drug Design; EGF Family of Proteins; Epidermal Growth Factor; Epiregulin; Female; Gene Expression; Glycoproteins; Gonadotropins; Humans; Intercellular Signaling Peptides and Proteins; Luteinizing Hormone; Ovarian Neoplasms; Signal Transduction; Transforming Growth Factor alpha

2004
Expression of epidermal growth factor-receptor related protein (ERRP) in human colorectal carcinogenesis.
    Cancer letters, 2004, Sep-30, Volume: 213, Issue:2

    We hypothesize that ERRP (EGFR-related protein), a recently identified negative regulator of EGFR may modulate EGFR function in colorectal carcinogenesis. The expression of ERRP and EGFR in normal and neoplastic colorectal tissue was examined. ERRP was highly expressed in normal colonic mucosa and benign colorectal adenomas, but lower in colorectal cancer. Mean scores for ERRP expression decreased significantly across well differentiated, moderately well differentiated and poorly differentiated (P = 0.002) tumors, respectively. ERRP expression became more attenuated in polyps with increasing grades of dysplasia. In contrast, expression of EGFR was inversely related to ERRP in representative samples of normal and neoplastic tissues.

    Topics: Adenoma; Cell Differentiation; Cell Transformation, Neoplastic; Colon; Colonic Polyps; Colorectal Neoplasms; Epidermal Growth Factor; ErbB Receptors; Gene Expression Regulation, Neoplastic; Glycoproteins; Humans; Oncogene Proteins

2004
Circulating growth factor levels are associated with tumorigenesis in neurofibromatosis type 1.
    Clinical cancer research : an official journal of the American Association for Cancer Research, 2004, Sep-01, Volume: 10, Issue:17

    Neurofibromatosis type 1 (NF1) is characterized by systemic development of neurofibromas. Early clinical diagnosis can be ambiguous, and genetic diagnosis can be prohibitively difficult. Dysregulation of a number of growth factors has been suggested to be a mechanism of pathogenesis. This study was performed to assess the contribution of circulating growth factors for diffuse tumorigenesis and the diagnostic value of circulating growth factor identification in serum.. The growth stimulation of neurofibroma-derived cells by serum from NF1 patients was tested, and serum growth factor levels in a cohort of NF1 patients (n = 39) between the ages of 7 and 70 years were analyzed.. Concentrations of midkine (MK) and stem cell factor, but not epidermal growth factor, were substantially increased in serum of NF1 patients when compared with healthy controls. Within the NF1 group, MK levels increased dramatically at puberty from an average of 0.79 ng/mL in patients <18 years to 1.18 ng/mL in patients >18 years old. Stem cell factor and MK concentrations above a defined threshold in serum of NF1 patients are of diagnostic benefit for 96% of patients in the cohort tested. Furthermore, serum from NF1 patients enhanced proliferation of human neurofibroma-derived primary Schwann cells and endothelial cells substantially better than normal serum.. Enhanced circulating growth factor levels contribute to diffuse tumorigenesis in NF1 and may provide the basis for molecular diagnosis.

    Topics: Adolescent; Adult; Aged; Case-Control Studies; Cell Proliferation; Cell Transformation, Neoplastic; Child; Child, Preschool; Cohort Studies; Cytokines; Endothelial Cells; Epidermal Growth Factor; Female; Gene Expression Regulation, Developmental; Gene Expression Regulation, Neoplastic; Humans; Male; Middle Aged; Midkine; Neurofibromatosis 1; Schwann Cells; Stem Cell Factor

2004
Telomerase prolongs the lifespan of normal human ovarian surface epithelial cells without inducing neoplastic phenotype.
    Journal of the Society for Gynecologic Investigation, 2004, Volume: 11, Issue:8

    The aim of this study was to determine the effects of exogenous expression of the catalytic subunit of telomerase (hTERT) on the lifespan, growth characteristics, and tumorigenicity of normal human ovarian surface epithelial (OSE) cells.. Low-passage primary cultures of normal human OSE cells were transfected with hTERT and the resulting cell lines were characterized.. The ectopic expression of hTERT stabilized the telomeres of the OSE cultures above 8 kb. The hTERT-transfected OSE cell lines grew beyond the normal lifespan seen in OSE cells and propagated in culture for more than 40 passages before senescing. Moreover, the hTERT-transfected cells demonstrated extensive proliferative capacity as evidenced by their ability to continuously grow even when seeded at low dilutions. The morphologic features and normal differentiation patterns seen in normal OSE cells were likewise retained by the hTERT-transfected cells. In addition, the cultures remained responsive to physiologic concentrations of epidermal growth factor and transforming growth factor-beta. Changes associated with neoplastic transformation like anchorage-independent growth, tumorigencity and karyotypic instability were not observed.. We were able to show that the ectopic expression of hTERT in normal human OSE: 1) resulted in cultures with greater growth potential and longer lifespan and 2) did not induce a transformed phenotype previously seen in viral oncogene-transfected OSE cells. The established cell lines would not only provide sufficient material for comprehensive studies to investigate the normal physiology of OSE cells, but could also help in the understanding of the early steps of ovarian carcinogenesis.

    Topics: Animals; Cell Differentiation; Cell Division; Cell Survival; Cell Transformation, Neoplastic; Cells, Cultured; DNA-Binding Proteins; Epidermal Growth Factor; Epithelial Cells; Female; Humans; Immunohistochemistry; Keratins; Mice; Mice, SCID; Ovarian Neoplasms; Ovary; Telomerase; Transfection; Transforming Growth Factor beta; Tumor Cells, Cultured

2004
A Nodal- and ALK4-independent signaling pathway activated by Cripto-1 through Glypican-1 and c-Src.
    Cancer research, 2003, Mar-15, Volume: 63, Issue:6

    Human Cripto-1 (CR-1) is a member of the epidermal growth factor-Cripto FRL1 Cryptic family that has been shown to function as a coreceptor with the type I Activin serine-threonine kinase receptor ALK4 for the transforming growth factor beta-related peptide Nodal. However, CR-1 can also activate the mitogen-activated protein kinase and Akt pathways independently of Nodal and ALK4 by an unknown mechanism. Here, we demonstrate that CR-1 specifically binds to Glypican-1, a membrane-associated heparan sulfate proteoglycan, and activates the tyrosine kinase c-Src, triggering the mitogen-activated protein kinase and Akt signaling pathways. Finally, an active Src kinase is necessary for CR-1 to induce in vitro transformation and migration in mouse mammary epithelial cells.

    Topics: Activin Receptors, Type I; Animals; Anti-HIV Agents; Antibodies, Monoclonal; Antibodies, Monoclonal, Humanized; Cell Line; Cell Movement; Cell Transformation, Neoplastic; Chlorocebus aethiops; COS Cells; Enzyme Activation; Enzyme-Linked Immunosorbent Assay; Epidermal Growth Factor; GPI-Linked Proteins; Heparan Sulfate Proteoglycans; HIV Antibodies; Humans; Intercellular Signaling Peptides and Proteins; Membrane Glycoproteins; Mice; Mitogen-Activated Protein Kinases; Neoplasm Proteins; Nodal Protein; Phosphatidylinositol Diacylglycerol-Lyase; Phosphorylation; Polysaccharide-Lyases; Precipitin Tests; Protein Binding; Protein Serine-Threonine Kinases; Proteins; Proto-Oncogene Proteins; Proto-Oncogene Proteins c-akt; Signal Transduction; src-Family Kinases; Substrate Specificity; Transforming Growth Factor beta; Type C Phospholipases

2003
Epidermal growth factor-induced epithelio-mesenchymal transition in human breast carcinoma cells.
    Laboratory investigation; a journal of technical methods and pathology, 2003, Volume: 83, Issue:3

    PMC42-LA cells display an epithelial phenotype: the cells congregate into pavement epithelial sheets in which E-cadherin and beta-catenin are localized at cell-cell borders. They abundantly express cytokeratins, although 5% to 10% of the cells also express the mesenchymal marker vimentin. Stimulation of PMC42-LA cells with epidermal growth factor (EGF) leads to epithelio-mesenchymal transition-like changes including up-regulation of vimentin and down-regulation of E-cadherin. Vimentin expression is seen in virtually all cells, and this increase is abrogated by treatment of cells with an EGF receptor antagonist. The expression of the mesenchyme-associated extracellular matrix molecules fibronectin and chondroitin sulfate proteoglycan also increase in the presence of EGF. PMC42-LA cells adhere rapidly to collagen I, collagen IV, and laminin-1 substrates and markedly more slowly to fibronectin and vitronectin. EGF increases the speed of cell adhesion to most of these extracellular matrix molecules without altering the order of adhesive preference. EGF also caused a time-dependent increase in the motility of PMC42-LA cells, commensurate with the degree of vimentin staining. The increase in motility was at least partly chemokinetic, because it was evident both with and without chemoattractive stimuli. Although E-cadherin staining at cell-cell junctions disappeared in response to EGF, beta-catenin persisted at the cell periphery. Further analysis revealed that N-cadherin was present at the cell-cell junctions of untreated cells and that expression was increased after EGF treatment. N- and E-cadherin are not usually coexpressed in human carcinoma cell lines but can be coexpressed in embryonic tissues, and this may signify an epithelial cell population prone to epithelio-mesenchymal-like responses.

    Topics: Actins; Breast Neoplasms; Cadherins; Carcinoma; Cell Adhesion; Cell Transformation, Neoplastic; Chemotaxis; Dose-Response Relationship, Drug; Epidermal Growth Factor; Extracellular Matrix Proteins; Keratins; Neoplasm Proteins; Tumor Cells, Cultured; Vimentin; Vinculin

2003
Inhibition of cell transformation by resveratrol and its derivatives: differential effects and mechanisms involved.
    Oncogene, 2003, Apr-10, Volume: 22, Issue:14

    Resveratrol, a constituent of grapes and other foods, has been reported to be a potential cancer chemopreventive agent. Our previous study showed that the antitumor activity of resveratrol occurs through mitogen-activated protein kinases-mediated p53 activation and induction of apoptosis. To develop more effective agents with fewer side effects for the chemoprevention of cancer, we investigated the effect of resveratrol and its structurally related derivatives on epidermal growth factor (EGF)-induced cell transformation. Our results provided the first evidence that one of the resveratrol derivatives exerted a more potent inhibitory effect than resveratrol on EGF-induced cell transformation, but had less cytotoxic effects on normal nontransformed cells. Compared to resveratrol, this compound also caused cell cycle arrest in the G1 phase, but did not induce p53 activation and apoptosis. Furthermore, this compound, but not resveratrol, markedly inhibited EGF-induced phosphatidylinositol-3 kinase (PI-3K) and Akt activation. Collectively, these data suggested that the higher antitumor effect of the compound compared to resveratrol, may act through a different mechanism by mainly targeting PI-3K/Akt signaling pathways.

    Topics: Animals; Anticarcinogenic Agents; Apoptosis; Cell Line; Cell Transformation, Neoplastic; Epidermal Growth Factor; G1 Phase; Gene Expression Regulation; Genes, p53; Mice; Resveratrol; Stilbenes

2003
Molecular characterization of human telomerase reverse transcriptase-immortalized human fibroblasts by gene expression profiling: activation of the epiregulin gene.
    Cancer research, 2003, Apr-15, Volume: 63, Issue:8

    Reconstitution of telomerase activity by ectopic expression of telomerase reverse transcriptase (hTERT) results in an immortal phenotype in various types of normal human cells, including fibroblasts. Despite lack of transformation characteristics, it is unclear whether hTERT-immortalized cells are physiologically and biochemically the same as their normal counterparts. Here, we compared the gene expression profiles of normal and hTERT-immortalized fibroblasts by using a cDNA microarray containing 20,736 cDNA clones and identified 172 dysregulated genes or expressed sequence tags (ESTs). One of the highly expressed genes in the hTERT-immortalized fibroblasts (hTERT-BJ cells) encodes epiregulin, a potent growth factor. Blockade of epiregulin reduced the growth of hTERT-BJ cells and colony formation of hTERT-transformed fibroblasts. Moreover, inhibition of epiregulin function in immortal hTERT-BJ cells triggered a senescence program. Our results suggest that both activation of telomerase and subsequent induction of epiregulin are required for sustained cell proliferation. Given the significant difference in gene expression profiles between normal and hTERT-immortalized fibroblasts and the close relationship between epiregulin and tumorigenesis, we conclude that hTERT-immortalized cells may not replace their normal counterparts for studies of normal cell biology and that the use of hTERT for expansion of normal human cells for therapeutic purposes must be approached with caution.

    Topics: Cell Division; Cell Transformation, Neoplastic; Cellular Senescence; DNA-Binding Proteins; DNA, Complementary; Enzyme Activation; Epidermal Growth Factor; Epiregulin; Fibroblasts; Gene Expression Profiling; Humans; Oligonucleotide Array Sequence Analysis; RNA, Messenger; Telomerase; Up-Regulation

2003
p38 Mitogen-activated protein kinase regulation of JB6 Cl41 cell transformation promoted by epidermal growth factor.
    The Journal of biological chemistry, 2003, Jul-18, Volume: 278, Issue:29

    The relationship between cell transformation and p38 MAP kinase, a major mitogen-activated protein (MAP) kinase pathway converting signals of various extracellular stimuli into expression of specific target genes through activation of transcription factors, still remains unclear. The aim of the present study was to investigate the role of the p38 MAP kinase pathway in epidermal growth factor (EGF)-induced cell transformation in JB6 cells. Our data show that a dominant negative mutant of p38 MAP (DN-p38) kinase inhibits EGF-promoted JB6 Cl41 cell transformation and that SB202190, an inhibitor of p38 MAP kinase, also inhibits JB6 Cl41 cell transformation in a dose-dependent manner. Moreover, our results show that DN-p38 MAP kinase inhibits the phosphorylation of EGF-stimulated activating transcription factor-2 (ATF-2) and signal transducer and activator of transcription 1 (STAT1). Additionally, DN-p38 MAP kinase inhibits EGF-induced phosphorylation of c-Myc (Thr58/Ser62). Gel shift assays indicate that DN-p38 MAP kinase inhibits EGF-induced activator protein-1 (AP-1) DNA binding in a dose-dependent manner. These results show that p38 MAP kinase plays a key role in the regulation of EGF-induced cell transformation in JB6 cells through regulation of phosphorylation of p38 MAP kinase and activation of its target genes in phosphorylation, c-Myc cell transformation-related genes, and AP-1 binding ability.

    Topics: Activating Transcription Factor 2; Animals; Cell Line; Cell Transformation, Neoplastic; Cyclic AMP Response Element-Binding Protein; DNA-Binding Proteins; Enzyme Inhibitors; Epidermal Growth Factor; Imidazoles; JNK Mitogen-Activated Protein Kinases; Mice; Mitogen-Activated Protein Kinases; Models, Biological; Mutagenesis, Site-Directed; Mutation; p38 Mitogen-Activated Protein Kinases; Phosphorylation; Proto-Oncogene Proteins c-myc; Pyridines; Signal Transduction; STAT1 Transcription Factor; Trans-Activators; Transcription Factor AP-1; Transcription Factors

2003
Transcription factor activation and mitogenic synergism in airway smooth muscle cells.
    The European respiratory journal, 2003, Volume: 21, Issue:5

    Simultaneous treatment of human airway smooth muscle (HASM) cells with lysophosphatidic acid (LPA) and epidermal growth factor (EGF) leads to strikingly synergistic stimulation of mitogenesis. The purpose of this study was to explore potential sites for signal integration mediating synergism, focusing on extracellular signal-regulated kinase (ERK) and transcription factors involved in proliferation and inflammation as likely candidates. Activation of ERK was analysed by immunoblotting. Transcription factor activation was assessed using HASM cells transduced with luciferase reporter gene constructs. LPA and EGF both activated ERK but had no synergistic effect when combined. LPA and EGF both activated activator protein (AP)-1, cyclic adenosine monophosphate response element-binding protein, nuclear factor of activated T-cells and the serum response element; however, only AP-1 activation exhibited synergism. Activation of the inhibitory guanine nucleotide-binding protein and of ERK signalling pathways were required for most transcription factor responses to LPA. In contrast, nuclear factor (NF)-kappaB was activated by LPA but not EGF and NF-kappaB activation was completely blocked only when Rho was inhibited. Rapid activation of Rho was observed in response to LPA but not to EGF. Importantly, inhibition of Rho selectively blocked synergism in both AP-1 activation and mitogenesis. In summary, extracellular signal-regulated kinase activation is required for many transcription factor responses to lysophosphatidic acid and epidermal growth factor, however it is not synergistic. Activation of activator protein-1 is synergistic, and Rho activation by lysophosphatidic acid is required for synergism in both activator protein-1 activation and mitogenesis.

    Topics: Acute-Phase Proteins; Botulinum Toxins; Cell Transformation, Neoplastic; Cells, Cultured; Epidermal Growth Factor; Humans; Lysophospholipids; MAP Kinase Signaling System; Mitogens; Myocytes, Smooth Muscle; Trachea; Transcription Factor AP-1; Transcription Factors

2003
Inhibition of caspase-9 through phosphorylation at Thr 125 by ERK MAPK.
    Nature cell biology, 2003, Volume: 5, Issue:7

    Many pro-apoptotic signals activate caspase-9, an initiator protease that activates caspase-3 and downstream caspases to initiate cellular destruction. However, survival signals can impinge on this pathway and suppress apoptosis. Activation of the Ras-Raf-MEK-ERK mitogen-activated protein kinase (MAPK) pathway is associated with protection of cells from apoptosis and inhibition of caspase-3 activation, although the targets are unknown. Here, we show that the ERK MAPK pathway inhibits caspase-9 activity by direct phosphorylation. In mammalian cell extracts, cytochrome c-induced activation of caspases-9 and -3 requires okadaic-acid-sensitive protein phosphatase activity. The opposing protein kinase activity is overcome by treatment with the broad-specificity kinase inhibitor staurosporine or with inhibitors of MEK1/2. Caspase-9 is phosphorylated at Thr 125, a conserved MAPK consensus site targeted by ERK2 in vitro, in a MEK-dependent manner in cells stimulated with epidermal growth factor (EGF) or 12-O-tetradecanoylphorbol-13-acetate (TPA). Phosphorylation at Thr 125 is sufficient to block caspase-9 processing and subsequent caspase-3 activation. We suggest that phosphorylation and inhibition of caspase-9 by ERK promotes cell survival during development and tissue homeostasis. This mechanism may also contribute to tumorigenesis when the ERK MAPK pathway is constitutively activated.

    Topics: 3T3 Cells; Animals; Apoptosis; Base Sequence; Caspase 3; Caspase 9; Caspases; Cell Survival; Cell Transformation, Neoplastic; Cytochrome c Group; Enzyme Inhibitors; Epidermal Growth Factor; Eukaryotic Cells; HeLa Cells; Humans; MAP Kinase Kinase 1; Mice; Mitogen-Activated Protein Kinase Kinases; Mitogen-Activated Protein Kinases; Molecular Sequence Data; Phosphorylation; Protein Serine-Threonine Kinases; Pyridines; Recombinant Fusion Proteins; Signal Transduction; Threonine

2003
Antibody blockade of the Cripto CFC domain suppresses tumor cell growth in vivo.
    The Journal of clinical investigation, 2003, Volume: 112, Issue:4

    Cripto, a cell surface-associated protein belonging to the EGF-CFC family of growth factor-like molecules, is overexpressed in many human solid tumors, including 70-80% of breast and colon tumors, yet how it promotes cell transformation is unclear. During embryogenesis, Cripto complexes with Alk4 via its unique cysteine-rich CFC domain to facilitate signaling by the TGF-beta ligand Nodal. We report, for the first time to our knowledge, that Cripto can directly bind to another TGF-beta ligand, Activin B, and that Cripto overexpression blocks Activin B growth inhibition of breast cancer cells. This result suggests a novel mechanism for antagonizing Activin signaling that could promote tumorigenesis by deregulating growth homeostasis. We show that an anti-CFC domain antibody, A8.G3.5, both disrupts Cripto-Nodal signaling and reverses Cripto blockade of Activin B-induced growth suppression by blocking Cripto's association with either Alk4 or Activin B. In two xenograft models, testicular and colon cancer, A8.G3.5 inhibited tumor cell growth by up to 70%. Both Nodal and Activin B expression was found in the xenograft tumor, suggesting that either ligand could be promoting tumorigenesis. These data validate that functional blockade of Cripto inhibits tumor growth and highlight antibodies that block Cripto signaling mediated through its CFC domain as an important class of antibodies for further therapeutic development.

    Topics: Activin Receptors, Type I; Activins; Animals; Antibodies, Monoclonal; Breast Neoplasms; Cell Division; Cell Separation; Cell Transformation, Neoplastic; CHO Cells; Cricetinae; Dose-Response Relationship, Drug; Epidermal Growth Factor; Epitopes; Flow Cytometry; GPI-Linked Proteins; Humans; Immunoblotting; Immunoglobulin G; Immunohistochemistry; Intercellular Signaling Peptides and Proteins; Ligands; Male; Membrane Glycoproteins; Mice; Mice, Nude; Neoplasm Proteins; Neoplasm Transplantation; Nodal Protein; Plasmids; Precipitin Tests; Protein Binding; Protein Structure, Tertiary; Proteins; Reverse Transcriptase Polymerase Chain Reaction; RNA, Messenger; Signal Transduction; Time Factors; Transfection; Transforming Growth Factor beta; Tumor Cells, Cultured

2003
Involvement of the Akt/mTOR pathway on EGF-induced cell transformation.
    Molecular carcinogenesis, 2003, Volume: 38, Issue:1

    Our previous study demonstrated that phosphatidylinositol 3-kinase (PI3K) is necessary for epidermal growth factor (EGF)-induced cell transformation in mouse epidermal JB6 cells. Akt and the mammalian target of rapamycin (mTOR) are regarded as PI3K downstream effectors. Therefore, in this study, we investigated the role of Akt and mTOR on EGF-induced cell transformation in JB6 cells using rapamycin, a specific mTOR inhibitor, and cells expressing dominant negative mutants of Akt1 (DNM-Akt1). We found that the treatment of cells with rapamycin inhibited EGF-induced cell transformation but only slightly inhibited JB6 cell proliferation at 72 h. Although LY294002, a PI3K inhibitor, attenuated EGF-induced activator protein 1 (AP-1) activation, treatment with rapamycin did not affect AP-1 activity. Treatment with rapamycin inhibited EGF-induced phosphorylation and activation of ribosomal p70 S6 protein kinase (p70 S6K), an mTOR downstream target, but had no effect on phosphorylation and activation of Akt. Rapamycin also had no effect on EGF-induced phosphorylation of extracellular signal-regulated protein kinases (ERKs). We showed that introduction of DNM-Akt1 into JB6 mouse epidermal Cl 41 (JB6 Cl 41) cells inhibits EGF-induced cell transformation without blocking cell proliferation. The expression of DNM-Akt1 also suppressed EGF-induced p70 S6K activation as well as Akt activation. These results indicated an involvement of the Akt/mTOR pathway in EGF-induced cell transformation in JB6 cells.

    Topics: Animals; Antibiotics, Antineoplastic; Cell Transformation, Neoplastic; Epidermal Growth Factor; Mice; Protein Kinases; Protein Serine-Threonine Kinases; Proto-Oncogene Proteins; Proto-Oncogene Proteins c-akt; Ribosomal Protein S6 Kinases, 70-kDa; Sirolimus; TOR Serine-Threonine Kinases

2003
Activated Cdc42 sequesters c-Cbl and prevents EGF receptor degradation.
    Cell, 2003, Sep-19, Volume: 114, Issue:6

    Cdc42 is a Ras-related protein that has been implicated in the control of normal cell growth, and when improperly regulated, in cellular transformation and invasiveness. A variety of extracellular stimuli, including epidermal growth factor (EGF), activate Cdc42. Here, we show that activation of Cdc42 protects the EGF receptor from the negative regulatory activity of the c-Cbl ubiquitin ligase. Activated Cdc42 binds to p85Cool-1 (for cloned-out-of-library)/beta-Pix (for Pak-interactive exchange factor), a protein that directly associates with c-Cbl. This inhibits the binding of Cbl by the EGF receptor and thus prevents Cbl from catalyzing receptor ubiquitination. The role played by Cdc42 in regulating the timing of EGF receptor-Cbl interactions is underscored by the fact that constitutively active Cdc42(F28L), by persistently blocking the binding of Cbl to these receptors, leads to their aberrant accumulation and sustained EGF-stimulated ERK activation, thus resulting in cellular transformation.

    Topics: 3T3 Cells; Animals; cdc42 GTP-Binding Protein; Cell Cycle Proteins; Cell Division; Cell Membrane; Cell Transformation, Neoplastic; Epidermal Growth Factor; ErbB Receptors; Feedback, Physiological; Guanine Nucleotide Exchange Factors; Mice; Mitogen-Activated Protein Kinases; Protein Binding; Proto-Oncogene Proteins; Proto-Oncogene Proteins c-cbl; Rho Guanine Nucleotide Exchange Factors; Ubiquitin-Protein Ligases

2003
Elevated JNK activation contributes to the pathogenesis of human brain tumors.
    Oncogene, 2002, Aug-01, Volume: 21, Issue:33

    The ERK pathway is typically associated with activation of the EGF receptor and has been shown to play a major role in promoting several tumor phenotypes. An analogous signaling module, the JNK pathway, has not been shown to be consistently activated by the EGF receptor but is instead more uniformly stimulated by cellular stresses and cytokines. The function of the JNK pathway in primary tumors is unclear as it has been implicated in both promoting apoptosis and cell growth in vitro, which may be a reflection of the cell lines chosen. Primary human brain tumors frequently show overexpression of the EGF receptor. To clarify the role of JNK in tumorigenesis, we have investigated the role of JNK in a large panel of primary human brain tumors and tumor derived cell lines. Here we present evidence that JNK has a major role in promoting tumorigenesis both in vivo and in vitro. Western blot analysis demonstrated that 86% (18 of 21) of primary brain tumors showed evidence of JNK activation but only 38% (8 of 21) showed evidence of ERK activation. Kinase assays revealed that 77% of brain tumor cell lines activated JNK in response to EGF (7 of 13) or had high levels of basal activity (3 of 13), whereas none of six normal cell lines analysed, including astrocytes, had these properties. Of several growth factors examined, EGF produced the highest level of JNK induction in tumor cell lines and the duration of activation was greater than that seen for ERK. Expression of a dominant-negative (dn) form of JNK potently inhibited EGF mediated anchorage independent growth and protection from cell death in two glial tumor cell lines. These findings demonstrate that enhanced JNK activation is frequently found in primary brain tumors and that this activation contributes to phenotypes related to transformation.

    Topics: Apoptosis; Blotting, Western; Brain Neoplasms; Cell Division; Cell Survival; Cell Transformation, Neoplastic; Contact Inhibition; Enzyme Activation; Epidermal Growth Factor; ErbB Receptors; Humans; JNK Mitogen-Activated Protein Kinases; Mitogen-Activated Protein Kinases; Time Factors; Tumor Cells, Cultured

2002
Effects of ErbB-2 overexpression on mitogenic signalling and cell cycle progression in human breast luminal epithelial cells.
    Oncogene, 2002, Sep-26, Volume: 21, Issue:43

    Most breast cancers arise from luminal epithelial cells and 25-30% of these tumours overexpress the ErbB-2 receptor. Herein, a non-transformed, immortalized cell system was used to investigate the effects of ErbB-2 overexpression in luminal epithelial cells. The phenotypic consequence of ErbB-2 overexpression is a shortening of the G1 phase of the cell cycle and early S phase entry, which leads to hyperproliferation. We show that this effect was mediated through the up-regulation of cdk6 and cyclins D1 and E, and enhanced degradation and relocalization of p27(Kip1). These changes were effected predominantly through enhanced MAPK signalling, resulting in cdk2 hyperactivation. PI3K signalling also participated in cell cycle progression, since PI3K and MAPK coordinately regulated changes in cyclin D1 and cdk6 expression. Cdk4 activity was not required for cell cycle progression in these cells, and was constitutively inhibited through its association with p16(INK4A). MAPK-dependent induction of p21(Cip1) was also necessary for G1 phase progression, although its degradation by the proteasome was required for S phase entry. These data provide new insights into the complex molecular mechanisms underlying mitogenic cell cycle control in luminal epithelial cells, the cell type relevant to primary breast cancer, and show how ErbB-2 overexpression subverts this normal control.

    Topics: Breast; Breast Neoplasms; CDC2-CDC28 Kinases; Cell Cycle; Cell Division; Cell Transformation, Neoplastic; Cyclin D1; Cyclin-Dependent Kinase 2; Cyclin-Dependent Kinase 6; Cyclin-Dependent Kinase Inhibitor p21; Cyclin-Dependent Kinases; Cyclins; Epidermal Growth Factor; Epithelial Cells; Female; Humans; Microfilament Proteins; Mitogen-Activated Protein Kinases; Muscle Proteins; Phosphatidylinositol 3-Kinases; Protein Serine-Threonine Kinases; Proto-Oncogene Proteins c-myc; Receptor, ErbB-2

2002
Epidermal growth factor up-regulates the transcription of mouse lon homology ATP-dependent protease through extracellular signal-regulated protein kinase- and phosphatidylinositol-3-kinase-dependent pathways.
    Experimental cell research, 2002, Oct-15, Volume: 280, Issue:1

    Epidermal growth factor (EGF) induces tumorigenic transformation of mouse epidermal cells (JB6 P(+)). We cloned a full-length EGF-responsive cDNA in JB6P(+) cells; EGF up-regulated mRNA expression of this gene 5- to 6-fold. The deduced amino acid sequence of this cDNA exhibited 84 and 96% homology with human and rat Lon homology ATP-dependent protease, respectively, and all conserved domains of Lon, such as ATPase/protease domains, are present in the mouse gene, indicating that this gene is mouse Lon. EGF increased the transcriptional rate without affecting the mRNA stability of m-Lon. The level of m-Lon in irreversibly transformed mouse epidermal cells (JB7) was 3.4-fold higher than that in parental JB6 P(+) cells. Similarly, human mammary epithelial cells overexpressing the proto-oncogene ErbB2 expressed significantly higher levels of Lon than normal mammary epithelial cells. EGF failed to regulate Lon expression in ERK-deficient JB6 P(-) cells or cells that expressed the dominant-negative p85 P13-K regulatory unit. Furthermore, selective chemical blockers for MEK1 and P13-K (PD98059 and LY294002) inhibited EGF-mediated induction. Mitochondria-localized Lon protease plays a critical role in the regulation of mitochondrial gene expression and genome integrity. Disruption of mitochondrial homeostasis is a general characteristic of tumorigenic transformation. Thus, the role of Lon in tumor promotion warrants further study.

    Topics: Amino Acid Sequence; Animals; ATP-Dependent Proteases; Breast; Cell Line; Cell Line, Transformed; Cell Transformation, Neoplastic; Cloning, Molecular; Conserved Sequence; Epidermal Cells; Epidermal Growth Factor; Epithelial Cells; Female; Heat-Shock Proteins; Humans; MAP Kinase Signaling System; Mice; Mitogen-Activated Protein Kinases; Molecular Sequence Data; Phosphatidylinositol 3-Kinases; Proto-Oncogene Mas; RNA, Messenger; Sequence Alignment; Sequence Homology, Amino Acid; Serine Endopeptidases; Signal Transduction; Transcription, Genetic; Transcriptional Activation; Transformation, Genetic; Up-Regulation

2002
Transforming growth factor-beta and epidermal growth factor synergistically stimulate epithelial to mesenchymal transition (EMT) through a MEK-dependent mechanism in primary cultured pig thyrocytes.
    Journal of cell science, 2002, Nov-15, Volume: 115, Issue:Pt 22

    Enhancement of tumor cell growth and invasiveness by transforming growth factor-beta (TGF-beta) requires constitutive activation of the ras/MAPK pathway. Here we have investigated how MEK activation by epidermal growth factor (EGF) influences the response of fully differentiated and growth-arrested pig thyroid epithelial cells in primary culture to TGF-beta1. The epithelial tightness was maintained after single stimulation with EGF or TGF-beta1 (both 10 ng/ml) for 48 hours. In contrast, co-stimulation abolished the transepithelial resistance and increased the paracellular flux of [(3)H]inulin within 24 hours. Reduced levels of the tight junction proteins claudin-1 and occludin accompanied the loss of barrier function. N-cadherin, expressed only in few cells of untreated or single-stimulated cultures, was at the same time increased 30-fold and co-localised with E-cadherin at adherens junctions in all cells. After 48 hours of co-stimulation, both E- and N-cadherin were downregulated and the cells attained a fibroblast-like morphology and formed multilayers. TGF-beta1 only partially inhibited EGF-induced Erk phosphorylation. The MEK inhibitor U0126 prevented residual Erk phosphorylation and abrogated the synergistic responses to TGF-beta1 and EGF. The observations indicate that concomitant growth factor-induced MEK activation is necessary for TGF-beta1 to convert normal thyroid epithelial cells to a mesenchymal phenotype.

    Topics: Adherens Junctions; Animals; Cadherins; Cell Differentiation; Cell Size; Cell Transformation, Neoplastic; Cells, Cultured; Claudin-1; Down-Regulation; Drug Interactions; Enzyme Inhibitors; Epidermal Growth Factor; Epithelial Cells; MAP Kinase Kinase 1; Membrane Proteins; Mesoderm; Microscopy, Electron; Mitogen-Activated Protein Kinase Kinases; Mitogen-Activated Protein Kinases; Neoplasm Invasiveness; Occludin; Protein Serine-Threonine Kinases; Sus scrofa; Thyroid Gland; Tight Junctions; Transforming Growth Factor beta

2002
Episomally mediated overexpression of wild-type erbB-2 transforms MCF-10A breast epithelial cells.
    Breast cancer research and treatment, 2002, Volume: 76, Issue:3

    MCF-10A are human non-transformed, EGF and insulin dependent breast epithelial cells. The cells were transfected with an episomal pCEP4 vector library containing cDNA from SKBR3 breast carcinoma cells, and selected in media without EGF. After two cycles of expression cloning, morphologically transformed cells appeared. Extracted episomes contained a high proportion of erbB-2 cDNA with wild-type transmembrane domains. Transfection of MCF-10A with individual erbB-2 containing episomes induced significant foci formation in low serum (0.1%) without EGF. MCF-10A sublines expanded from these foci contained a high number of erbB-2 gene copies, highly expressed erbB-2, and lost E-cadherin expression. These results show that if wild-type erbB-2 is sufficiently overexpressed, erbB-2 alone can cause EGF independent transformation of these nonmalignant breast cells. This expression system may be useful for expression cloning in MCF-10A cells, and simulating the effects of high erbB-2 gene amplification in breast epithelial cells.

    Topics: Breast; Breast Neoplasms; Cell Line, Transformed; Cell Transformation, Neoplastic; Clone Cells; DNA, Recombinant; Epidermal Growth Factor; Epithelial Cells; Gene Amplification; Gene Expression Profiling; Genes, erbB-2; Genetic Vectors; Humans; Transfection

2002
Role of GTP-binding proteins in reversing the antiproliferative effects of tocotrienols in preneoplastic mammary epithelial cells.
    Asia Pacific journal of clinical nutrition, 2002, Volume: 11 Suppl 7

    Tocotrienols are a subclass of vitamin E compounds that display potent anticancer activity. Determining the anticancer mechanism of action of tocotrienols will provide essential information necessary for understanding the potential health benefits of these compounds in reducing the risk of breast cancer in women. Epidermal growth factor (EGF) is a potent mitogen for normal and neoplastic mammary epithelial cells. Initial events in EGF-receptor (EGF-R) mitogenic-signalling are G-protein activation, stimulation of adenylyl cyclase and cyclic AMP (cAMP) production. Studies were conducted to determine if the antiproliferative effects of tocotrienols are associated with reduced EGF-induced G-protein and cAMP-dependent mitogenic signalling. Preneoplastic CL-S1 mouse mammary epithelial cells were grown in culture and maintained on serum-free media containing 0-25 micro mol/L tocotrienol-rich fraction of palm oil and/or different doses of pharmacological agents that alter intracellular cAMP levels. Tocotrienol-induced effects on EGF-receptor levels of tyrosine kinase activity, as well as EGF-dependent mitogen-activated pathway kinase (MAPK) and Akt activation, were determined by western blot analysis. Results demonstrate that the antiproliferative effects of tocotrienols in preneoplastic mammary epithelial cells do not reflect a reduction in EGF-receptor mitogenic responsiveness, but rather, result from an inhibition in early post-receptor events involved in cAMP production upstream from EGF-dependent MAPK and phosphoinositide 3-kinase/Akt mitogenic signalling. In summary, these data further characterise the mechanism of action of tocotrienols in suppressing preneoplastic mammary epithelial cell proliferation, and advance the current understanding of the potential health benefits of these compounds in reducing the risk of breast cancer in women.

    Topics: Animals; Anticarcinogenic Agents; Antioxidants; Breast Neoplasms; Cell Division; Cell Transformation, Neoplastic; Cyclic AMP; Dose-Response Relationship, Drug; Epidermal Growth Factor; Epithelial Cells; ErbB Receptors; Female; GTP-Binding Proteins; Humans; Mammary Glands, Animal; Mammary Neoplasms, Animal; Mice; Mice, Inbred BALB C; Mitogen-Activated Protein Kinase 1; Mitogen-Activated Protein Kinase 3; Mitogen-Activated Protein Kinases; Protein-Tyrosine Kinases; Tocotrienols; Tumor Cells, Cultured

2002
Ser-10 phosphorylation of histone H3 and immediate early gene expression in oncogene-transformed mouse fibroblasts.
    Cancer research, 2002, Jan-01, Volume: 62, Issue:1

    Stimulation of the Ras-mitogen-activated protein kinase (MAPK) pathway by growth factors, phorbol esters, and oncoproteins results in the phosphorylation of histone H3. Rsk-2 and MSK1 have been reported to be H3 kinases activated by the Ras-MAPK signal transduction pathway. In this study, we used inhibitors of Rsk-2 and MSK1 to decide which of these kinases was responsible for the 12-O-tetradecanoylphorbol-13-acetate (TPA)-induced phosphorylation of H3 in 10T(1/2) and Ciras-3 (H-ras-transformed 10T(1/2)) mouse fibroblasts. These studies demonstrated that MSK1, but not Rsk-2, was the H3 kinase activated in these cells. Furthermore, assays with Rsk-2 showed that this kinase phosphorylates H2B but not H3 in vitro. H89, a potent MSK1 inhibitor, prevented TPA induction of H3 phosphorylation and diminished the TPA-induced expression of the c-fos and urokinase plasminogen activator genes. We propose that persistent activation of the Ras-MAPK pathway and MSK1 resulting in the elevation of phosphorylated H3 levels may contribute to the aberrant gene expression observed in the oncogene-transformed cells.

    Topics: 3T3 Cells; Animals; Calcium-Calmodulin-Dependent Protein Kinases; Cell Transformation, Neoplastic; Epidermal Growth Factor; Fibroblasts; Gene Expression Regulation; Genes, fos; Genes, Immediate-Early; Histones; MAP Kinase Signaling System; Mice; Phosphorylation; ras Proteins; Ribosomal Protein S6 Kinases; Ribosomal Protein S6 Kinases, 90-kDa; Serine; Tetradecanoylphorbol Acetate; Urokinase-Type Plasminogen Activator

2002
Growth factor requirements and basal phenotype of an immortalized mammary epithelial cell line.
    Cancer research, 2002, Jan-01, Volume: 62, Issue:1

    Carcinogenesis involves a multistep process whereby a normal healthy cell undergoes both immortalization and oncogenesis to become fully transformed. Immortalization results from the subversion of critical cell cycle regulatory checkpoints, thereby allowing a cell to extend its finite life span and to maintain telomeric length. Oncogenesis is the manifestation of additional genetic events that are capable of conferring upon the cell an actual growth advantage. Such an advantage may relieve a cell of its normal requirements for a particular growth factor or may enhance the ability of a cell to proliferate outside of its normal microenvironment. To further investigate this multistep process, we developed an immortalized mammary epithelial cell line by overexpressing the catalytic subunit of telomerase (human telomerase reverse transcriptase) in primary human mammary epithelial cell lines. We present evidence that the overexpression of human telomerase reverse transcriptase was sufficient to extend the life span of the cells and allow for additional events that lead to immortalization. The result was the establishment of an IMEC line. Biochemical analysis of these cells indicates a basal epithelial phenotype with expression of high molecular weight cytokeratins. We show that continued growth of the IMECs is rigorously dependent upon both insulin and epidermal growth factor, and that the mitogenic effects of these factors on the IMECs are mediated in part by AKT. In addition, IMECs express the p53 family member DeltaN-p63-alpha, which is found in basal epithelial cells of many tissues and has been implicated as playing an essential role in normal epithelial development. Our studies suggest that the immortalization of basal epithelial cells of the mammary gland may be an early step in the initiation of a subset of breast cancers with a basal epithelial phenotype.

    Topics: Breast; Breast Neoplasms; Cell Division; Cell Line, Transformed; Cell Transformation, Neoplastic; DNA-Binding Proteins; Epidermal Growth Factor; Epithelial Cells; Genes, Tumor Suppressor; Humans; Insulin; Membrane Proteins; Phenotype; Phosphoproteins; Protein Serine-Threonine Kinases; Proto-Oncogene Proteins; Proto-Oncogene Proteins c-akt; Telomerase; Trans-Activators; Transcription Factors; Tumor Suppressor Proteins

2002
Clonal differences in expression of 25-hydroxyvitamin D(3)-1alpha-hydroxylase, of 25-hydroxyvitamin D(3)-24-hydroxylase, and of the vitamin D receptor in human colon carcinoma cells: effects of epidermal growth factor and 1alpha,25-dihydroxyvitamin D(3).
    Experimental cell research, 2002, Jun-10, Volume: 276, Issue:2

    Human colon carcinoma cells express 25-hydroxyvitamin D(3)-1alpha-hydroxylase (CYP27B1) and thus produce the vitamin D receptor (VDR) ligand 1alpha,25-dihydroxyvitamin D(3) (1,25-D3), which can be metabolized by 25-hydroxyvitamin D(3)-24-hydroxylase (CYP24). Expression of VDR, CYP27B1, and CYP24 determines the efficacy of the antimitotic action of 1,25-D3 and is distinctly related to the degree of differentiation of cancerous lesions. In the present study we addressed the question of whether the effects of epidermal growth factor (EGF) and of 1,25-D3 on VDR, CYP27B1, and CYP24 gene expression in human colon carcinoma cell lines also depend on the degree of cellular differentiation. We were able to show that slowly dividing, highly differentiated Caco-2/15 cells responded in a dose-dependent manner to both EGF and 1,25-D3 by up-regulation of VDR and CYP27B1 expression, whereas in highly proliferative, less differentiated cell lines, such as Caco-2/AQ and COGA-1A and -1E, negative regulation was observed. CYP24 mRNA was inducible in all clones by 1,25-D3 but not by EGF. From the observed clonal differences in the regulatory effects of EGF and 1,25-D3 on VDR and CYP27B1 gene expression we suggest that VDR-mediated growth inhibition by 1,25-D3 would be efficient only in highly differentiated carcinomas even when under mitogenic stimulation by EGF.

    Topics: 25-Hydroxyvitamin D3 1-alpha-Hydroxylase; Calcitriol; Carcinoma; Cell Differentiation; Cell Division; Cell Transformation, Neoplastic; Clone Cells; Colonic Neoplasms; Cytochrome P-450 Enzyme System; Dose-Response Relationship, Drug; Epidermal Growth Factor; Gene Expression Regulation, Neoplastic; Humans; Receptors, Calcitriol; RNA, Messenger; Steroid Hydroxylases; Tumor Cells, Cultured; Vitamin D3 24-Hydroxylase

2002
Ras induces NBT-II epithelial cell scattering through the coordinate activities of Rac and MAPK pathways.
    Journal of cell science, 2002, Jun-15, Volume: 115, Issue:Pt 12

    Cell dissociation and cell migration are the two main components of epithelium-mesenchyme transitions (EMT). We previously demonstrated that Ras is required for the accomplishment of both of these processes during the EGF-induced EMT of the NBT-II rat carcinoma cell line in vitro. In this study, we examined the downstream targets of Ras that are responsible for the dissociation and motility of NBT-II cells. Overexpression of activated forms of c-Raf and MEK1 (a component of the mitogen-activated protein kinase pathway, MAPK) led to cell dissociation, as inferred by the loss of desmosomes from the cell periphery. By contrast, active PI3K, RalA and RalB did not induce desmosome breakdown. The MEK1 inhibitor PD098059 inhibited EGF- and Ras-induced cell dispersion, whereas the PI3K inhibitor LY294002 had no effect. Accordingly, among the partial loss-of-function mutants of Ras (RasV12) that were used to distinguish between downstream targets of Ras, we found that the Raf-specific Ras mutants RasV12S35 and RasV12E38 induced cell dissociation. The PI3K- and RalGDS-activating Ras mutants had, in contrast, no effect on cell dispersion. However, MEK1 was unable to promote cell motility, whereas RasV12S35 and RasV12E38 induced cell migration, suggesting that another Ras effector was responsible for cell motility. We found that the small GTPase Rac is necessary for EGF-mediated cell dispersion since overexpression of a dominant-negative mutant of Rac1 (Rac1N17) inhibited EGF-induced NBT-II cell migration. All stimuli that promoted cell migration also induced Rac activation. Finally, coexpression of active Rac1 and active MEK1 induced the motility of NBT-II cells, suggesting that Ras mediates NBT-II cell scattering through the coordinate activation of Rac and the Raf/MAPK pathway.

    Topics: Animals; Cell Adhesion; Cell Differentiation; Cell Movement; Cell Transformation, Neoplastic; Desmosomes; Endocytosis; Enzyme Inhibitors; Epidermal Growth Factor; Epithelial Cells; Epithelium; Genetic Vectors; MAP Kinase Kinase 1; MAP Kinase Signaling System; Mesoderm; Mitogen-Activated Protein Kinase Kinases; Mutation; Protein Serine-Threonine Kinases; rac GTP-Binding Proteins; ras Proteins; Rats; Receptors, Cell Surface; Tumor Cells, Cultured

2002
Downregulated AP-1 activity is associated with inhibition of Protein-Kinase-C-dependent CD44 and ezrin localisation and upregulation of PKC theta in A431 cells.
    Journal of cell science, 2002, Jul-01, Volume: 115, Issue:Pt 13

    Progression to an invasive, metastatic tumour requires the coordinated expression and function of a number of gene products, as well as their regulation in the context of invasion. The transcription factor AP-1 regulates expression of many of those genes necessary for implementation of the invasion programme. Two such gene products, CD44 and ezrin, are both upregulated in fibroblasts transformed by v-fos and are commonly implicated in cell motility and invasion. Here we report that CD44 and ezrin colocalise to membrane ruffles and microvilli of A431 cells after treatment with EGF. However, A431 cells expressing dominant-negative c-Jun (TAM67), and which as a consequence fail to invade in response to EGF, also fail to correctly localise CD44 and ezrin. CD44 and ezrin are both substrates for Protein Kinase C, and we show that their EGF-dependent colocalisation requires Protein Kinase C activity. Associated with TAM67 expression and disrupted CD44 and ezrin colocalisation is the increased expression and activation of the novel PKC theta isoform. Expression of PKC theta in A431 cells results in the inhibition of cell motility and disrupted localisation of CD44 and ezrin. We propose that AP-1 regulates the integrity of Protein Kinase C signalling and identifies PKC theta as a potential suppressor of the invasion programme.

    Topics: Animals; Cell Compartmentation; Cell Membrane; Cell Transformation, Neoplastic; Cells, Cultured; Cytoskeletal Proteins; Down-Regulation; Enzyme Inhibitors; Epidermal Growth Factor; Fluorescent Antibody Technique; Humans; Hyaluronan Receptors; Isoenzymes; Neoplasm Invasiveness; Neoplasm Metastasis; Neoplasms; Phenotype; Phorbol Esters; Phosphoproteins; Protein Isoforms; Protein Kinase C; Protein Kinase C-theta; Protein Transport; Proto-Oncogene Proteins c-jun; Transcription Factor AP-1; Up-Regulation

2002
ERK5 and ERK2 cooperate to regulate NF-kappaB and cell transformation.
    The Journal of biological chemistry, 2001, Mar-16, Volume: 276, Issue:11

    We have previously demonstrated an involvement of MEK5 and ERK5 in RafBXB-stimulated focus formation in NIH3T3 cells. We find here that MEK5 and ERK5 cooperate with the RafBXB effectors MEK1/2 and ERK1/2 to induce foci. To further understand MEK5-ERK5-dependent signaling, we examined potential MEK5-ERK5 effectors that might influence focus-forming activity. Consistent with results from our focus-formation assays, constitutively active variants of MEK5 and MEK1 synergize to activate NF-kappaB, and MEK5 and ERK5 are required for activation of NF-kappaB by RafBXB. The MEK5-ERK5 pathway is also sufficient to activate both NF-kappaB and p90 ribosomal S6 kinase. Our results support the hypothesis that NF-kappaB and p90 ribosomal S6 kinase are involved in MEK5-ERK5-dependent focus formation and may serve as integration points for ERK5 and ERK1/2 signaling.

    Topics: 3T3 Cells; Animals; Cell Division; Cell Transformation, Neoplastic; Epidermal Growth Factor; Heparin-binding EGF-like Growth Factor; Intercellular Signaling Peptides and Proteins; MAP Kinase Kinase 1; MAP Kinase Kinase 5; Mice; Mitogen-Activated Protein Kinase 1; Mitogen-Activated Protein Kinase 7; Mitogen-Activated Protein Kinase Kinases; Mitogen-Activated Protein Kinases; NF-kappa B; Protein Serine-Threonine Kinases; Proto-Oncogene Proteins c-raf; Response Elements; Ribosomal Protein S6 Kinases; Transcription, Genetic

2001
Inhibition of epidermal growth factor-induced cell transformation and activator protein 1 activation by [6]-gingerol.
    Cancer research, 2001, Feb-01, Volume: 61, Issue:3

    Many spices, including plants of the ginger family, possess anticarcinogenic activity. However, the molecular mechanisms by which they exert their antitumorigenic effects are unknown. Activator protein 1 (AP-1) has a critical role in tumor promotion, and blocking of tumor promoter-induced activation of AP-1 inhibits neoplastic transformation. Epidermal growth factor induces cell transformation and AP-1 activity. The purpose of this study was to investigate the effect of two structurally related compounds of the ginger family, [6]-gingerol and [6]-paradol, on EGF-induced cell transformation and AP-1 activation. Our results provide the first evidence that both block EGF-induced cell transformation but act by different mechanisms.

    Topics: Animals; Anticarcinogenic Agents; Catechols; Cell Death; Cell Line; Cell Transformation, Neoplastic; DNA; Enzyme Induction; Epidermal Cells; Epidermal Growth Factor; Epidermis; Fatty Alcohols; Guaiacol; Ketones; Mice; Mitogen-Activated Protein Kinases; p38 Mitogen-Activated Protein Kinases; Phosphorylation; Plants, Medicinal; Transcription Factor AP-1

2001
Hydrogen peroxide promotes transformation of rat liver non-neoplastic epithelial cells through activation of epidermal growth factor receptor.
    Molecular carcinogenesis, 2001, Volume: 30, Issue:4

    Previous studies demonstrated that hydrogen peroxide (H(2)O(2)) is a tumor promoter in the rat liver epithelial cell line T51B. We investigated the pathway linking H(2)O(2) to tumor promotion. H(2)O(2) can directly induce tyrosine phosphorylation of epidermal growth factor receptor (EGFR). H(2)O(2) and epidermal growth factor exerted similar effects on the induction of early growth response genes, disruption of gap junction communication, triggering of calcium inflow, and promotion of transformation. Furthermore, the effect of H(2)O(2) on tumor promotion was blocked by abrogation of EGFR activation. Our results suggested that tumor promotion by H(2)O(2) is mediated mainly through activation of EGFR in T51B cells.

    Topics: Animals; Apoptosis; Blotting, Northern; Blotting, Western; Calcium; Cell Survival; Cell Transformation, Neoplastic; Colony-Forming Units Assay; Connexins; DNA-Binding Proteins; Early Growth Response Protein 1; Enzyme Activation; Enzyme Inhibitors; Epidermal Growth Factor; Epithelial Cells; ErbB Receptors; Hydrogen Peroxide; Immediate-Early Proteins; Liver; Phosphorylation; Precipitin Tests; Protein-Tyrosine Kinases; Rats; Transcription Factors; Tyrosine

2001
Mutant epidermal growth factor receptor enhances induction of vascular endothelial growth factor by hypoxia and insulin-like growth factor-1 via a PI3 kinase dependent pathway.
    British journal of cancer, 2001, May-18, Volume: 84, Issue:10

    Over-expression of truncated epidermal growth factor receptor (EGFR) occurs in a variety of malignancies including glioblastoma multiforme, breast and lung cancer. The truncation deletes an extracellular domain and results in constitutive activation of the receptor. NIH3T3 cells were transfected with full length or truncated human EGFR and differences in growth rates in vivo and in vitro analysed. A growth advantage was seen for cells expressing mutant receptor compared to full length EGFR in vivo only. Administration of an anti-mutant EGFR antibody to mice transiently reduced the growth rates of mutant tumours, confirming that the mutant receptor itself was important in this enhanced tumorigenicity. This showed that stimuli present in vivo and not in vitro may be contributing to growth. We therefore analysed the regulation of the angiogenic factor vascular endothelial growth factor (VEGF). Although levels of secreted VEGF did not differ significantly between wild-type and mutant EGFR cell lines when grown in vitro under normoxic conditions, following exposure to 0.1% hypoxia levels of VEGF produced by mutant cells increased 3.5-6.6 fold compared to 2 or less for full length EGFR cells. The fold induction was influenced by experimental conditions, including cell confluence and percentage of fetal bovine serum, but was consistently higher for mutant cell lines. The increase in VEGF under hypoxic conditions was blocked by the addition of PI3 kinase inhibitors, indicating that the latter pathway is important in the hypoxic stress response. Basal levels were not affected. Addition of insulin-like growth factor-1 also increased levels of VEGF under normoxic conditions in the mutant cells and no further increase was seen when added to cells exposed to 0.1% oxygen, indicating that levels of VEGF were already maximally stimulated. These results show that the mutant EGFR interacts with other growth factors and hypoxia to regulate VEGF via a PI3 kinase pathway, and suggests a specific role for anti-mutant EGFR antibodies and PI3 kinase inhibitors as therapy of this specific tumour target.

    Topics: 3T3 Cells; Androstadienes; Animals; Cell Division; Cell Hypoxia; Cell Transformation, Neoplastic; Chromones; Endothelial Growth Factors; Enzyme Inhibitors; Epidermal Growth Factor; ErbB Receptors; Gene Expression Regulation; Humans; Insulin-Like Growth Factor I; Lymphokines; Mice; Mice, Nude; Morpholines; Phosphatidylinositol 3-Kinases; Phosphoinositide-3 Kinase Inhibitors; Recombinant Proteins; Sequence Deletion; Transfection; Transplantation, Heterologous; Vascular Endothelial Growth Factor A; Vascular Endothelial Growth Factors; Wortmannin

2001
Inhibition of anchorage-independent growth and lung metastasis of A549 lung carcinoma cells by IkappaBbeta.
    Oncogene, 2001, Apr-26, Volume: 20, Issue:18

    To evaluate the role of the NF-kappaB signaling pathway in oncogenic transformation, we expressed IkappaBbeta, a specific inhibitor of NF-kappaB, in two human lung adenocarcinoma cell lines, A549 and H441. Expression of IkappaBbeta significantly reduced NF-kappaB activation induced by cotransfection with p65/RelA or TNF-alpha and abrogated the basal NF-kappaB activity in A549 cells. Transfection of IkappaBbeta into A549, H441 and K-ras-transformed NIH3T3 cells suppressed anchorage-independent growth as measured by colony formation in soft agar. Anchorage-independent growth of vector-transfected A549 cells in reduced serum could be enhanced by both EGF and IGF-I. In contrast, only EGF but not IGF-I could induce anchorage-independent growth of IkappaBbeta-expressing A549 cells, suggesting that the IGF-I signaling pathway regulating growth and survival may be blocked by IkappaBbeta. Interestingly, expression of IkappaBbeta suppressed growth of A549 cells in low serum in vitro without affecting in vivo growth subcutaneously in nude mice. However, metastatic growth of IkappaBbeta-expressing A549 cells in the lungs of nude mice was significantly inhibited. These results provide evidence that NFkappaB plays an important role in anchorage-independent growth and metastatic growth of lung carcinoma cells.

    Topics: 3T3 Cells; Adenocarcinoma; Animals; Cell Adhesion; Cell Division; Cell Line, Transformed; Cell Transformation, Neoplastic; DNA-Binding Proteins; Epidermal Growth Factor; Humans; I-kappa B Proteins; Lung Neoplasms; Mice; Mice, Nude; NF-kappa B; Signal Transduction; Transcription Factor RelA; Transfection; Transplantation, Heterologous; Tumor Cells, Cultured; Tumor Necrosis Factor-alpha

2001
Omega 3 but not omega 6 fatty acids inhibit AP-1 activity and cell transformation in JB6 cells.
    Proceedings of the National Academy of Sciences of the United States of America, 2001, Jun-19, Volume: 98, Issue:13

    Epidemiological and animal-based investigations have indicated that the development of skin cancer is in part associated with poor dietary practices. Lipid content and subsequently the derived fatty acid composition of the diet are believed to play a major role in the development of tumorigenesis. Omega 3 (omega3) fatty acids, including docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA), can effectively reduce the risk of skin cancer whereas omega 6 (omega6) fatty acids such as arachidonic acid (AA) reportedly promote risk. To investigate the effects of fatty acids on tumorigenesis, we performed experiments to examine the effects of the omega3 fatty acids EPA and DHA and of the omega6 fatty acid AA on phorbol 12-tetradecanoate 13-acetate (TPA)-induced or epidermal growth factor (EGF)-induced transcription activator protein 1 (AP-1) transactivation and on the subsequent cellular transformation in a mouse epidermal JB6 cell model. DHA treatment resulted in marked inhibition of TPA- and EGF-induced cell transformation by inhibiting AP-1 transactivation. EPA treatment also inhibited TPA-induced AP-1 transactivation and cell transformation but had no effect on EGF-induced transformation. AA treatment had no effect on either TPA- or EGF-induced AP-1 transactivation or transformation, but did abrogate the inhibitory effects of DHA on TPA- or EGF-induced AP-1 transactivation and cell transformation in a dose-dependent manner. The results of this study demonstrate that the inhibitory effects of omega3 fatty acids on tumorigenesis are more significant for DHA than for EPA and are related to an inhibition of AP-1. Similarly, because AA abrogates the beneficial effects of DHA, the dietary ratio of omega6 to omega3 fatty acids may be a significant factor in mediating tumor development.

    Topics: Animals; Cell Adhesion; Cell Line; Cell Transformation, Neoplastic; Epidermal Growth Factor; Fatty Acids, Omega-3; Fatty Acids, Omega-6; Fatty Acids, Unsaturated; Genes, Reporter; JNK Mitogen-Activated Protein Kinases; Kinetics; Luciferases; Mice; Mitogen-Activated Protein Kinases; Oligodeoxyribonucleotides; p38 Mitogen-Activated Protein Kinases; Skin; Tetradecanoylphorbol Acetate; Transcription Factor AP-1; Transcriptional Activation; Transfection

2001
Immortalized bovine pancreatic duct cells become tumorigenic after transfection with mutant k-ras.
    Virchows Archiv : an international journal of pathology, 2001, Volume: 438, Issue:6

    Mutation of the K-ras gene is thought to be an early and important event in pancreatic carcinogenesis. In order to study the role of this molecular alteration in the transition from the normal to the neoplastic pancreatic cell, bovine pancreatic duct cells were first immortalized by SV40 large T antigen (Ag) complementary (c)DNA transfection and then transfected with a mutated K-ras gene. As did primary duct cells, the immortalized duct cells (more than 100 passages) expressed cytokeratins, carbonic anhydrase type-II, cystic fibrosis transmembrane conductance regulator (CFTR), and multidrug resistance (mdr). They grew as a single layer after transplantation under plastic domes and formed three-dimensional structures resembling ducts when grown on Matrigel. Cell growth was stimulated by insulin, epidermal growth factor (EGF), transforming growth factor (TGF)-alpha, but cells did not respond to gastrin and CCK-8. They did not form colonies in soft agar nor did they form tumors in nude mice. Immortalized cells transfected with mutated K-ras acquired the ability to form tumors after orthotopic injection into the nude mouse pancreas. It is concluded that SV 40 immortalized bovine pancreatic

    Topics: Animals; Antigens, Polyomavirus Transforming; Biomarkers; Cattle; Cell Division; Cell Line, Transformed; Cell Transformation, Neoplastic; Clone Cells; DNA, Complementary; Epidermal Growth Factor; Epithelial Cells; Fluorescent Antibody Technique, Indirect; Genes, ras; Insulin; Mice; Mice, Nude; Mutation; Pancreatic Ducts; Pancreatic Neoplasms; Polymerase Chain Reaction; RNA, Viral; Transfection; Transforming Growth Factor alpha

2001
Two novel glycosides from the fruits of Morinda citrifolia (noni) inhibit AP-1 transactivation and cell transformation in the mouse epidermal JB6 cell line.
    Cancer research, 2001, Aug-01, Volume: 61, Issue:15

    The fruit juice of Morinda citrifolia (noni), a plant originally grown in the Hawaiian and Tahitian islands, has long been used by islanders to treat diseases, including cancer. Two novel glycosides, 6-O-(beta-D-glucopyranosyl)-1-O-octanoyl-beta-D-glucopyranose and asperulosidic acid, extracted from the juice of noni fruits, were used to examine their effects on 12-O-tedtradecanoylphorbol-13-acetate (TPA)- and epidermal growth factor (EGF)-induced AP-1 transactivation and cell transformation in mouse epidermal JB6 cells. The results indicated that both compounds were effective in suppressing TPA- or EGF-induced cell transformation and associated AP-1 activity. TPA- or EGF-induced phosphorylation of c-Jun, but not extracellular signal-regulated kinases or p38 kinases, was also blocked by the compounds, indicating that c-Jun N-terminal kinases were critical in mediating TPA- or EGF-induced AP-1 activity and subsequent cell transformation in JB6 cells.

    Topics: Animals; Anticarcinogenic Agents; Cell Adhesion; Cell Line; Cell Transformation, Neoplastic; DNA; Epidermal Growth Factor; Fruit; Glucosides; Glycosides; Mice; Mitogen-Activated Protein Kinases; p38 Mitogen-Activated Protein Kinases; Phosphorylation; Plant Extracts; Plants, Medicinal; Proto-Oncogene Proteins c-jun; Rubiaceae; Skin; Skin Neoplasms; Tetradecanoylphorbol Acetate; Transcription Factor AP-1; Transcriptional Activation

2001
A functional screen for genes inducing epidermal growth factor autonomy of human mammary epithelial cells confirms the role of amphiregulin.
    Oncogene, 2001, Jul-05, Volume: 20, Issue:30

    To gain better understanding of the molecular alterations responsible for the aggressive growth potential of epidermal growth factor receptor (EGFR)-positive breast cancers, we utilized an expression cloning strategy to seek gene products that mediate the EGF-independent growth of human breast cancer cells. A retroviral cDNA expression library was constructed from the EGFR-positive SUM-149PT cell line, and transduced into growth factor-dependent human mammary epithelial (HME) cells. Recipient cells were functionally selected for their ability to proliferate in serum-free, EGF-free medium. Library cDNAs were recovered from EGF-independent colonies by PCR amplification or by biological rescue. Clone H55a#1 contained a library insert encoding amphiregulin. This EGFR ligand was able to confer EGF independence when transduced into HME cells. SUM-149PT and H55a#1 cells overexpressed amphiregulin transcripts, and secreted moderate EGF-like activity in conditioned media, indicating a possible autocrine loop. EGFR membrane levels and constitutive phosphorylation were consistent with this hypothesis, as well as the sensitivity of the cells to an ErbB-specific kinase inhibitor. Expression of the WT1 Wilms' tumor suppressor gene, a transcriptional activator of amphiregulin, did not parallel amphiregulin transcript levels, suggesting that another factor regulates amphiregulin in SUM-149PT. Our data confirm the importance of amphiregulin in the etiology of breast cancer.

    Topics: Amphiregulin; Breast; Breast Neoplasms; Cell Division; Cell Transformation, Neoplastic; Cells, Cultured; Clone Cells; Culture Media, Conditioned; Culture Media, Serum-Free; DNA, Complementary; EGF Family of Proteins; Epidermal Growth Factor; Epithelial Cells; ErbB Receptors; Female; Gene Library; Genes, Wilms Tumor; Genetic Complementation Test; Genetic Techniques; Genetic Vectors; Glycoproteins; Growth Substances; Humans; Intercellular Signaling Peptides and Proteins; Neoplasm Proteins; Neoplastic Stem Cells; Phenotype; Retroviridae; Transfection; Tumor Cells, Cultured

2001
The role of alpha 6 beta 1 integrin and EGF in normal and malignant acinar morphogenesis of human prostatic epithelial cells.
    Mutation research, 2001, Sep-01, Volume: 480-481

    Complex multiple interactions between cells and extracellular matrix occur during acinar morphogenesis involving integrin receptors and growth factors. Changes in these interactions occur during carcinogenesis as cells progress from a normal to a malignant, invasive phenotype. We have developed human prostatic epithelial cell lines of the same lineage, which represent multiple steps in carcinogenesis, similar to prostatic intraepithelial neoplasia and subsequent tumor progression. The non-tumorigenic, RWPE-1 and the tumorigenic WPE1-NB27 and WPE1-NB26 cell lines were used to examine their ability to undergo acinar morphogenesis in a 3-D cell culture model and its relationship to invasion, integrin expression and EGF presence. An inverse relationship between the degree of acinar formation and invasive ability was observed. The non-tumorigenic, non-invasive RWPE-1 and the low tumorigenic, low invasive, WPE1-NB27 cells show high and decreased acinar forming ability, respectively, while the more invasive WPE1-NB26 cells show a loss of acinar formation. While RWPE-1 acini show basal expression of alpha 6 beta 1 integrin, which correlates with their ability to polarize and form acini, WPE1-NB27 cells lack alpha 6 but show basal, but weaker expression of beta 1 integrin. WPE1-NB26 cells show loss alpha 6 and abnormal, diffused beta 1 integrin expression. A dose-dependent decrease in acinar formation was observed in RWPE-1 cells when cell proliferation was induced by EGF. Anti-functional antibody to EGF caused an increase in acinar formation in RWPE-1 cells. These results suggest that malignant cells lose the ability to undergo acinar morphogenesis and that the degree of this loss appears to be related to invasive ability, EGF levels and alterations in laminin-specific integrin expression. This model system mimics different steps in prostate carcinogenesis and has applications in the secondary and tertiary prevention of prostate cancer.

    Topics: Antibodies; Cell Division; Cell Line; Cell Transformation, Neoplastic; Collagen; Disease Progression; Dose-Response Relationship, Drug; Drug Combinations; Epidermal Growth Factor; Epithelial Cells; Humans; Integrin alpha6beta1; Integrins; Laminin; Male; Morphogenesis; Neoplasm Invasiveness; Precancerous Conditions; Prostate; Prostatic Intraepithelial Neoplasia; Prostatic Neoplasms; Proteoglycans

2001
Mig-6 is a negative regulator of the epidermal growth factor receptor signal.
    Biological chemistry, 2001, Volume: 382, Issue:12

    In contrast to signal generation and transmission, the mechanisms and molecules that negatively regulate receptor tyrosine kinase (RTK) signaling are poorly understood. Here we characterize Mig-6 as a novel negative feedback regulator of the epidermal growth factor receptor (EGFR) and potential tumor suppressor. Mig-6 was identified in a yeast two-hybrid screen with the kinase active domain of the EGFR as bait. Upon EGF stimulation Mig-6 binds to the EGFR involving a highly acidic region between amino acids 985-995. This interaction is kinase activity-dependent, but independent of tyrosine 992. Mig-6 overexpression results in reduced activation of the mitogenactivated protein kinase ERK2 in response to EGF, but not FGF or PDGF, stimulation and in enhanced receptor internalization without affecting the rate of degradation. The induction of Mig-6 mRNA expression in response to EGF, but not FGF, indicates the existence of a negative regulatory feedback loop. Consistent with these findings, a possible role as tumor suppressor is indicated by Mig-6-mediated inhibition of EGFR overexpression-induced transformation of Rati cells.

    Topics: Amino Acid Sequence; Animals; Blotting, Northern; Carrier Proteins; Cell Transformation, Neoplastic; Chlorocebus aethiops; COS Cells; Down-Regulation; Epidermal Growth Factor; ErbB Receptors; Intracellular Signaling Peptides and Proteins; Mitogen-Activated Protein Kinases; Molecular Sequence Data; Phosphoamino Acids; Precipitin Tests; Rats; RNA, Messenger; Sequence Homology, Amino Acid; Signal Transduction; Tumor Cells, Cultured; Two-Hybrid System Techniques

2001
Phospholipase D and RalA cooperate with the epidermal growth factor receptor to transform 3Y1 rat fibroblasts.
    Molecular and cellular biology, 2000, Volume: 20, Issue:2

    3Y1 rat fibroblasts overexpressing the epidermal growth factor (EGF) receptor (EGFR cells) become transformed when treated with EGF. A common response to oncogenic and mitogenic stimuli is elevated phospholipase D (PLD) activity. RalA, a small GTPase that functions as a downstream effector molecule of Ras, exists in a complex with PLD1. In the EGFR cells, EGF induced a Ras-dependent activation of RalA. The activation of PLD by EGF in these cells was dependent upon both Ras and RalA. In contrast, EGF-induced activation of Erk1, Erk2, and Jun kinase was dependent on Ras but independent of RalA, indicating divergent pathways activated by EGF and mediated by Ras. The transformed phenotype induced by EGF in the EGFR cells was dependent upon both Ras and RalA. Importantly, overexpression of wild-type RalA or an activated RalA mutant increased PLD activity in the absence of EGF and transformed the EGFR cells. Although overexpression of PLD1 is generally toxic to cells, the EGFR cells not only tolerated PLD1 overexpression but also became transformed in the absence of EGF. These data demonstrate that either RalA or PLD1 can cooperate with EGF receptor to transform cells.

    Topics: Animals; Cell Division; Cell Line; Cell Transformation, Neoplastic; Enzyme Activation; Epidermal Growth Factor; ErbB Receptors; Fibroblasts; Gene Expression; Genes, Dominant; GTP Phosphohydrolases; JNK Mitogen-Activated Protein Kinases; Mitogen-Activated Protein Kinases; Mutation; Phenotype; Phospholipase D; ral GTP-Binding Proteins; ras Proteins; Rats; Signal Transduction; Transfection

2000
Role of TGF-beta in EGF-induced transformation of NRK cells is sustaining high-level EGF-signaling.
    FEBS letters, 2000, Jan-21, Volume: 466, Issue:1

    We have been isolating and analyzing NRK cell mutants, which fail to transform by epidermal growth factor (EGF) and transforming growth factor (TGF)-beta. One such mutant, R14, can respond to the growth inhibitory signal of TGF-beta to the same extent as parental NRK but fail to respond to the growth stimulatory signal of EGF. This mutant has a defect in EGF receptor (EGFR) expression. When R14 mutant expressed a high level of EGFR, however, EGF not only induced proliferation in this mutant but also induced transformation without the aid of TGF-beta. These findings suggest that the major role of TGF-beta in this transformation system should be to counteract the ligand-dependent down-regulation of EGFR, thereby sustaining high-level EGF-signaling.

    Topics: Animals; Base Sequence; Cell Line; Cell Transformation, Neoplastic; DNA Primers; Epidermal Growth Factor; ErbB Receptors; Gene Expression; Molecular Sequence Data; Mutation; Promoter Regions, Genetic; Rats; Signal Transduction; Transforming Growth Factor beta

2000
Annexin 1 expression and phosphorylation are upregulated during liver regeneration and transformation in antithrombin III SV40 T large antigen transgenic mice.
    Hepatology (Baltimore, Md.), 2000, Volume: 31, Issue:2

    We have used a transgenic animal model, which constitutively develops hepatocarcinoma (Antithrombin III SV40 T large Antigen: ASV), to study the involvement of Annexin 1 (ANX1) in liver regeneration and malignant transformation. Primary hepatocytes isolated from normal mice did not express ANX1. In contrast, ANX1 was strongly expressed in hepatocytes of transgenic mice during constitutive development of hepatocarcinoma. In ASV transgenic mice, an elevated ANX1 level preceded the appearance of the tumor, indicating that it could be a good marker in the diagnosis of cancer. One-third hepatectomy in normal mice resulted in stimulation of ANX1 synthesis and phosphorylation. This upregulation correlated with increased synthesis of EGF and consequently with increased phosphorylation of the EGF receptor (EGF-R). Stable transfection of a hepatocyte cell line derived from ASV transgenic mice (mhAT2) with antisense complementary DNA for ANX1 reduced the proliferation rate as well as cytosolic phospholipase A(2) (cPLA(2)) activity. Thus, ANX1 expression and phosphorylation could be a factor implicated in liver regeneration and tumorigenesis, either through modulation of cPLA(2) activity or EGF-R function.

    Topics: Animals; Annexin A1; Antigens, Polyomavirus Transforming; Antithrombin III; Carcinoma, Hepatocellular; Cell Transformation, Neoplastic; Cells, Cultured; Epidermal Growth Factor; Hepatectomy; Liver Neoplasms; Liver Regeneration; Mice; Mice, Inbred Strains; Mice, Transgenic; Phosphorylation; Postoperative Period; Promoter Regions, Genetic; Up-Regulation

2000
Rap1-suppressed tumorigenesis is concomitant with the interference in ras effector signaling.
    FEBS letters, 2000, Feb-11, Volume: 467, Issue:2-3

    Expression of Rap1 blocks epithelial growth factor-induced extracellular signal-regulated kinases (ERKs) activation. However, recent studies demonstrated that Rap1 mediates ERKs activation induced by nerve growth factor. The anti-oncogenic effect of Rap1 has been reported but its mechanism remains unclear. To evaluate the correlation between the anti-transforming effect and the activation of ERKs, we transfected rap1 cDNA into Hep3B cells and selected stable transfectants. The Rap1 transfectants completely lost their intrinsic tumorigenicity in Balb/c nude mice. Both insulin and 12-O-tetradecanoyl phorbol-13-acetate (TPA)-stimulated ERK activations were also blocked. Our findings suggest that Rap1-suppressed tumorigenicity is concomitant with ERKs inhibition.

    Topics: Animals; Cell Division; Cell Transformation, Neoplastic; Enzyme Activation; Epidermal Growth Factor; Humans; Insulin; Mice; Mice, Inbred BALB C; Mice, Nude; Mitogen-Activated Protein Kinases; Nerve Growth Factor; rap1 GTP-Binding Proteins; Tetradecanoylphorbol Acetate; Transfection; Tumor Cells, Cultured

2000
Oncogenic epidermal growth factor receptor mutants with tandem duplication: gene structure and effects on receptor function.
    Oncogene, 2000, Feb-10, Volume: 19, Issue:6

    A number of epidermal growth factor receptor (EGFR) deletion mutants have been identified in gliomas, in which the EGFR gene is frequently amplified and rearranged. We have previously characterized the structure of a gene in A-172 human glioma cells that encodes a 190-kDa EGFR mutant with tandem duplication of the tyrosine kinase (TK) and calcium-mediated internalization (CAIN) domains. Here we describe a 185-kDa tandem duplication mutant (TDM) that is expressed in KE and A-1235 glioma cells, along with certain functional characteristics of the mutants. The corresponding transcripts in KE and A-1235 cells contain 1053 additional nucleotides representing an in-frame duplication of exons 18 through 25 which encode the entire TK region and a portion of the CAIN domain. As with duplication of the entire TK/CAIN region (exons 18-26) in A-172 cells, duplication of exons 18-25 is associated with a specific genomic rearrangement between flanking introns. Involved introns contain homology to recombination signal sequence (RSS) heptamers present in the V(D)J region of the T lymphocyte receptor gene. In defined medium, both oncogenic TDM are constitutively autophosphorylated and inefficiently downregulated. High-affinity binding is reduced in EGFR.TDM/18-26, although the t1/2 of receptor internalization is not prolonged.

    Topics: Animals; Biopsy; Brain Neoplasms; Cell Transformation, Neoplastic; Epidermal Growth Factor; ErbB Receptors; Exons; Fibroblasts; Gene Duplication; Gene Expression Regulation, Neoplastic; Glioma; Humans; Introns; Mice; Molecular Weight; Phosphorylation; Protein Processing, Post-Translational; Protein Structure, Tertiary; Radioligand Assay; Reverse Transcriptase Polymerase Chain Reaction; RNA, Messenger; Sequence Homology, Nucleic Acid; Tumor Cells, Cultured

2000
Epidermal growth factor-stimulated tyrosine phosphorylation of caveolin-1. Enhanced caveolin-1 tyrosine phosphorylation following aberrant epidermal growth factor receptor status.
    The Journal of biological chemistry, 2000, Mar-17, Volume: 275, Issue:11

    Caveolin-1 is the major coat protein of caveolae and has been reported to interact with various intracellular signaling molecules including the epidermal growth factor (EGF) receptor. To investigate the involvement of caveolin-1 in EGF receptor action, we used mouse B82L fibroblasts transfected with (a) wild type EGF receptor, (b) a C-terminally truncated EGF receptor at residue 1022, (c) a C-terminally truncated EGF receptor at residue 973, or (d) a kinase-inactive EGF receptor (K721M). Following EGF treatment, there was a distinct electrophoretic mobility shift of the caveolin-1 present in cells expressing the truncated forms of the EGF receptor, but this shift was not detectable in cells bearing either normal levels of the wild type EGF receptor or a kinase-inactive receptor. This mobility shift was also not observed following the addition of other cell stimuli, such as platelet-derived growth factor, insulin, basic fibroblast growth factor, or phorbol 12-myristate 13-acetate. Analysis of caveolin-1 immunoprecipitates from EGF-stimulated or nonstimulated cells demonstrated that the EGF-induced mobility shift of caveolin-1 was associated with its tyrosine phosphorylation in cells expressing truncated EGF receptors. Maximal caveolin-1 phosphorylation was achieved within 5 min after exposure to 10 nM EGF and remained elevated for at least 2 h. Additionally, several distinct phosphotyrosine-containing proteins (60, 45, 29, 24, and 20 kDa) were co-immunoprecipitated with caveolin-1 in an EGF-dependent manner. Furthermore, the Src family kinase inhibitor, PP1, does not affect autophosphorylation of the receptor, but it does inhibit the EGF-induced mobility shift and phosphorylation of caveolin-1. Conversely, the MEK inhibitors PD98059 and UO126 could attenuate EGF-induced mitogen-activated protein kinase activation, they do not affect the EGF-induced mobility shift of caveolin-1. Because truncation and overexpression of the EGF receptor have been linked to cell transformation, these results provide the first evidence that the tyrosine phosphorylation of caveolin-1 occurs via an EGF-sensitive signaling pathway that can be potentiated by an aberrant activity or expression of various forms of the EGF receptor.

    Topics: Animals; Caveolin 1; Caveolins; Cell Transformation, Neoplastic; Cells, Cultured; Dose-Response Relationship, Drug; Epidermal Growth Factor; ErbB Receptors; Flavonoids; Membrane Proteins; Mice; Mitogen-Activated Protein Kinase Kinases; Mutation; Phosphorylation; Protein Isoforms; Pyrazoles; Pyrimidines; Recombinant Proteins; Signal Transduction; src-Family Kinases; Tyrosine

2000
Cross-talk between epidermal growth factor receptor and c-Met signal pathways in transformed cells.
    The Journal of biological chemistry, 2000, Mar-24, Volume: 275, Issue:12

    In rat liver epithelial cells constitutively expressing transforming growth factor alpha (TGFalpha), c-Met is constitutively phosphorylated in the absence of its ligand, hepatocyte growth factor. We proposed that TGFalpha and the autocrine activation of its receptor, epidermal growth factor receptor (EGFR), leads to phosphorylation and activation of c-Met. We found that there is constitutive c-Met phosphorylation in human hepatoma cell lines and the human epidermoid carcinoma cell line, A431 which express TGFalpha, but not in normal human hepatocytes. Constitutive c-Met phosphorylation in A431, HepG2, AKN-1, and HuH6 cells was inhibited by neutralizing antibodies against TGFalpha and/or EGFR. Exposure to exogenous TGFalpha or EGF increased the phosphorylation of c-Met in the human epidermoid carcinoma cell line, A431. The increase of c-Met phosphorylation by TGFalpha in A431 cells was inhibited by neutralizing antibodies against TGFalpha and/or EGFR and by the EGFR-specific inhibitor tyrphostin AG1478. These results indicate that constitutive c-Met phosphorylation, and the increase of c-Met phosphorylation by TGFalpha or EGF, in tumor cell lines is the result of the activation via EGFR. We found that c-Met in tumor cells co-immunoprecipitates with EGFR regardless of the existence of their ligands in tumor cells, but not in normal human hepatocytes. We conclude that c-Met associates with EGFR in tumor cells, and this association facilitates the phosphorylation of c-Met in the absence of hepatocyte growth factor. This cross-talk between c-Met and EGFR may have significant implications for altered growth control in tumorigenesis.

    Topics: Animals; Carcinoma, Hepatocellular; Carcinoma, Squamous Cell; Cell Transformation, Neoplastic; Epidermal Growth Factor; ErbB Receptors; Hepatocyte Growth Factor; Humans; Liver Neoplasms; Phosphorylation; Protein Binding; Proto-Oncogene Proteins c-met; Quinazolines; Rats; Receptor Cross-Talk; Transforming Growth Factor alpha; Tumor Cells, Cultured; Tyrphostins

2000
Epidermal growth factor receptor expression in neurofibromatosis type 1-related tumors and NF1 animal models.
    The Journal of clinical investigation, 2000, Volume: 105, Issue:9

    We have found that EGF-R expression is associated with the development of the Schwann cell-derived tumors characteristic of neurofibromatosis type 1 (NF1) and in animal models of this disease. This is surprising, because Schwann cells normally lack EGF-R and respond to ligands other than EGF. Nevertheless, immunoblotting, Northern analysis, and immunohistochemistry revealed that each of 3 malignant peripheral nerve sheath tumor (MPNST) cell lines from NF1 patients expressed the EGF-R, as did 7 of 7 other primary MPNSTs, a non-NF1 MPNST cell line, and the S100(+) cells from each of 9 benign neurofibromas. Furthermore, transformed derivatives of Schwann cells from NF1(-/-) mouse embryos also expressed the EGF-R. All of the cells or cell lines expressing EGF-R responded to EGF by activation of downstream signaling pathways. Thus, EGF-R expression may play an important role in NF1 tumorigenesis and Schwann cell transformation. Consistent with this hypothesis, growth of NF1 MPNST lines and the transformed NF1(-/-) mouse embryo Schwann cells was greatly stimulated by EGF in vitro and could be blocked by agents that antagonize EGF-R function.

    Topics: Animals; Cell Transformation, Neoplastic; Epidermal Growth Factor; ErbB Receptors; Humans; Mice; Mice, Mutant Strains; Neurilemmoma; Neurofibromatosis 1; Neurofibromin 1; Proteins; Rats; Tumor Cells, Cultured

2000
RAS transformation causes sustained activation of epidermal growth factor receptor and elevation of mitogen-activated protein kinase in human mammary epithelial cells.
    International journal of cancer, 2000, Oct-01, Volume: 88, Issue:1

    Activation of the ras oncogene is an important step in carcinogenesis. Human MCF-10A mammary epithelial cells were transformed with a point-mutated form of the Ha-ras oncogene. Epidermal growth factor receptor (EGFR) phosphorylation levels were chronically elevated after EGF induction and the EGFR ligand-driven internalization rate was slower in Ha-ras transformed MCF-10A cells. Additionally, basal levels of p42/44 mitogen-activated protein kinase (MAPK) expression and enzyme activity were significantly higher in Ha-ras transformed cells, localized predominantly in the nucleus. The anti-EGFR monoclonal antibody (MAb) 225 and the EGFR tyrosine kinase inhibitor PD153035 blocked anchorage-independent growth of Ha-ras transformed cells in soft agar and were more effective when used in combination. The MEK inhibitor PD98059 and anti-erbB-2 MAb L26 also suppressed colony formation of Ha-ras transformed cells in soft agar. Therefore, Ha-ras transformation leads to an augmentation in signaling through the EGFR as a result of an increase in ligand-dependent phosphorylation, a decrease in its internalization and an up-regulation in basal p44/42 MAPK levels. These effects may contribute to uncontrolled growth of Ha-ras-transformed human mammary epithelial cells.

    Topics: Antibodies, Monoclonal; Antibody Specificity; Breast; Cell Line, Transformed; Cell Transformation, Neoplastic; Cells, Cultured; Enzyme Activation; Enzyme Inhibitors; Epidermal Growth Factor; ErbB Receptors; Female; Gene Expression Regulation; Genes, ras; Growth Inhibitors; Humans; MAP Kinase Signaling System; Mitogen-Activated Protein Kinase 1; Mitogen-Activated Protein Kinase 3; Mitogen-Activated Protein Kinases; Phosphorylation; Point Mutation; Proto-Oncogene Proteins c-raf; Proto-Oncogene Proteins p21(ras); Subcellular Fractions; Substrate Specificity; Transfection

2000
Expression of a restrictive receptor for interleukin 13 is associated with glial transformation.
    Journal of neuro-oncology, 2000, Volume: 48, Issue:2

    We have previously documented that the vast majority of high-grade gliomas over-express binding sites for interleukin 13 (IL13) in situ. We now extend this analysis to evaluate the distribution of the binding of IL13 among other brain tumors. Tumor specimens from patients with low-grade gliomas, oligodendrogliomas, ependymomas, pilocytic astrocytomas, gliosarcomas, medulloblastomas, meningiomas, and metastases to the brain were analyzed and compared to a new series of glioblastoma multiforme (GBM) samples. Serial tumor tissue sections were incubated with 125I-labeled (i) IL13, (ii) antibody against transferrin (Tf) receptor, and (iii) epidermal growth factor (EGF). Most (17/18) GBMs stained specifically for IL13 binding sites while sections from 3/11 low-grade gliomas, 5/5 high-grade gliomas (grade III), 3/5 oligodendrogliomas (all three were anaplastic), and 1/2 gliosarcomas also showed specific binding for IL13. We did not detect IL13 binding sites in medulloblastomas (0/4) and found them only in 2/20 meningiomas. Metastases to the brain (4/12, i.e., lung adenocarcinomas and renal cell carcinoma) showed some binding of 125I-IL13. The presence of receptors for Tf was ubiquitous among all studied tumors while EGF receptor expression was much more variable. Since it appears that primarily the least differentiated forms of gliomas possess IL13 binding sites in abundance, it is plausible that IL 13 receptor expressed in low-grade gliomas might be a prognostically significant marker associated with their progression to high-grade gliomas. Finally, we demonstrate that the glioma-associated IL13 receptor is truly more restrictive in nature also due to its selective representation among brain tumors of glial origin.

    Topics: Adenocarcinoma; Biomarkers, Tumor; Brain Neoplasms; Carcinoma; Cell Transformation, Neoplastic; Disease Progression; Ependymoma; Epidermal Growth Factor; ErbB Receptors; Gene Expression Regulation, Neoplastic; Glioma; Gliosarcoma; Humans; Interleukin-13; Interleukin-13 Receptor alpha1 Subunit; Interleukin-4; Medulloblastoma; Meningeal Neoplasms; Meningioma; Neoplasm Proteins; Oligodendroglioma; Receptors, Interleukin; Receptors, Interleukin-13; Receptors, Transferrin; Recombinant Proteins; Substrate Specificity

2000
The Jun kinase 2 isoform is preferentially required for epidermal growth factor-induced transformation of human A549 lung carcinoma cells.
    Molecular and cellular biology, 1999, Volume: 19, Issue:3

    We have previously found that epidermal growth factor (EGF) mediates growth through the Jun N-terminal kinase/stress-activated kinase (JNK/SAPK) pathway in A549 human lung carcinoma cells. As observed here, EGF treatment also greatly enhances the tumorigenicity of A549 cells, suggesting an important role for JNK in cancer cell growth (F. Bost, R. McKay, N. Dean, and D. Mercola, J. Biol. Chem. 272:33422-33429, 1997). Several isoforms families of JNK, JNK1, JNK2, and JNK3, have been isolated; they arise from alternative splicing of three different genes and have distinct substrate binding properties. Here we have used specific phosphorothioate oligonucleotides targeted against the two major isoforms, JNK1 and JNK2, to discriminate their roles in EGF-induced transformation. Multiple antisense sequences have been screened, and two high-affinity and specific candidates have been identified. Antisense JNK1 eliminated steady-state mRNA and JNK1 protein expression with a 50% effective concentration (EC50) of <0.1 microM but did not alter JNK2 mRNA or protein levels. Conversely, antisense JNK2 specifically eliminated JNK2 steady-state mRNA and protein expression with an EC50 of 0.1 microM. Antisense JNK1 and antisense JNK2 inhibited by 40 and 70%, respectively, EGF-induced total JNK activity, whereas sense and scrambled-sequence control oligonucleotides had no effect. The elimination of mRNA, protein, and JNK activities lasted 48 and 72 h following a single Lipofectin treatment with antisense JNK1 and JNK2, respectively, indicating sufficient duration for examining the impact of specific elimination on the phenotype. Direct proliferation assays demonstrated that antisense JNK2 inhibited EGF-induced doubling of growth as well as the combination of active antisense oligonucleotides did. EGF treatment also induced colony formation in soft agar. This effect was completely inhibited by antisense JNK2 and combined-antisense treatment but not altered by antisense JNK1 alone. These results show that EGF doubles the proliferation (growth in soft agar as well as tumorigenicity in athymic mice) of A549 lung carcinoma cells and that the JNK2 isoform but not JNK1 is utilized for mediating the effects of EGF. This study represents the first demonstration of a cellular phenotype regulated by a JNK isoform family, JNK2.

    Topics: Agar; Animals; Calcium-Calmodulin-Dependent Protein Kinases; Cell Division; Cell Transformation, Neoplastic; Epidermal Growth Factor; Female; Gene Expression; Humans; Isoenzymes; JNK Mitogen-Activated Protein Kinases; Mice; Mice, Nude; Mitogen-Activated Protein Kinase 9; Mitogen-Activated Protein Kinases; Oligonucleotides; Oligonucleotides, Antisense; Protein Kinases; RNA, Antisense; RNA, Messenger; Time Factors; Tumor Cells, Cultured; Ultraviolet Rays

1999
Malignant transformation of p53-deficient astrocytes is modulated by environmental cues in vitro.
    Cell growth & differentiation : the molecular biology journal of the American Association for Cancer Research, 1999, Volume: 10, Issue:2

    The early incidence of p53 mutation in astrocytomas suggests that it plays an important role in astrocyte transformation. Astrocytes isolated from homozygous p53 knockout mice grow rapidly, lack contact inhibition, and are immortal. Here we tested whether the loss of p53 is sufficient for progression to tumorigenicity of astrocytes. We grew primary astrocytes under three conditions for over 120 population doublings and assessed their antigenic phenotype, chromosome number, and expression of glioma-associated genes as well as their ability to form colonies in soft agarose and tumors s.c. and intracranially in nude mice. Under two conditions (10% FCS and 0.5% FCS plus 20 ng/ml EGF), cells acquired the ability to form colonies in soft agarose and tumors in nude mice, and this was accompanied by the expression of genes, including epidermal growth factor receptor, platelet-derived growth factor receptor alpha and beta, protein kinase Cdelta, and vascular endothelial growth factor, which are known to be aberrantly regulated in human astrocytomas. Under the third condition (0.5% FCS plus 10 ng/ml basic fibroblast growth factor), astrocytes gained the ability to form colonies in soft agarose and had abnormal chromosome numbers similar to cells in the first two conditions but did not form tumors in nude mice or overexpress glioma-associated genes. These data provide experimental evidence for the idea that the malignant progression initiated by the loss of p53 may be subject to modulation by extracellular environmental influences.

    Topics: Aneuploidy; Animals; Astrocytes; Blotting, Northern; Brain; Brain Neoplasms; Carcinogenicity Tests; Cell Culture Techniques; Cell Division; Cell Transformation, Neoplastic; Cells, Cultured; Culture Media; Epidermal Growth Factor; ErbB Receptors; Fibroblast Growth Factor 2; Glioma; Humans; Mice; Mice, Nude; Receptors, Platelet-Derived Growth Factor; Time Factors; Tumor Stem Cell Assay; Tumor Suppressor Protein p53

1999
Heparin-binding epidermal growth factor-like growth factor, a v-Jun target gene, induces oncogenic transformation.
    Proceedings of the National Academy of Sciences of the United States of America, 1999, May-11, Volume: 96, Issue:10

    Jun is a transcription factor belonging to the activator protein 1 family. A mutated version of Jun (v-Jun) transduced by the avian retrovirus ASV17 induces oncogenic transformation in avian cell cultures and sarcomas in young galliform birds. The oncogenicity of Jun probably results from transcriptional deregulation of v-Jun-responsive target genes. Here we describe the identification and characterization of a growth-related v-Jun target, a homolog of heparin-binding epidermal growth factor-like growth factor (HB-EGF). HB-EGF is strongly expressed in chicken embryo fibroblasts (CEF) transformed by v-Jun. HB-EGF expression is not detectable or is marginal in nontransformed CEF. Using a hormone-inducible Jun-estrogen receptor chimera, we found that HB-EGF expression is correlated with v-Jun activity. In this system, induction of v-Jun is followed within 1 hr by elevated levels of HB-EGF. In CEF infected with various Jun mutants, HB-EGF expression is correlated with the oncogenic potency of the mutant. Constitutive expression of HB-EGF conveys to CEF the ability to grow in soft agar and to form multilayered foci of transformed cells on a solid substrate. These observations suggest that HB-EGF is an effector of Jun-induced oncogenic transformation.

    Topics: Amino Acid Sequence; Animals; Cell Division; Cell Transformation, Neoplastic; Cells, Cultured; Chick Embryo; Cloning, Molecular; Epidermal Growth Factor; Gene Expression Regulation, Neoplastic; Heparin-binding EGF-like Growth Factor; Intercellular Signaling Peptides and Proteins; Molecular Sequence Data; Oncogene Protein p65(gag-jun); Oncogenes; Receptors, Estrogen; Recombinant Fusion Proteins; RNA, Messenger; Sequence Alignment; Transfection

1999
Vitamin D3 receptor (VDR) expression in HC-11 mammary cells: regulation by growth-modulatory agents, differentiation, and Ha-ras transformation.
    Breast cancer research and treatment, 1999, Volume: 54, Issue:2

    HC-11 mammary epithelial cells which originate from midpregnant BALB/c mice are able to differentiate in culture after epidermal (EGF) or basic fibroblast (FGF) growth factor pretreatment followed by lactogenic hormone stimulation (Dexamethasone, Insulin, and Prolactin - DIP). In our study, HC-11 cells exhibited specific vitamin D3 receptors (VDR) determined by Northern analysis or flow cytometry and responded to 10 nM vitamin D3 treatment displaying strong growth inhibition, arrest in G0/G1 phase without evidence of apoptosis, and VDR mRNA reduction, although the percentage of cells expressing VDR protein remained unchanged. In an attempt to verify if there was a correlation between the growth state of the cells and VDR levels, we have examined the effects of growth modulators such as EGF/bFGF and confluency and transformation by Ha-ras. A down-regulation of VDR expression was observed after Ha-ras transformation of HC-11 cells which desensitized the cells to the growth inhibitory effects of vitamin D3. EGF or bFGF decreased VDR in parental cells and EGF antagonized the antiproliferative activity of vitamin D3. As well, transition from proliferating to confluent state significantly reduced VDR levels only in parental cells. DIP-induced HC-11 cell differentiation (monitored by beta-casein transcripts), although leading to cell cycle arrest, increased VDR mRNA content, which seems to be rather related to lactogenic hormone induction than to differentiation itself. In fact, DIP-stimulated HC-11 cells in the absence of EGF pretreatment, or DIP-treated HC-11ras cultures, also displayed up-regulated VDR level even in the absence of differentiation. Concluding, mammary VDR levels might be regulated by growth modulating agents, by physiological conditions of the gland, and by the ras-mediated malignant transformation.

    Topics: Animals; Cell Cycle; Cell Differentiation; Cell Line; Cell Transformation, Neoplastic; Dexamethasone; Epidermal Growth Factor; Female; Gene Expression Regulation; Genes, ras; Growth Substances; Insulin; Mammary Glands, Animal; Mice; Mice, Inbred BALB C; Pregnancy; Prolactin; Receptors, Calcitriol; RNA, Messenger; Transcription, Genetic

1999
Production of epidermal growth factor related ligands in tumorigenic and benign human lung epithelial cells.
    Cancer letters, 1999, Jul-19, Volume: 142, Issue:1

    We recently demonstrated that human lung epithelial cells, overexpressing ErbB-2, formed tumors in nude mice only when high levels of transforming growth factor alpha (TGFalpha) were produced. Cells transfected with a TGFalpha antisense vector failed to form tumors in nude mice. In order to further evaluate the importance, for tumorigenicity, of TGFalpha and its stimulation of ErbB family signalling, the production of other EGF family growth factors by these human lung epithelial cells was studied. We demonstrate for the first time that both tumorigenic and non-tumorigenic human lung epithelial cells produced, in addition to TGFalpha, amphiregulin, betacellulin, heparin-binding EGF and heregulin. These data suggest that human lung epithelial cells have the potential for multifactorial modulation of ErbB receptor family signalling through control of ligand as well as receptor production. In this system, the probable importance of TGFalpha-stimulated signaling for tumorigenicity is supported by its 13-fold higher production in tumorigenic as compared with non-tumorigenic cells and the 2-fold or lower differences observed in production of the other epidermal growth factor (EGF) family ligands.

    Topics: Animals; Cell Transformation, Neoplastic; Epidermal Growth Factor; Gene Expression Regulation, Neoplastic; Genes, erbB-2; Humans; Ligands; Lung; Lung Neoplasms; Mice; Neoplasms, Experimental; Receptor, ErbB-2; Transforming Growth Factor alpha

1999
Perturbation of EGF-induced MAP kinase activation by TGF-beta 1.
    Brazilian journal of medical and biological research = Revista brasileira de pesquisas medicas e biologicas, 1999, Volume: 32, Issue:7

    TGF-beta 1 regulates both cellular growth and phenotypic plasticity important for maintaining a growth advantage and increased invasiveness in progressively malignant cells. Recent studies indicate that TGF-beta-1 stimulates the conversion of epitheliod to fibroblastoid phenotype which presumably leads to the inactivation of growth-inhibitory effects by TGF-beta 1 (Portella et al. (1998) Cell Growth and Differentiation, 9: 393-404). Therefore, the investigation of TGF-beta 1 signaling that leads to altered growth and migration may provide novel targets for the prevention of increased cell growth and invasion. Although much attention has been paid to TGF-beta 1 responses in epithelial cells, the above studies suggest that examination of signal transduction pathways in fibroblasts are important as well. Data from our laboratory are consistent with the concept that TGF-beta 1 can act as a regulatory switch in density-dependent C3H 10T1/2 fibroblasts capable of either promoting or delaying G1 traverse. The regulation of this switch is proposed to occur prior to pRb phosphorylation, namely prior to activation of cyclin-dependent kinases. The current study is concerned with the evaluation of a key cyclin (cyclin D1) which activates cdk4 and p27KIP1 which in turn inhibit cdk2 in the proliferative responses of epidermal growth factor (EGF) and platelet-derived growth factor (PDGF) and their modulation by TGF-beta 1. Although the molecular events that lead to elevation of cyclin D1 are not completely understood, it appears likely that activation of p42/p44MAPK kinases is involved in its transcriptional regulation. TGF-beta 1 delayed EGF- or PDGF-induced cyclin D1 expression and blocked the induction of active p42/p44MAPK. The mechanism by which TGF-beta 1 induces a block in p42/p44MAPK activation is being examined and the possibility that TGF-beta 1 regulates phosphatase activity is being tested.

    Topics: Cell Transformation, Neoplastic; Cyclin D1; Enzyme Activation; Epidermal Growth Factor; Humans; Mitogen-Activated Protein Kinases; Transforming Growth Factor beta

1999
Controlled dimerization of ErbB receptors provides evidence for differential signaling by homo- and heterodimers.
    Molecular and cellular biology, 1999, Volume: 19, Issue:10

    The four members of the ErbB family of receptor tyrosine kinases are involved in a complex array of combinatorial interactions involving homo- and heterodimers. Since most cell types express more than one member of the ErbB family, it is difficult to distinguish the biological activities of different homo- and heterodimers. Here we describe a method for inducing homo- or heterodimerization of ErbB receptors by using synthetic ligands without interference from the endogenous receptors. ErbB receptor chimeras containing synthetic ligand binding domains (FK506-binding protein [FKBP] or FKBP-rapamycin-binding domain [FRB]) were homodimerized with the bivalent FKBP ligand AP1510 and heterodimerized with the bifunctional FKBP-FRB ligand rapamycin. AP1510 treatment induced tyrosine phosphorylation of ErbB1 and ErbB2 homodimers and recruitment of Src homology 2 domain-containing proteins (Shc and Grb2). In addition, ErbB1 and ErbB2 homodimers activated downstream signaling pathways leading to Erk2 and Akt phosphorylation. However, only ErbB1 homodimers were internalized upon AP1510 stimulation, and only ErbB1 homodimers were able to associate with and induce phosphorylation of c-Cbl. Cells expressing AP1510-induced ErbB1 homodimers were able to associate with and induce phosphorylation of c-Cbl. Cells expressing AP1510-induced ErbB1 homodimers were able to form foci; however, cells expressing ErbB2 homodimers displayed a five- to sevenfold higher focus-forming ability. Using rapamycin-inducible heterodimerization we show that c-Cbl is unable to associate with ErbB1 in a ErbB1-ErbB2 heterodimer most likely because ErbB2 is unable to phosphorylate the c-Cbl binding site on ErbB1. Thus, we demonstrate that ErbB1 and ErbB2 homodimers differ in their abilities to transform fibroblasts and provide evidence for differential signaling by ErbB homodimers and heterodimers. These observations also validate the use of synthetic ligands to study the signaling and biological specificity of selected ErbB dimers in any cell type.

    Topics: Animals; Cell Cycle; Cell Transformation, Neoplastic; Dimerization; Dose-Response Relationship, Drug; Endocytosis; Epidermal Growth Factor; ErbB Receptors; Ligands; Phosphorylation; Proto-Oncogene Proteins; Proto-Oncogene Proteins c-cbl; Rats; Receptor, ErbB-2; Recombinant Fusion Proteins; Signal Transduction; Tacrolimus; Ubiquitin-Protein Ligases

1999
Antagonistic effects of protein kinase C alpha and delta on both transformation and phospholipase D activity mediated by the epidermal growth factor receptor.
    Molecular and cellular biology, 1999, Volume: 19, Issue:11

    Downregulation of protein kinase C delta (PKC delta) by treatment with the tumor-promoting phorbol ester 12-O-tetradecanoylphorbol-13-acetate (TPA) transforms cells that overexpress the non-receptor class tyrosine kinase c-Src (Z. Lu et al., Mol. Cell. Biol. 17:3418-3428, 1997). We extended these studies to cells overexpressing a receptor class tyrosine kinase, the epidermal growth factor (EGF) receptor (EGFR cells); like c-Src, the EGF receptor is overexpressed in several human tumors. In contrast with expectations, downregulation of PKC isoforms with TPA did not transform the EGFR cells; however, treatment with EGF did transform these cells. Since TPA downregulates all phorbol ester-responsive PKC isoforms, we examined the effects of PKC delta- and PKC alpha-specific inhibitors and the expression of dominant negative mutants for both PKC delta and alpha. Consistent with a tumor-suppressing function for PKC delta, the PKC delta-specific inhibitor rottlerin and a dominant negative PKC delta mutant transformed the EGFR cells in the absence of EGF. In contrast, the PKC alpha-specific inhibitor Go6976 and expression of a dominant negative PKC alpha mutant blocked the transformed phenotype induced by both EGF and PKC delta inhibition. Interestingly, both rottlerin and EGF induced substantial increases in phospholipase D (PLD) activity, which is commonly elevated in response to mitogenic stimuli. The elevation of PLD activity in response to inhibiting PKC delta, like transformation, was dependent upon PKC alpha and restricted to the EGFR cells. These data demonstrate that PKC isoforms alpha and delta have antagonistic effects on both transformation and PLD activity and further support a tumor suppressor role for PKC delta that may be mediated by suppression of tyrosine kinase-dependent increases in PLD activity.

    Topics: Acetophenones; Animals; Benzopyrans; Carbazoles; Cell Transformation, Neoplastic; Cells, Cultured; Crosses, Genetic; Epidermal Growth Factor; ErbB Receptors; Indoles; Isoenzymes; Models, Genetic; Mutagenesis, Insertional; Phospholipase D; Protein Kinase C; Protein Kinase C-alpha; Protein Kinase C-delta; Rats; Recombinant Proteins; Tetradecanoylphorbol Acetate

1999
Sustained mitogen-activated protein kinase activation is induced by transforming erbB receptor complexes.
    DNA and cell biology, 1999, Volume: 18, Issue:10

    We used a genetic approach to characterize features of mitogen-activated protein kinase (MAPK) activation occurring as a consequence of expression of distinct erbB receptor combinations in transformed human cells. Kinase-deficient erbB proteins reduced epidermal growth factor (EGF)-induced tyrosine phosphorylation of endogenous Shc proteins and also reduced immediate and sustained EGF-induced ERK MAPK activities in human glioblastoma cells, although basal ERK MAPK activities were unaffected. Basal and EGF-induced JNK and p38 MAPK kinase activities were equivalent in parental cancer cells and EGFR-inhibited subclones. When ectopically overexpressed in murine fibroblasts and human glioblastoma cells, a constitutively activated human EGF receptor oncoprotein (deltaEGFR) induced EGF-independent elevation of basal ERK MAPK activity. Basal JNK MAPK kinase activity was also specifically induced by deltaEGFR, which correlated with increased phosphorylation of a 54-kDa JNK2 protein observed in deltaEGFR-containing cells. The JNK activities in response to DNA damage were comparably increased in cells containing wildtype EGFR or deltaEGFR. Consistent with the notion that transforming erbB complexes induce sustained and unregulated MAPK activities, coexpression of p185(neu) and EGFR proteins to levels sufficient to transform murine fibroblasts also resulted in prolonged EGF-induced ERK in vitro kinase activation. Transforming erbB complexes, including EGFR homodimers, deltaEGFR homodimers, and p185(neu)/EGFR heterodimers, appear to induce sustained, unattenuated activation of MAPK activities that may contribute to increased transformation and resistance to apoptosis in primary human glioblastoma cells.

    Topics: Adaptor Proteins, Signal Transducing; Adaptor Proteins, Vesicular Transport; Animals; Brain Neoplasms; Cell Line, Transformed; Cell Transformation, Neoplastic; Dimerization; DNA Damage; Enzyme Activation; Epidermal Growth Factor; ErbB Receptors; Fibroblasts; Genes, erbB; Glioblastoma; Humans; JNK Mitogen-Activated Protein Kinases; MAP Kinase Signaling System; Mice; Mitogen-Activated Protein Kinase 1; Mitogen-Activated Protein Kinase 3; Mitogen-Activated Protein Kinase Kinases; Mitogen-Activated Protein Kinases; Neoplasm Proteins; p38 Mitogen-Activated Protein Kinases; Phosphorylation; Protein Multimerization; Protein Processing, Post-Translational; Protein Structure, Tertiary; Proteins; Receptor, ErbB-2; Recombinant Fusion Proteins; Sequence Deletion; Shc Signaling Adaptor Proteins; Src Homology 2 Domain-Containing, Transforming Protein 1; Transfection; Tumor Cells, Cultured

1999
Changes associated with delay of mammary cancer by retinoid analogues in transgenic mice bearing c-neu oncogene.
    Breast cancer research and treatment, 1999, Volume: 58, Issue:3

    Breast cancer is one of the common cancers and is a leading cause of cancer mortality in women. The TG.NK transgenic mouse line on FVB strain background expresses the c-neu oncogene under the control of a MMTV promoter in mammary tissue and appears to be a useful animal model for evaluation of strategies to delay or prevent mammary cancer. Fiber-rich nonpurified diet (NTP-2000) and some retinoid analogues have delayed mammary cancer in the TG.NK model. Four week old hemizygous TG.NK female mice with MMTV/c-neu (erbB2) activated oncogene were fed NTP-2000 diet containing the retinoid analogue 4-hydroxyphenylretinamide (4-HPR) at 7 mmol/kg or the arotinoid Ro 40-8757 at 1.5 and 2.5 mmol/kg for 26 weeks. The 4-HPR at 7 mmol/kg diet delayed the development of palpable tumors up to 24 weeks, but by 26 weeks, the incidence markedly increased and was closer to the NTP-2000 diet control group. However, the 4-HPR diet markedly decreased the average weight of the tumors at 26 weeks with no decrease in multiplicity. The 4-HPR also caused significant increase in liver weights without an effect on body weight. Arotinoid Ro 40-8757 caused marked decrease in the number and branching of mammary ducts, and inhibited mammary tumor development with significant decrease in the incidence, multiplicity, and tumor weights compared to the NTP-2000 diet control. Arotinoid also caused a significant dose-related increase in liver weights without a significant effect on body weights. At the doses tested, the arotinoid but not 4-HPR decreased the circulating levels of IGF-1. However, there was no association between the IGF-1 levels and the size, incidence, or absence of tumors when evaluated for any treatment group or for all mice in the study irrespective of treatment. The oncogene erbB2 (c-neu) and the growth factor EGF expression were more prominent in the small tumors of the mice treated with arotinoid than in the larger tumors of the control group. PCNA staining was observed in areas where there was high erbB2 and EGF staining. The delay in onset of mammary tumors by the above retinoid analogues may be related to the delay in development of mammary glands.

    Topics: Animals; Body Weight; Cell Transformation, Neoplastic; Diet; Epidermal Growth Factor; Female; Genes, erbB-2; Mammary Glands, Animal; Mammary Neoplasms, Experimental; Mice; Mice, Transgenic; Retinoids

1999
Shortage of mitogen-activated protein kinase is responsible for resistance to AP-1 transactivation and transformation in mouse JB6 cells.
    Proceedings of the National Academy of Sciences of the United States of America, 1998, Jan-06, Volume: 95, Issue:1

    The JB6 mouse epidermal cell system, which includes tumor promotion-sensitive (P+) and tumor promotion-resistant (P-) cells, is a well-established and extensively used cell culture model for studying the mechanism of late-stage tumor promotion. Tumor promoters, such as 12-O-tetradecanoylphorbol 13-acetate (TPA) or epidermal growth factor (EGF), induce high levels of activator protein 1 (AP-1) activity and large, tumorigenic, anchorage-independent colonies in soft agar at a high frequency in JB6 P+ cells, but not in JB6 P- cells. We report here a molecular explanation for the defect in the AP-1 activation and promotion-resistant phenotype of P- cells. We demonstrate that the lack of AP-1 activation and cell transformation responses to TPA and EGF in P- cells appears attributable to the low level of mitogen-activated protein kinase (MAPK) (extracellular signal-regulated protein kinase, Erk) in these cells. TPA and EGF induce transactivation of AP-1 activity in P+ cells but not in P- cells. Nonphosphorylated forms and TPA- or EGF-induced phosphorylated forms of Erks (Erk1 and Erk2) in P- cells were much lower than those in P+ cells. Stable transfection of wild-type MAPK (Erk2) into P- cells restored its response to TPA and EGF for both AP-1 activation and cell transformation. These results suggest that the shortage of MAPK (Erk1 and Erk2) appears to be an important contributor to the tumor promotion-resistant phenotype in JB6 cells.

    Topics: Animals; Calcium-Calmodulin-Dependent Protein Kinases; Carcinogens; Cell Line; Cell Transformation, Neoplastic; Epidermal Growth Factor; Epidermis; Mice; Phosphorylation; Tetradecanoylphorbol Acetate; Transcription Factor AP-1; Transcriptional Activation

1998
Murine Ksr interacts with MEK and inhibits Ras-induced transformation.
    Current biology : CB, 1998, Jan-01, Volume: 8, Issue:1

    Ksr (kinase supressor of Ras) was identified as a regulator of the Ras-MAP kinase (mitogen-activated protein kinase) pathway by genetic screens in Drosophila and Caenorhabditis elegans. Ksr is a kinase with similarities to the three conserved regions of Raf kinases, especially within the kinase domain. To investigate whether these structural similarities correlated with common functional properties, we examined the ability of mKsr-1, the murine homolog of Ksr, to interact with components of the vertebrate MAP kinase pathway.. In the yeast two-hybrid interaction assay, mKsr-1 did not bind to either Ras, B-Raf or Raf-1, but interacted strongly with both MEK-1 and MEK-2, activators of MAP kinase. The Ksr-MEK interaction was confirmed by co-immunoprecipitation experiments. Ectopically expressed mKsr-1 co-precipitated with endogenous MEK-1 in COS-1 cells, and endogenous Ksr and MEK co-precipitated from PC12 cells. Phosphorylation of MEK by mKsr-1 was not detected, however. In contrast, the MEK subpopulation complexed with mKsr-1 in COS-1 cells or PC12 cells did not display kinase activity. This ability of Ksr to block MEK in an inactive form correlated with a biological response: mKsr-1 did not transform NIH3T3 cells, and, furthermore, mKsr-1 reduced Ras-induced transformation. Similarly, mKsr-1 inhibited the proliferation of embryonic neuroretina cells induced by Ras and B-Raf but not that induced by MEK.. Our results suggest a novel mechanism for Ksr in regulating the MAP kinase pathway, at least in part through an ability to interact with MEK.

    Topics: 3T3 Cells; Animals; Cell Division; Cell Transformation, Neoplastic; Chick Embryo; COS Cells; Epidermal Growth Factor; MAP Kinase Kinase 1; MAP Kinase Kinase 2; Mice; Mitogen-Activated Protein Kinase Kinases; Nerve Growth Factors; PC12 Cells; Protein Kinases; Protein Serine-Threonine Kinases; Protein-Tyrosine Kinases; Proto-Oncogene Proteins c-raf; ras Proteins; Rats; Retina; Signal Transduction

1998
Overexpression of the MEN/ELL protein, an RNA polymerase II elongation factor, results in transformation of Rat1 cells with dependence on the lysine-rich region.
    The Journal of biological chemistry, 1998, Feb-27, Volume: 273, Issue:9

    The MEN gene (also called ELL) encodes an RNA polymerase II elongation factor that has been implicated in t(11;19)(q23;p13.1) translocation in myeloid leukemias. The function of another elongation factor, elongin, is known to be inhibited by VHL tumor suppressor protein in vitro, suggesting the possible relationship of aberrant transcriptional elongation to oncogenesis. We overexpressed the MEN protein in Rat1 fibroblasts to evaluate its transforming activity. MEN-overexpressing cells acquired the capacity for anchorage-independent growth. In addition, the growth factor requirement was decreased in these cells. However, cells expressing a deletion mutant of MEN lacking the lysine-rich region did not exhibit such biological abilities. c-Fos protein expression and AP-1 activity were elevated in the MEN-expressing cells, which might be part of the mechanism responsible for the transformation. The c-fos mRNA, the expression of which is known to be regulated partly at the stage of transcriptional elongation, appeared earlier in the MEN-expressing cells than in cells transfected with an empty vector or the deletion mutant lacking the lysine-rich region after stimulation with epidermal growth factor. The RNA polymerase II elongation factor MEN may play an important role in the regulation of cell proliferation.

    Topics: 3T3 Cells; Animals; Cell Nucleus; Cell Transformation, Neoplastic; Cells, Cultured; DNA-Binding Proteins; Epidermal Growth Factor; Fibroblasts; Gene Expression; Gene Transfer Techniques; Humans; Leukemia, Myeloid; Lysine; Mice; Mutation; Neoplasm Proteins; Peptide Elongation Factors; Proto-Oncogene Proteins c-fos; Rats; Recombinant Proteins; Retroviridae; RNA Polymerase II; RNA, Messenger; Sequence Deletion; Transcription Factor AP-1; Transcription Factors; Transcription, Genetic; Transcriptional Elongation Factors

1998
Vascular endothelial growth factor expression and vascular density as prognostic markers of survival in patients with low-grade astrocytoma.
    Journal of neurosurgery, 1998, Volume: 88, Issue:3

    It has long been recognized that some patients with low-grade astrocytoma may survive for many years, whereas in others the disease follows a more malignant course resulting in a short survival time, usually due to malignant transformation into higher-grade tumors.. The aim of this study was to investigate angiogenesis in the initial biopsy specimen of tumor tissue as a biological marker to identify patients with low-grade astrocytoma who are at high risk of malignant tumor transformation or death.. Tumor tissue was studied in 74 consecutively treated adult patients in whom a diagnosis of diffuse supratentorial hemispheric histologically proven fibrillary low-grade astrocytoma was made and who underwent surgery between January 1972 and January 1994. Studies were conducted using monoclonal antibodies to the antigens of the proliferation-associated Ki-67 (MIB-1), factor VIII, vascular endothelial growth factor (VEGF), basic fibroblast growth factor (bFGF), and epidermal growth factor (EGF). The overall 5-year survival rate for the entire patient population was 65%, with a median survival time of 7.5 years. The total mean follow-up period was 6.1 years. All tumors showed a low proliferative potential at the time of the initial operation, as demonstrated by an MIB-1 labeling index of less than 1.5%. Patients with more than seven microvessels in tumor tissue (29 cases) had a shorter survival time (mean 3.8 years) than those with seven or fewer microvessels (mean survival 11.2 years). This difference in survival times was significant by univariate (p = 0.001) and stepwise multivariate analyses (p < 0.001). Tumors with a larger number of microvessels also had a greater chance of undergoing malignant transformation (p = 0.001). Similarly, significant staining for VEGF was correlated with shorter survival times when using univariate (p = 0.003) and multivariate (p = 0.008) analyses and with a greater chance of malignant transformation (p = 0.002). Patients with tumors staining positive for VEGF (39 individuals) had a median survival time of 5.3 years, and those with tumors negative for VEGF (35 patients) had a median survival time of 11.2 years. No association was observed between bFGF, EGF, and survival or malignant transformation. The stepwise multivariate analysis included histological and clinical variables simultaneously.. The authors have shown that microvessel density and VEGF levels are independent prognostic markers of survival in fibrillary low-grade astrocytoma. This finding leads them to propose that fibrillary diffuse low-grade astrocytoma is not a single pathological entity but is composed of a spectrum of tumors with differing propensities to undergo malignant transformation that is at least partly based on their inherent angiogenic potential.

    Topics: Adolescent; Adult; Aged; Analysis of Variance; Antibodies, Monoclonal; Astrocytoma; Biomarkers, Tumor; Capillaries; Cell Division; Cell Transformation, Neoplastic; Coloring Agents; Endothelial Growth Factors; Epidermal Growth Factor; Factor VIII; Female; Fibroblast Growth Factor 2; Follow-Up Studies; Gene Expression Regulation, Neoplastic; Glioblastoma; Humans; Ki-67 Antigen; Lymphokines; Male; Microcirculation; Middle Aged; Multivariate Analysis; Neovascularization, Pathologic; Prognosis; Supratentorial Neoplasms; Survival Rate; Treatment Outcome; Vascular Endothelial Growth Factor A; Vascular Endothelial Growth Factors; Venules

1998
Advances in the understanding of malignant transformation of keratinocytes: an immunohistochemical study.
    Journal of the European Academy of Dermatology and Venereology : JEADV, 1998, Volume: 10, Issue:2

    We set out to investigate the interactions between malignant transformation of keratinocytes, presence of oncoproteins and immunosurveillance in squamous cell carcinoma (SCC) and in a preneoplastic lesion, actinic keratosis (AK).. Samples of SCC, AK and normal skin (NS) were subjected to quantitative analysis using the following antibodies: anti-p53, Ki67, OKT6, OK-DR, B7/BB1, anti-CD54, anti-CD11, OKT3, OKT4, OKT8; positivity for ras-p21, EGFr and bcl-2 was evaluated by semiquantitative analysis.. Oncoprotein alterations and increased keratinocyte proliferative activity were observed both in AK and SCC. The number of Langerhans cells (CD1a+ cells) was similar in the two lesions but lower in SCC compared to AK. The proportion of CD1a(+)-B7/BB1+ cells was slightly higher in AK and SCC than in NS. The Langerhans cells expressed the HLA-DR antigen in all groups. Values were highest in AK and NS, and quite low in SCC. Cytotoxic T lymphocytes were more numerous in SCC than in AK and NS. Interestingly, the total CD4/CD8 ratio was much lower in SCC than in AK and NS, which indicates an increase in the CD8+ subpopulation in samples of SCC. In the epithelia of SCC samples there were a considerable number of B7/BB1+ keratinocytes.. We suggest that alterations in the immunodefence mechanisms have an important role in the transformation of AK into SCC, and that these changes affect not only lymphocytes, but also professional (i.e., Langerhans cells) and non-professional (i.e., keratinocytes) antigen presenting cells.

    Topics: Aged; Analysis of Variance; Biopsy, Needle; Carcinoma, Squamous Cell; CD3 Complex; CD4 Antigens; CD8 Antigens; Cell Transformation, Neoplastic; Cells, Cultured; Epidermal Growth Factor; Female; HLA-DR Antigens; Humans; Immunohistochemistry; Intercellular Adhesion Molecule-1; Keratinocytes; Keratosis; Male; Middle Aged; Oncogene Proteins; Precancerous Conditions; Skin Neoplasms

1998
Gene trapping identifies inhibitors of oncogenic transformation. The tissue inhibitor of metalloproteinases-3 (TIMP3) and collagen type I alpha2 (COL1A2) are epidermal growth factor-regulated growth repressors.
    The Journal of biological chemistry, 1998, May-29, Volume: 273, Issue:22

    A gene trap strategy has been used to identify genes that are repressed in cells transformed by an activated epidermal growth factor (EGF)/EGF receptor signal transduction pathway. EGF receptor-expressing NIH3T3 cells (HER1 cells) were infected with a retrovirus containing coding sequences for the human CD2 antigen and for secreted alkaline phosphatase in the U3 region. By selecting for and against CD2 expression, we obtained clones in which the gene trap had integrated into genes selectively repressed by EGF. Two of these clones encoded for the secreted extracellular matrix proteins TIMP3 and COL1A2. We show here that both genes are downstream targets of RAS and are specifically repressed by EGF-induced transformation. Moreover, this strategy tags tumor suppressor genes in their normal chromosomal location, thereby improving target-specific screens for antineoplastic drugs.

    Topics: 3T3 Cells; Animals; Base Sequence; Cell Transformation, Neoplastic; Collagen; DNA; Epidermal Growth Factor; ErbB Receptors; Humans; Mice; Molecular Sequence Data; Phenotype; RNA, Messenger; Sequence Homology, Nucleic Acid; Signal Transduction; Tissue Inhibitor of Metalloproteinase-3

1998
Interleukin-6 and epidermal growth factor promote anchorage-independent growth of immortalized human prostatic epithelial cells treated with N-methyl-N-nitrosourea.
    The Prostate, 1998, Jun-01, Volume: 35, Issue:4

    Epidermal growth factor (EGF) and interleukin (IL)-6 are implicated in the growth of benign and malignant prostatic epithelial cells. We investigated the role of EGF and IL-6 during the process of prostate carcinogenesis.. Using growth in soft agar as an index of transformation, we examined the effect of EGF and IL-6 on the enhancement of N-methyl-N-nitrosourea (MNU)-initiated transformation of immortalized, nontumorigenic prostatic epithelial cell lines (PWR-1E and RWPE-1) developed in our laboratory. The effect of EGF and IL-6 on the growth of MNU-induced transformants isolated from soft agar was assessed both in monolayer culture and in a soft agar.. After a 1 hr exposure to N-methyl-N-nitrosourea (50 microg/ml), cells (5 x 10(4)) were grown in soft agar in the presence of EGF (5 ng/ml) or IL-6 (10 or 100 ng/ml). Addition of EGF or IL-6 significantly increased colony formation in soft agar of both immortalized prostatic epithelial cell lines initiated with MNU (P < 0.001-0.05). Only a very small number of colonies was observed with the parental cell lines PWR-1E and RWPE-1 not exposed to MNU, and their numbers increased by the addition of EGF or IL-6. All of the transformants, derived by exposure to MNU and isolated from soft agar, exhibited a higher cell growth potential in monolayer cultures than did their parental cell lines. Furthermore, as compared to the parental cell lines, growth response of MNU-transformants to 5alpha-dihydrotestosterone (5alpha-DHT), EGF, or IL-6 in monolayer culture was better in 5 of 8, 6 of 8, and 7 of 8 cell lines, respectively. All of the MNU-transformants exhibited a far higher colony-forming efficiency in soft agar than did the parental cell lines. However, the degree of responsiveness to EGF or IL-6 in soft agar varied among the MNU-transformants.. The results of the present study suggest that IL-6 and EGF may enhance prostate carcinogenesis in vitro by preferentially stimulating the growth of transformed cells.

    Topics: Cell Adhesion; Cell Culture Techniques; Cell Division; Cell Line; Cell Transformation, Neoplastic; Epidermal Growth Factor; Epithelial Cells; Humans; Interleukin-6; Kinetics; Male; Methylnitrosourea; Prostate; Receptors, Interleukin-6; Time Factors

1998
Constitutive association of EGF receptor with the CrkII-23 mutant that inhibits transformation of NRK cells by EGF and TGF-beta.
    Cellular signalling, 1998, Volume: 10, Issue:4

    Crk belongs to the adapter proteins that participate in many signalling pathways from cell surface receptors. We have characterised the CrkII-23 mutant that inhibits the transformation of NRK cells induced by epidermal growth factor (EGF) and transforming growth factor (TGF)-beta. To study the biochemical difference, cDNAs of the wild-type CrkII and the CrkII-23 mutant were introduced stably into NIH 3T3 cells expressing EGF receptor (EGFR). Both CrkII and CrkII-23 were phosphorylated on tyrosine upon EGF simulation with similar time course and dose dependency. Whereas the wild-type CrkII bound to EGFR only after EGF stimulation, CrkII-23 bound to EGFR from before stimulation. Mutation in the Src homology (SH) 2 or amino-terminal SH3 domain did not abolish the binding of CrkII-23 to EGFR in the quiescent cells, suggesting that the binding is mediated by a novel mechanism. These CrkII-23-derived mutants, however, did not suppress transformation of NRK cells by EGF and TGF-beta. Hence, both the SH2 and amino-terminal SH3 domains are required to inhibit transformation of NRK cells. These results suggest that persistent signalling from CrkII-23 bound to EGFR suppresses transformation by EGF and TGF-beta in NRK23 cells.

    Topics: 3T3 Cells; Adaptor Proteins, Signal Transducing; Adaptor Proteins, Vesicular Transport; Animals; Cell Differentiation; Cell Transformation, Neoplastic; COS Cells; DNA Mutational Analysis; Epidermal Growth Factor; ErbB Receptors; Eukaryotic Initiation Factor-2; Guanine Nucleotide Exchange Factors; Humans; Mice; Mutation; Neurons; PC12 Cells; Phosphorylation; Protein Binding; Protein Kinases; Protein Structure, Tertiary; Proteins; Proto-Oncogene Proteins; Proto-Oncogene Proteins c-abl; Proto-Oncogene Proteins c-cbl; Proto-Oncogene Proteins c-crk; Rats; Shc Signaling Adaptor Proteins; Src Homology 2 Domain-Containing, Transforming Protein 1; src Homology Domains; Transforming Growth Factor beta; Tyrosine; Ubiquitin-Protein Ligases

1998
Absence of autophosphorylation site Y882 in the p185neu oncogene product correlates with a reduction of transforming potential.
    Oncogene, 1998, Jun-04, Volume: 16, Issue:22

    Autophosphorylation of type I receptor tyrosine kinases (RTKs) comprises one step in the signaling events mediated by erbB receptors such as p185neu and EGFR. Previous analysis of p185neu has indicated that there are at least five tyrosine autophosphorylation sites, Y882, Y1028, Y1143, Y1226/7 and Y1253, of which Y882 might be important because of its location in the kinase activity domain. We have specifically analysed the effect of a Y882F (phenylalanine substituted for tyrosine at position 882) mutation in the enzymatic active domain. We also deleted the carboxyl terminal 122 amino acids which contained three other autophosphorylation sites (TAPstop) and combined mutants of that deletion with Y882F (Y882F/APstop). Both in vitro and in vivo transformation assays showed that substitution of tyrosine882 by phenylalanine significantly decreased the transforming potential of activated, oncogenic p185neu, although no significant difference in the total phosphotyrosine levels of the mutant proteins were observed. To analyse mitogenic signaling in response to ligand, the intracellular domains of p185neu and Y882F were fused with the extracellular domain of the EGF receptor. The proliferation of cells expressing these chimeric receptors was EGF-dependent, and cells expressing EGFR/Y882F chimeric receptors were less responsive to EGF stimulation than those expressing EGFR/neu receptors. In vitro kinase assays demonstrated that abolishing the autophosphorylation site Y882 diminished the enzymatic tyrosine kinase activity of p185neu. These studies, taken together with the phenotypic inhibition observed with cells expressing Y882F, suggest that the tyrosine882 residue may be important for p185neu-mediated transformation by affecting the enzymatic kinase function of the p185neu receptor.

    Topics: Amino Acid Substitution; Animals; Binding Sites; Cell Division; Cell Transformation, Neoplastic; Epidermal Growth Factor; ErbB Receptors; Mitogens; Mutagenesis, Site-Directed; Oncogenes; Phenylalanine; Phosphorylation; Rats; Receptor, ErbB-2; Tyrosine

1998
STAT3 mediates the survival signal in oncogenic ras-transfected intestinal epithelial cells.
    International journal of cancer, 1998, Oct-29, Volume: 78, Issue:3

    The oncogenic ras mutation is a common and critical step in gastrointestinal carcinogenesis. In a previous study, we demonstrated that oncogenic ras activated the EGF-related peptide autocrine loop and that the apoptosis resistance observed in the oncogenic ras-stimulated cell (IEC-ras cell) was dependent on this activated EGF-related peptide autocrine loop. STATs (signal transducers and activators of transcription), first identified as intracellular signal transducers stimulated by cytokines, are known to also be activated by EGF. However, the role of STATs in the survival signal of IEC-ras cells is not clear. In the present study, we demonstrate that STAT3 is constitutively activated in ras-stimulated cells and that STAT3 activation is considerably suppressed by the EGF-specific receptor kinase inhibitor AG 1478. We also show that disruption of the STAT3 pathway by introduction of a dominant-negative STAT3 mutant abolishes the apoptosis resistance against UVC and MMC treatment observed in IEC-ras cells without affecting proliferation. Moreover, the expression of Bcl-2 and Bcl-xL, apoptosis-suppressive proteins, is reduced in dominant-negative STAT3-transfected cells. Thus, STAT3 appears to be an important mediator of the antiapoptotic signal in IEC-ras cells.

    Topics: Acute-Phase Proteins; Animals; bcl-X Protein; Cell Line; Cell Survival; Cell Transformation, Neoplastic; DNA Fragmentation; DNA-Binding Proteins; Enzyme Inhibitors; Epidermal Growth Factor; ErbB Receptors; Genes, ras; Intestinal Mucosa; Phosphorylation; Proto-Oncogene Proteins c-bcl-2; Quinazolines; Rats; Signal Transduction; STAT1 Transcription Factor; STAT3 Transcription Factor; Trans-Activators; Transfection; Tyrphostins

1998
Stat3 plays an important role in oncogenic Ros- and insulin-like growth factor I receptor-induced anchorage-independent growth.
    The Journal of biological chemistry, 1998, Oct-23, Volume: 273, Issue:43

    The role of signal transducers and activators of transcription (STATs) in receptor protein-tyrosine kinase (PTK)-induced cell growth and transformation was investigated using an inducible epidermal growth factor receptor-Ros chimeric receptor called ER2 and a constitutively activated insulin-like growth factor I receptor called NM1, both of which are able to induce anchorage-independent growth of NIH 3T3 cells. ER2 and NM1 receptor PTKs are able to cause Stat3 activation. Co-expressing the dominant negative Stat3 mutant with ER2 or NM1 in transiently or stable transfected cells resulted in a dramatic inhibition of colonies induced by these receptor PTKs and a moderate inhibition of their mitogenicity in monolayer. Therefore, Stat3 is not only important for initiation of transformation, as demonstrated by inhibition of the epidermal growth factor-inducible colony formation of the ER2 cells by the mutant, but it is also required for the maintenance of transformation, as evidenced by reversion of the NM1 transformed cells. The DNA binding and transcriptional activities of the endogenous Stat3 were greatly inhibited in the ER2 and NM1 cells co-expressing the Stat3 mutants. We conclude that activated function of Stat3 is required for the establishment and maintenance of Ros and insulin-like growth factor I receptor PTK-induced cell transformation.

    Topics: 3T3 Cells; Animals; Cell Transformation, Neoplastic; DNA-Binding Proteins; Epidermal Growth Factor; Mice; Mutation; Oncogene Proteins, Viral; Phosphorylation; Protein Binding; Receptor Protein-Tyrosine Kinases; Receptor, IGF Type 1; Signal Transduction; STAT3 Transcription Factor; Trans-Activators; Transcription, Genetic

1998
Mutagenic analysis of Vav reveals that an intact SH3 domain is required for transformation.
    Oncogene, 1998, Sep-24, Volume: 17, Issue:12

    The vav proto-oncogene encodes a protein with multiple modulae domains that enable it to function as a mediator, linking tyrosine signaling to downstream events in hematopoietic cells. Circumstantial evidence suggests that protein-protein interactions exerted by two of these domains, the Src homology 2 (SH2) and the Src homology 3 (SH3), play an important role in the regulation of Vav activity. To study the relevance of the SH3 domain for the function of vav as a transforming gene, we have created several mutations in the SH3 domain located at its carboxy region. Substitution of the non-conserved aspartic acid 797 (to asparagine, D797N) retained the transforming potential of the vav oncogene, whereas substitutions of five highly conserved amino-acids: alanine 789 (to asparagine, A789N), leucine 801 (to arginine, L801R), tryptophan 821 (to arginine, W821R), glycine 830 (to valine, G830V) and valine 837 (to glutamic acid, V837E) greatly reduced its transforming potential. The mutant proteins resemble Vav in many biochemical properties; however, while the transforming mutant protein (D797N) associates with several unidentified proteins in a manner similar to that of Vav, the non-transforming mutant Vav proteins react very poorly with these proteins. Among the known Vav-interacting proteins, hnRNP-K associates with all mutant proteins except A789N and V837E whereas binding of Zyxin to any of the mutant proteins is not affected. Taken together, our results clearly demonstrate that the SH3 domain has a positive effect on vav activity and is needed for vav transformation. The vavSH3C associating protein(s) that are crucial for its activity as a transforming gene have probably not yet been identified.

    Topics: 3T3 Cells; Amino Acid Sequence; Animals; Binding Sites; Cell Transformation, Neoplastic; Epidermal Growth Factor; Humans; Mice; Molecular Sequence Data; Mutagenesis, Site-Directed; Oncogene Proteins; Phosphorylation; Protein Binding; Protein Conformation; Proto-Oncogene Mas; Proto-Oncogene Proteins c-vav; Sequence Homology, Amino Acid; Signal Transduction; src Homology Domains; Transfection

1998
KRAB-independent suppression of neoplastic cell growth by the novel zinc finger transcription factor KS1.
    The Journal of clinical investigation, 1998, Dec-01, Volume: 102, Issue:11

    The study of zinc finger proteins has revealed their potential to act as oncogenes or tumor suppressors. Here we report the molecular, biochemical, and functional characterization of KS1 (KRAB/zinc finger suppressor protein 1), a novel, ubiquitously expressed zinc finger gene initially isolated from a rat pancreas library. KS1 contains 10 C2H2 zinc fingers, a KRAB-A/B motif, and an ID sequence that has been shown previously to participate in growth factor-regulated gene expression. Northern blot analysis using pancreatic cell lines demonstrates that KS1 mRNA is inducible by serum and epidermal growth factor, suggesting a role for this gene in cell growth regulation. Biochemical analysis reveals that KS1 is a nuclear protein containing two transcriptional repressor domains, R1 and R2. R1 corresponds to the KRAB-A motif, whereas R2 represents a novel sequence. Transformation assays using NIH3T3 cells demonstrate that KS1 suppresses transformation by the potent oncogenes Ha-ras, Galpha12, and Galpha13. Deletion of the R1/ KRAB-A domain does not modify the transformation suppressive activity of KS1, whereas deletion of R2 abolishes this function. Thus, KS1 is a novel growth factor-inducible zinc finger transcriptional repressor protein with the potential to protect against neoplastic transformation induced by several oncogenes.

    Topics: 3T3 Cells; Amino Acid Sequence; Animals; Base Sequence; Cell Nucleus; Cell Transformation, Neoplastic; DNA-Binding Proteins; Epidermal Growth Factor; Gene Expression Regulation; Gene Library; Genes; Genes, ras; Genes, Tumor Suppressor; Genes, Wilms Tumor; Kruppel-Like Transcription Factors; Mice; Molecular Sequence Data; Multigene Family; Neoplasm Proteins; Nuclear Proteins; Pancreas; Pancreatic Neoplasms; Protein Conformation; Rats; Repressor Proteins; Sequence Alignment; Sequence Homology, Amino Acid; Transcription Factors; Transcription, Genetic; Tumor Cells, Cultured; Zinc Fingers

1998
Expression of an oncogenic mutant G alpha i2 activates Ras in Rat-1 fibroblast cells.
    FEBS letters, 1998, Nov-27, Volume: 440, Issue:1-2

    It has been reported that expression of the active mutant of heterotrimeric GTP-binding protein alpha subunit G alpha i2 transforms Rat-1 cells. However, the G alpha i2-mediated mitogenic signaling pathways remain to be elucidated. Here, we demonstrate that inducible expression of the active mutant of G alpha i2 (G alpha i2(Q205L)) activates Ras and c-Jun N-terminal kinase (JNK) in addition to extracellular signal-regulated kinase (ERK) in Rat-1 cells. Our findings suggest that Ras may play a critical role in the G alpha i2-induced transformation and G alpha i2 can transduce signals from the Gi-coupled receptor to JNK and ERK in certain types of mammalian cells.

    Topics: Animals; Calcium-Calmodulin-Dependent Protein Kinases; Cell Line; Cell Transformation, Neoplastic; Enzyme Activation; Epidermal Growth Factor; Escherichia coli; Fibroblasts; Flavonoids; GTP-Binding Protein alpha Subunit, Gi2; GTP-Binding Protein alpha Subunits, Gi-Go; GTP-Binding Proteins; Guanosine Triphosphate; JNK Mitogen-Activated Protein Kinases; Mitogen-Activated Protein Kinase 1; Mitogen-Activated Protein Kinases; Mitogens; Mutation; Phosphorylation; Proto-Oncogene Proteins; ras Proteins; Rats; Recombinant Fusion Proteins; Signal Transduction; Trypsin

1998
Expression of dominant negative Erk2 inhibits AP-1 transactivation and neoplastic transformation.
    Oncogene, 1998, Dec-31, Volume: 17, Issue:26

    The mitogen activated protein (MAP) kinases or extracellular signal-regulated kinases (Erks) are activated in response to Ras expression or exposure to tumor promoters or to growth factors, and have been implicated in AP-1 transactivation in some models. We have shown that tumor promoter induced activation of the transcription factor AP-1 is required for induced neoplastic transformation in the Balb/C JB6 cell model. Jun and Fos family protein levels have been found not to be limiting for AP-1 response. The present study asks whether activation of Erks1 and 2 is required for AP-1 transactivation and transformation of JB6 cells and whether Erks might be targeted for cancer prevention. Expression of either of two different dominant negative kinase inactive Erk2 mutants in transformation sensitive (P+) JB6 cells substantially inhibited the tumor promoter induced activation of Erks1 and 2 and of AP-1 measured by a collagenase-luciferase reporter. Multiple mutant Erk2 expressing clonal lines were also rendered non-responsive to induced neoplastic transformation. These observations, together with our recent finding attributing AP-1 non-responsiveness to Erk deficiency in a clonal line of transformation resistant (P-) cells, argue for a requirement for Erks1 and/or 2 activation in AP-1 transactivation in the mouse JB6 neoplastic progression model, and suggest the utility of Erks as a prevention target.

    Topics: Animals; Calcium-Calmodulin-Dependent Protein Kinases; Carcinogens; Cell Line; Cell Transformation, Neoplastic; Epidermal Cells; Epidermal Growth Factor; Gene Expression Regulation, Neoplastic; Genes, Dominant; Mice; Mice, Inbred BALB C; Mitogen-Activated Protein Kinase 1; Mitogen-Activated Protein Kinase 3; Mitogen-Activated Protein Kinases; Mutation; Phosphorylation; Recombinant Proteins; Tetradecanoylphorbol Acetate; Transcription Factor AP-1; Transcriptional Activation; Transfection

1998
Cripto: roles in mammary cell growth, survival, differentiation and transformation.
    Cell death and differentiation, 1998, Volume: 5, Issue:5

    Cripto-1 (Cr-1) protein, encoded by the teratocarcinoma-derived growth factor gene (TDGF-1), is highly correlated with transformation in breast cancer. Eighty-two percent of breast carcinomas express Cr-1 whereas it is undetected in normal human breast tissue. We confirmed and extended findings that Cr-1 protein is expressed during the pregnancy and lactating stages of normal murine mammary glands but is barely detectable in glands from virgin animals and is undetectable in involuted glands. Cr-1 was found to be expressed in CID 9 cells, a line of mammary epithelial cells derived from 14.5 day pregnant mice and we have used these cells to investigate the roles of this gene. Exogenous mouse Cr-1 expression from a retroviral vector caused CID 9 cells to grow at an increased rate and to increased cell densities compared to parental and control cells. CID 9 cells overexpressing Cr-1 did not differentiate efficiently. Infection of CID 9 cells with a Cr-1 antisense vector caused these cells to change in morphology, to grow slowly, to undergo apoptosis at a higher rate and to achieve lower saturation densities but the cells were still capable of differentiating. We concluded that Cr-1 is an autocrine growth factor for normal breast cells, that when over-expressed stimulates excessive cell proliferation at the expense of differentiation. In transplantation studies, Cr-1 over-expression stimulated the growth and survival of mammary cells, but did not stimulate tumorigenesis in vivo.

    Topics: Animals; Apoptosis; Cell Differentiation; Cell Division; Cell Line; Cell Survival; Cell Transformation, Neoplastic; Epidermal Growth Factor; Epithelial Cells; Female; Gene Expression Regulation; Growth Substances; In Situ Nick-End Labeling; Mammary Glands, Animal; Membrane Glycoproteins; Mice; Neoplasm Proteins; Oligonucleotides, Antisense; Pregnancy; Transfection

1998
AP-1-mediated invasion requires increased expression of the hyaluronan receptor CD44.
    Molecular and cellular biology, 1997, Volume: 17, Issue:2

    Fibroblasts transformed by Fos oncogenes display increased expression of a number of genes implicated in tumor cell invasion and metastasis. In contrast to normal 208F rat fibroblasts, Fos-transformed 208F fibroblasts are growth factor independent for invasion. We demonstrate that invasion of v-Fos- or epidermal growth factor (EGF)-transformed cells requires AP-1 activity. v-Fos-transformed cell invasion is inhibited by c-jun antisense oligonucleotides and by expression of a c-jun dominant negative mutant, TAM-67. EGF-induced invasion is inhibited by both c-fos and c-jun antisense oligonucleotides. CD44s, the standard form of a transmembrane receptor for hyaluronan, is implicated in tumor cell invasion and metastasis. We demonstrate that increased expression of CD44 in Fos- and EGF-transformed cells is dependent upon AP-1. CD44 antisense oligonucleotides reduce expression of CD44 in v-Fos- or EGF-transformed cells and inhibit invasion but not migration. Expression of a fusion protein between human CD44s and Aequorea victoria green fluorescent protein (GFP) in 208F cells complements the inhibition of invasion by the rat-specific CD44 antisense oligonucleotide. We further show that both v-Fos and EGF transformations result in a concentration of endogenous CD44 or exogenous CD44-GFP at the ends of pseudopodial cell extensions. These results support the hypothesis that one role of AP-1 in transformation is to activate a multigenic invasion program.

    Topics: Animals; Cell Line, Transformed; Cell Transformation, Neoplastic; Epidermal Growth Factor; Fibroblasts; Gene Expression Regulation; Green Fluorescent Proteins; Humans; Hyaluronan Receptors; Luminescent Proteins; Neoplasm Invasiveness; Oligonucleotides, Antisense; Oncogene Proteins v-fos; Proto-Oncogene Proteins c-fos; Proto-Oncogene Proteins c-jun; Rats; Recombinant Fusion Proteins; Transcription Factor AP-1

1997
Opposite effects of the p52shc/p46shc and p66shc splicing isoforms on the EGF receptor-MAP kinase-fos signalling pathway.
    The EMBO journal, 1997, Feb-17, Volume: 16, Issue:4

    Shc proteins are targets of activated tyrosine kinases and are implicated in the transmission of activation signals to Ras. The p46shc and p52shc isoforms share a C-terminal SH2 domain, a proline- and glycine-rich region (collagen homologous region 1; CH1) and a N-terminal PTB domain. We have isolated cDNAs encoding for a third Shc isoform, p66shc. The predicted amino acid sequence of p66shc overlaps that of p52shc and contains a unique N-terminal region which is also rich in glycines and prolines (CH2). p52shc/p46shc is found in every cell type with invariant reciprocal relationship, whereas p66shc expression varies from cell type to cell type. p66shc differs from p52shc/p46shc in its inability to transform mouse fibroblasts in vitro. Like p52shc/p46shc, p66shc is tyrosine-phosphorylated upon epidermal growth factor (EGF) stimulation, binds to activated EGF receptors (EGFRs) and forms stable complexes with Grb2. However, unlike p52shc/p46shc it does not increase EGF activation of MAP kinases, but inhibits fos promoter activation. The isolated CH2 domain retains the inhibitory effect of p66shc on the fos promoter. p52shc/p46shc and p66shc, therefore, appear to exert different effects on the EGFR-MAP kinase and other signalling pathways that control fos promoter activity. Regulation of p66shc expression might, therefore, influence the cellular response to growth factors.

    Topics: 3T3 Cells; Adaptor Proteins, Signal Transducing; Adaptor Proteins, Vesicular Transport; Amino Acid Sequence; Animals; Calcium-Calmodulin-Dependent Protein Kinases; Cell Transformation, Neoplastic; Cloning, Molecular; DNA, Complementary; Enzyme Activation; Epidermal Growth Factor; ErbB Receptors; Genes, fos; GRB2 Adaptor Protein; Humans; Mice; Molecular Sequence Data; Phosphorylation; Promoter Regions, Genetic; Proteins; RNA Splicing; Shc Signaling Adaptor Proteins; Signal Transduction; Src Homology 2 Domain-Containing, Transforming Protein 1; Tyrosine

1997
Expression of the activated p185erbB2 tyrosine kinase in human epithelial cells leads to MAP kinase activation but does not confer oncogenicity.
    Experimental cell research, 1997, Mar-15, Volume: 231, Issue:2

    Amplification of the c-erbB2 gene and overexpression of p185erbB2 is found in approximately one-third of primary breast and ovarian cancers and also in some colon carcinomas. Moreover, a single point mutation in erbB2(V 664 E) confers transforming potential to erbB2 in NIH3T3 cells, even when expressed at low levels. To examine the transformation potential of erbB2 or erbB2(V-E) in colon epithelial cells, we have transfected a nontumorigenic clone of SW 613-S cells with either wild-type p185erbB2 or mutated p185erbB2(V-E). In contrast to p185erbB2, p185erbB2(V-E) associated constitutively with members of the Shc protein family, leading to phosphorylation of Shc and to stimulation of mitogen-activated protein kinase (MAP kinase). However, constitutive activation of MAP kinase activation in p185erbB2(V-E) expressing cells did not result in a tumorigenic phenotype. In addition, p185erbB2(V-E) expressing cells displayed a reduced ability to grow in soft agar compared to the parental cell line. In contrast these transfected cells were able to grow in three-dimensional collagen gels, whereas parental cells were not. Thus, expression of erbB2(V-E) in SW 613-S cells induced multiple changes in intracellular signaling and in growth requirement phenotype, particularly in response to the extracellular environment.

    Topics: Adaptor Proteins, Signal Transducing; Adaptor Proteins, Vesicular Transport; Adenocarcinoma; Animals; Calcium-Calmodulin-Dependent Protein Kinases; Cell Differentiation; Cell Division; Cell Transformation, Neoplastic; Collagen; Colonic Neoplasms; Enzyme Activation; Enzyme Induction; Epidermal Growth Factor; Epithelial Cells; Epithelium; Female; Gels; Humans; Intestinal Mucosa; Mice; Neoplasm Proteins; Neoplasm Transplantation; Oncogenes; Phosphorylation; Point Mutation; Protein Processing, Post-Translational; Proteins; Receptor, ErbB-2; Recombinant Fusion Proteins; Shc Signaling Adaptor Proteins; Src Homology 2 Domain-Containing, Transforming Protein 1; Transfection; Tumor Cells, Cultured

1997
EGF receptor binding and transformation by v-cbl is ablated by the introduction of a loss-of-function mutation from the Caenorhabditis elegans sli-1 gene.
    Oncogene, 1997, May-08, Volume: 14, Issue:18

    The 120 kD product of the c-cbl oncogene is rapidly tyrosine phosphorylated and recruited to the EGF receptor following ligand binding. Cbl's oncogenic potential is activated by a large carboxy-terminal truncation that generated v-cbl and removes the Ring finger and proline-rich SH3-binding domains. Here we show that this truncation reveals a novel and highly conserved domain that can interact directly with the EGF receptor in a phosphorylation dependent manner. Furthermore we demonstrate that the v-cbl domain is not utilized by c-cbl for recruitment to the receptor since this binding property is not evident in c-cbl constructs with proline domain deletions, and it is only revealed following deletion of the Ring finger. We also analyse a loss-of-function mutation from the C. elegans homologue, sli-1, and show that the corresponding mutation in v-cbl ablates transformation and EGF receptor association. Thus our findings provide further evidence that v-cbl possesses a novel and evolutionarily conserved phosphotyrosine binding domain and that the dual capability of EGF receptor binding by cbl involves two distinct mechanisms. In addition these findings raise the possibility that v-cbl may transform by competing with c-cbl for phosphorylated binding sites on activated receptor complexes.

    Topics: 3T3 Cells; Amino Acid Sequence; Animals; Binding Sites; Caenorhabditis elegans; Caenorhabditis elegans Proteins; Cell Transformation, Neoplastic; Conserved Sequence; Epidermal Growth Factor; ErbB Receptors; Helminth Proteins; Mice; Mutation; Oncogene Protein v-cbl; Phosphorylation; Proline; Proto-Oncogene Proteins; Proto-Oncogene Proteins c-cbl; Rabbits; Retroviridae Proteins, Oncogenic; Tyrosine; Ubiquitin-Protein Ligases

1997
Insulin-like growth factor I receptor signaling in transformation by src oncogenes.
    Molecular and cellular biology, 1997, Volume: 17, Issue:7

    R- cells, a line of mouse embryo fibroblasts with a targeted disruption of the insulin-like growth factor I (IGF-I) receptor genes, are refractory to transformation by several viral and cellular oncogenes. Using colony formation in soft agar as a measure of full transformation, we report here that R- cells can be transformed by v-src, although they still cannot be transformed by the activated c-src527 (mutation at tyrosine 527 to phenylalanine), which readily transforms mouse embryo cells with a wild-type number of IGF-I receptors (W cells). Although v-src is a more potent inducer of tyrosine phosphorylation than c-src527, the extent of phosphorylation of either insulin receptor substrate 1 or Shc, two of the major substrates of the IGF-I receptor, does not seem sufficiently different to explain the qualitative difference in soft agar growth. v-src, however, is considerably more efficient than c-src527 in its ability to tyrosyl phosphorylate, in R- cells, the focal adhesion kinase, Stat1, and p130cas. These results indicate that v-src, but not c-src527, can bypass the requirement for a functional IGF-I receptor in the full transformation of mouse embryo fibroblasts and suggest that qualitative and quantitative differences between the two oncogenes can be used to identify some of the signals relevant to the mechanism(s) of transformation.

    Topics: 3T3 Cells; Adaptor Proteins, Signal Transducing; Adaptor Proteins, Vesicular Transport; Animals; Cell Adhesion Molecules; Cell Division; Cell Transformation, Neoplastic; Cells, Cultured; DNA-Binding Proteins; Epidermal Growth Factor; Focal Adhesion Kinase 1; Focal Adhesion Protein-Tyrosine Kinases; Genes, src; Insulin Receptor Substrate Proteins; Mice; Oncogenes; Phosphoproteins; Phosphorylation; Phosphotyrosine; Protein-Tyrosine Kinases; Proteins; Receptor, IGF Type 1; Shc Signaling Adaptor Proteins; Signal Transduction; Src Homology 2 Domain-Containing, Transforming Protein 1; STAT1 Transcription Factor; Trans-Activators; Transfection

1997
Inositol hexaphosphate inhibits cell transformation and activator protein 1 activation by targeting phosphatidylinositol-3' kinase.
    Cancer research, 1997, Jul-15, Volume: 57, Issue:14

    Inositol hexaphosphate (InsP6) is the most abundant inositol phosphate found in plants. In mammalian cells, the concentrations of InsP6 are between 10 and 100 microM. Previous work has indicated that InsP6 is an effective cancer chemopreventive and chemotherapeutic agent. However, the molecular mechanisms involved in the inhibition of carcinogenesis by InsP6 remain unclear. In this study, we investigated the influence of InsP6 on tumor promoter-induced cell transformation and signal transduction pathways leading to activator protein 1 activation, which is considered to play a crucial role in tumor promotion. InsP6 markedly blocks epidermal growth factor-induced phosphatidylinositol-3 (PI-3) kinase activity in a dose-dependent manner in JB6 cells and directly in vitro. Blocking PI-3 kinase activity by InsP6 profoundly impairs epidermal growth factor- or phorbol ester-induced JB6 cell transformation and extracellular signal-regulated protein kinases activation, as well as activator protein 1 activation. These results provide the first evidence that the molecular mechanism of InsP6 antitumor promotion effect targets and blocks PI-3 kinase activation and demonstrate that PI-3 kinase can serve as a molecular target for the development of cancer chemopreventive agents.

    Topics: Animals; Anticarcinogenic Agents; Cell Line; Cell Transformation, Neoplastic; Epidermal Growth Factor; Mice; Phosphatidylinositol 3-Kinases; Phosphotransferases (Alcohol Group Acceptor); Phytic Acid; Tetradecanoylphorbol Acetate; Transcription Factor AP-1

1997
[Inhibition of EGF and TGF-beta dependent transformation of NRK23 cells by Crk II-23 mutant].
    [Hokkaido igaku zasshi] The Hokkaido journal of medical science, 1997, Volume: 72, Issue:4

    Adaptor proteins participate in many signaling pathways from cell surface receptors. Crk protein was the first example of the adaptor protein. We have examined the function of Crk II mutant, Crk II-23. The Crk II-23 mutant contains two amino-acid substitutions in the carboxyl-terminal SH3 domain and is known to inhibit the transformation of NRK cells induced by epidermal growth factor (EGF) and transforming growth factor-beta (TGF-beta). There was no remarkable difference between Crk II and Crk II-23 in EGF-dependent binding to EGF receptor (EGFR). However, in contrast to the wild-type Crk II, the Crk II-23 mutant bound to EGFR in quiescent NIH 3T3 cells. To clarify the difference, both the Crk II and Crk II-23, proteins were expressed in E. coli and examined their binding capacity in vitro. They bound to EGFR from EGF-stimulated NIH 3T3 cells in vitro to a similar extent. Expression of Crk II-23 in NIH 3T3 cells did not affect the binding of bacterially expressed Crk II and Crk II-23 to EGFR. These results suggest that post-translational modification of Crk II-23, such as physical association to cellular proteins, induces binding of Crk II-23 to EGFR in quiescent cells. We also demonstrated that mutation of either the SH2 or the SH3 domain abolished the anti-oncogenic activity of Crk II-23, although both mutants bound to EGFR in the quiescent cells. From these results, it could be concluded that persistent signaling through Crk II-23 bound to EGFR is responsible for the suppression of transformation by EGF and TGF-beta.

    Topics: Animals; Cell Transformation, Neoplastic; Cells, Cultured; Epidermal Growth Factor; ErbB Receptors; Humans; Mutation; Proto-Oncogene Proteins; Proto-Oncogene Proteins c-crk; Signal Transduction; Transforming Growth Factor beta

1997
Loss of response to epidermal growth factor and retinoic acid accompanies the transformation of human prostatic epithelial cells to tumorigenicity with v-Ki-ras.
    Carcinogenesis, 1997, Volume: 18, Issue:8

    Growth factor-independent proliferation and loss of response to differentiation factors are believed to be critical elements in carcinogenesis. We have developed an in vitro model of human prostatic carcinogenesis by the introduction of SV40 DNA into normal prostatic epithelial cells to create a transformed, immortal cell line, pRNS-1-1. This non-tumorigenic cell line responded similarly to normal prostatic epithelial cells to most growth- and differentiation-regulatory factors, with the notable exception of loss of response to the inhibitory factor 1,25-dihydroxyvitamin D3. In this study, we describe the introduction of the ras oncogene into pRNS-1-1 cells to create a tumorigenic cell line, pRNS-1-1/ras. In addition to an attenuated response to 1,25-dihydroxyvitamin D3, these cells also became unresponsive to retinoic acid and gained the ability to undergo clonal proliferation in the absence of epidermal growth factor (EGF). EGF-independent growth could not be linked to the production of autocrine transforming growth factor-alpha, but instead was likely due to sustained signaling by the ras oncogene, bypassing ligand-activation of the EGF receptor. Ligand-independent proliferation, coupled with the loss of response to the growth-inhibitory and differentiation agent retinoic acid, may be important elements in the conversion of human prostatic epithelial cells to tumorigenicity.

    Topics: Animals; Calcitriol; Cell Division; Cell Transformation, Neoplastic; Cells, Cultured; Epidermal Growth Factor; Epithelium; Fibroblast Growth Factor 1; Genes, ras; Humans; Keratins; Male; Mice; Mice, Nude; Neoplasm Transplantation; Prostate; Somatomedins; Transforming Growth Factor alpha; Tretinoin; Tumor Necrosis Factor-alpha

1997
Mutations of critical amino acids affect the biological and biochemical properties of oncogenic A-Raf and Raf-1.
    Oncogene, 1997, Aug-28, Volume: 15, Issue:9

    The catalytic domains of the Raf family of protein kinases (deltaRaf) differ in their ability to activate MEK in vitro and in vivo and in their ability to oncogenically transform mammalian cells. The kinase domain of B-Raf is more active than the equivalent portion of Raf-1 which in turn is more active than A-Raf. In Raf-1 the phosphorylation or mutation to aspartic acid of two key tyrosine residues upstream of the ATP binding site has been demonstrated to significantly potentiate catalytic activity. In A-Raf the analogous amino acids are also tyrosine whereas in B-Raf they are aspartic acid. To determine if these differences in amino acid sequence influence the relative catalytic activity of the Raf kinase domains we constructed forms of deltaA-Raf, deltaB-Raf and deltaRaf-1 that encode either aspartic acid [DD], phenylalanine [FF] or tyrosine [YY] at these positions. These proteins were expressed both in mammalian cells as fusions with the hormone binding domain of the estrogen receptor and as epitope-tagged proteins in Sf9 insect cells to test their oncogenic and catalytic potentials. When expressed in Rat1 or 3T3 cells in the presence of hormone all of the deltaRaf-1:ER and deltaA-Raf:ER proteins were transforming with the exception of the [FF] form of deltaA-Raf. In general the [DD] forms of the deltaRaf-1:ER and deltaA-Raf:ER proteins were the most potently oncogenic which correlated with their ability to elicit activation of the MAP kinase pathway. Consistent with the transformation data, the catalytic activity of the [DD] forms of deltaA-Raf:ER and deltaRaf-1:ER was about ten times greater than the cognate [FF] and [YY] forms of the proteins. By contrast all of the deltaB-Raf:ER proteins were highly transforming and deltaB-Raf catalytic activity was largely unaffected by mutation of the aforementioned aspartic acids to either tyrosine or phenylalanine. Similar results were obtained with epitope-tagged forms of deltaA-Raf, deltaB-Raf and deltaRaf-1 expressed in Sf9 cells. These data provide support for the model that key tyrosine residues in the protein kinase domains of A-Raf and Raf-1 are important in the regulation of catalytic activity. In addition they demonstrate that the higher intrinsic activity of B-Raf cannot be explained simply by the presence of aspartic acids at the analogous positions.

    Topics: Amino Acids; Animals; Baculoviridae; Calcium-Calmodulin-Dependent Protein Kinases; Catalysis; Cell Line; Cell Transformation, Neoplastic; Enzyme Activation; Epidermal Growth Factor; Gene Expression Regulation, Neoplastic; Heparin; Humans; Mice; Mutagenesis, Site-Directed; Protein Serine-Threonine Kinases; Proto-Oncogene Proteins; Proto-Oncogene Proteins c-raf; Receptors, Estrogen; Spodoptera

1997
Differential roles of IRS-1 and SHC signaling pathways in breast cancer cells.
    International journal of cancer, 1997, Sep-04, Volume: 72, Issue:5

    Several polypeptide growth factors stimulate breast cancer growth and may be involved in tumor progression. However, the relative importance of diverse growth factor signaling pathways in the development and maintenance of the neoplastic phenotype is largely unknown. The activation of such growth factor receptors as the insulin-like growth factor I receptor (IGF-I R), erbB-type receptors (erbB Rs) and FGF receptors (FGF Rs) controls the phenotype of a model breast cancer cell line MCF-7. To evaluate the function of 2 post-receptor signaling molecules, insulin receptor substrate-1 (IRS-1) (a major substrate of the IGF-IR) and SHC (a common substrate of tyrosine kinase receptors), we developed several MCF-7-derived cell clones in which the synthesis of either IRS-1 or SHC was blocked by antisense RNA. In MCF-7 cells, down-regulation of IRS-1 by 80-85% strongly suppressed anchorage-dependent and -independent growth and induced apoptotic cell death under growth factor- and estrogen-reduced conditions. The reduction of SHC levels by approximately 50% resulted in the inhibition of monolayer and anchorage-independent growth but did not decrease cell survival. Importantly, cell aggregation and the ability of cells to survive on the extracellular matrix were inhibited in MCF-7/anti-SHC clones, but not in MCF-7/anti-IRS-1 clones. Cell motility toward IGF was not attenuated in any of the tested cell lines, but motility toward EGF was decreased in MCF-7/anti-SHC clones. Our results suggest that in MCF-7 cells: 1) both IRS-1 and SHC are implicated in the control of monolayer and anchorage-independent growth; 2) IRS-1 is critical to support cell survival; 3) SHC is involved in EGF-dependent motility; and 4) normal levels of SHC, but not IRS-1, are necessary for the formation and maintenance of cell-cell interactions.

    Topics: Adaptor Proteins, Signal Transducing; Adaptor Proteins, Vesicular Transport; Apoptosis; Blotting, Western; Breast Neoplasms; Cell Adhesion; Cell Aggregation; Cell Division; Cell Movement; Cell Survival; Cell Transformation, Neoplastic; Epidermal Growth Factor; Female; Humans; Insulin Receptor Substrate Proteins; Phosphoproteins; Proteins; RNA, Antisense; Shc Signaling Adaptor Proteins; Signal Transduction; Somatomedins; Src Homology 2 Domain-Containing, Transforming Protein 1; Tumor Cells, Cultured

1997
Modulation of epidermal growth factor signal transduction by linoleic acid metabolites.
    Advances in experimental medicine and biology, 1997, Volume: 407

    Topics: Animals; Calcium-Calmodulin-Dependent Protein Kinases; Cell Division; Cell Line, Transformed; Cell Transformation, Neoplastic; Cricetinae; Embryo, Mammalian; Enzyme Activation; Epidermal Growth Factor; ErbB Receptors; Fibroblasts; Hybrid Cells; Linoleic Acid; Linoleic Acids; Lipid Peroxides; Mesocricetus; Phosphorylation; Protein Processing, Post-Translational; Signal Transduction; Stimulation, Chemical

1997
The role of linoleic acid metabolism in the proliferative response of cells overexpressing the erbB-2/HER2 oncogene.
    Advances in experimental medicine and biology, 1997, Volume: 407

    Topics: Animals; Arachidonic Acid; Cell Division; Cell Line, Transformed; Cell Transformation, Neoplastic; Cricetinae; Cyclooxygenase Inhibitors; Dexamethasone; DNA Replication; Epidermal Growth Factor; Fibroblasts; Hydroxyeicosatetraenoic Acids; Indomethacin; Linoleic Acid; Linoleic Acids; Lipoxygenase; Masoprocol; Mesocricetus; Receptor, ErbB-2; Recombinant Fusion Proteins; Transfection

1997
Inhibition of tumor promoter-induced activator protein 1 activation and cell transformation by tea polyphenols, (-)-epigallocatechin gallate, and theaflavins.
    Cancer research, 1997, Oct-01, Volume: 57, Issue:19

    (-)-Epigallocatechin gallate (EGCG) and theaflavins are believed to be key active components in tea for the chemoprevention against cancer. However, the molecular mechanisms by which EGCG and theaflavins block carcinogenesis are not clear. We have used the JB6 mouse epidermal cell line, a system that has been used extensively as an in vitro model for tumor promotion studies, to examine the anti-tumor promotion effects of EGCG and theaflavins at the molecular level. EGCG and theaflavins inhibited epidermal growth factor- or 12-O-tetradecanoylphorbol-13-acetate-induced cell transformation in a dose-dependent manner. At the dose range (5-20 microM) that inhibited cell transformation, EGCG and theaflavins also inhibited AP-1-dependent transcriptional activity and DNA binding activity. The inhibition of AP-1 activation occurs through the inhibition of a c-Jun NH2-terminal kinase-dependent, but not an extracellular signal-regulated protein kinase (Erk) 1-dependent or Erk2-dependent, pathway. Because the transcription factor AP-1 is important for tumor promoter-induced neoplastic transformation, the inhibitory effects on AP-1 activation by EGCG and theaflavins may further explain the anti-tumor promotion action of these tea constituents.

    Topics: Animals; Anticarcinogenic Agents; Biflavonoids; Catechin; Cell Transformation, Neoplastic; Chemoprevention; Epidermal Growth Factor; Mice; Phosphorylation; Protein Processing, Post-Translational; Protein-Tyrosine Kinases; Proto-Oncogene Proteins c-jun; Signal Transduction; Tea; Tetradecanoylphorbol Acetate; Transcription Factor AP-1; Transcriptional Activation

1997
In vitro pancreatic ductal cell carcinogenesis.
    International journal of cancer, 1997, Sep-17, Volume: 72, Issue:6

    Our experiments were designed to identify initial biochemical and biological changes that occur during pancreatic carcinogenesis. TAKA-1, an immortal hamster pancreatic ductal cell line, was treated in vitro for up to 11 weeks with the pancreatic carcinogen N-nitorosobis(2-oxopropyl)amine (BOP). These treated cells were designated TAKA-1 + BOP. The growth of TAKA-1 and TAKA-1 + BOP cell lines was investigated in soft agar and in hamsters intradermally. The resulting tumor from TAKA-1 + BOP was re-cultured in vitro and designated TAKA-1 + BOP-T. Mutation of c-K-ras and p53 oncogenes, chromosomal changes, expression of transforming growth factor alpha (TGF-alpha) and epidermal growth factor (EGF) receptor and several biochemical markers were examined in all cell lines. TAKA-1 + BOP but not TAKA-1 cells grew in soft agar and produced an invasive tumor in vivo. However, there were no differences in cell growth rate, DNA flow cytometry, or immunohistochemical findings between the non-transformed and transformed cells. TAKA-1, TAKA-1 + BOP and TAKA-1 + BOP-T cells all expressed mRNA of TGF-alpha and EGF receptor in a comparable pattern. DNA sequence analysis following polymerase chain reaction showed that neither TAKA-1 nor TAKA-1 + BOP cells has a mutation of c-K-ras or p53. Karyotype analysis demonstrated that TAKA-1 + BOP cells had more chromosomal abnormalities compared with TAKA-1 cells. Mutation of c-K-ras and p53 was not essential for carcinogenesis in hamster pancreatic ductal cells in vitro. In conclusion, immortality of the TAKA-1 cells caused expression of TGF-alpha to the same extent as in malignant cells. Chromosomal and ultrastructural patterns were the only differences detected between the non-transformed and BOP-transformed cells.

    Topics: Amino Acid Sequence; Animals; Base Sequence; Carcinogens; Cell Division; Cell Line; Cell Transformation, Neoplastic; Cricetinae; Epidermal Growth Factor; ErbB Receptors; Genes, p53; Genes, ras; Karyotyping; Keratins; Kinetics; Mice; Microscopy, Electron; Molecular Sequence Data; Mutagenesis; Nitrosamines; Pancreatic Ducts; Pancreatic Neoplasms; Rats; Sequence Alignment; Transforming Growth Factor alpha

1997
[Growth hormones and oncogenes in mammary adenocarcinomas induced by medroxyprogesterone acetate in BALB/c mice].
    Medicina, 1997, Volume: 57 Suppl 2

    We have studied the involvement of growth factors (GF), their receptors (GF-R) and oncogenes in modulating tumor growth in the medroxyprogesterone acetate (MPA)-induced mammary tumor model in BALB/c mice. We demonstrated the presence of both ligands of the insulin-like growth factor family (IGF-I, IGF-II) and the two types of receptors (IGF-RI, IGF-RII). MPA upregulated IGF-II mRNA and protein levels in hormone-dependent lines (MPA-D). The progression to a hormone-independent phenotype was accompanied by a high constitutive expression of IGF-II and by a significant decrease in IGF-IIR number. An antisense strategy used to evaluate the role of IGF in the MPA-induced growth of epithelial MPA-D cells showed that IGF mediate progestin-induced mammary tumor growth by autocrine/intracrine pathways. We also studied the role of heregulins (HRG), the recently identified ligands for the c-erbB3 and c-erbB4 oncogenes. HRG mRNA expression was restricted to tumors of ductal origin. MPA induced an in vivo up-regulation of HRG expression. Finally, we also found that MPA may be exerting its proliferative effect on MPA-D lines by inhibiting the expression of transforming growth factor beta 1, (TGF-beta 1) and the lack of expression of TGF-beta 1 in hormone-independent tumors may be related to the acquisition of autonomous growth.

    Topics: Adenocarcinoma; Animals; Cell Transformation, Neoplastic; Epidermal Growth Factor; Female; Growth Substances; Insulin-Like Growth Factor I; Insulin-Like Growth Factor II; Mammary Neoplasms, Experimental; Medroxyprogesterone Acetate; Mice; Mice, Inbred BALB C; Oncogenes; Receptors, Growth Factor; Transforming Growth Factor beta

1997
ras effector loop mutations that dissociate p120GAP and neurofibromin interactions.
    Molecular carcinogenesis, 1996, Volume: 15, Issue:1

    ras proteins are positively regulated by nucleotide exchange factors and negatively regulated by GTPase-activating proteins (GAPs). Two GAPs have been found in mammalian cells, p120GAP and neurofibromin, the product of the type 1 neurofibromatosis (NF1) gene. A library of substitutions in the effector loop region of ras in an Escherichia coli plasmid expression system was screened for c-Ha-ras species with altered GAP interactions. Several substitutions preferentially disrupted the interaction of ras with p120GAP as compared with the interaction with the recombinant GAP-related domain of neurofibromin (NF1-GRD). The most extreme example, Tyr32His, encoded a ras species that was unaffected by p120GAP but was stimulated normally by NF1-GRD. Tyr32His was weakly transforming in Rat2 cells. Tyr32His ras was primarily GDP-bound in quiescent Rat2 cells, although it rapidly associated with GTP after treatment of cells with epidermal growth factor. These results show that the NF1 product has less stringent requirements than p120GAP for ras effector domain structure and that negative regulation of ras can be achieved in rat fibroblasts by the product of NF1.

    Topics: Animals; Cell Division; Cell Transformation, Neoplastic; Cells, Cultured; Epidermal Growth Factor; GTP-Binding Proteins; GTPase-Activating Proteins; Guanine Nucleotides; Guanosine Triphosphate; Humans; Neurofibromin 1; Point Mutation; Proteins; Proto-Oncogene Proteins p21(ras); ras GTPase-Activating Proteins; Rats; Recombinant Fusion Proteins; Signal Transduction; Structure-Activity Relationship

1996
Trisomy 7p and malignant transformation of human breast epithelial cells following epidermal growth factor withdrawal.
    Cancer research, 1996, May-01, Volume: 56, Issue:9

    We have reported previously on the first spontaneously immortalized, nonmalignant human breast epithelial cell line, HMT-3522, which is entirely dependent on exogenous epidermal growth factor (EGF). In passage 118, cells were adapted to grow in medium without EGF and a new growth-transformed subline, HMT-3522/gt-1, was generated and propagated at high growth rate without exogenous EGF (Madsen et al., Cancer Res., 52: 1210-1217, 1992). Here we have used this subline and the continuum of the parent line, HMT-3522/wt, to pose the question whether a relevant change in a physiological parameter of the microenvironment will induce malignant transformation. The two cell lines were cultured under identical conditions with the only exception that EGF was omitted in the medium for gt-1. Initially, wt and gt-1 were identical in terms of karyotype as well as morphology, growth rate, and protein expression as revealed by two-dimensional gel electrophoresis. A highly dramatic shift to phenotype was observed in passage 238 when the gt-1 line became tumorigenic in nude mice. After two mouse-culture passages, the resulting malignantly transformed cell line (HMT-3522/mt-1) was refractory to the growth-modulating effect of EGF and presented an extra copy of a chromosome marker, 7q-, as the only cytogenetic difference from the gt-1. Our results suggest that microenvironmental cues are powerful factors in the induction of malignancy. A major role of EGF receptor in the malignant transformation is emphasized by loss of EGF sensitivity and acquisition of an extra chromosome 7p harboring the EGF receptor gene. We hypothesize that during premalignant hyperplasia, a population of EGF/transforming growth factor alpha autonomous epithelial cells in situ may develop as a consequence of local transforming growth factor alpha deprivation, a condition reflected in the culture model as autonomy after EGF withdrawal.

    Topics: Animals; Breast Neoplasms; Cell Division; Cell Transformation, Neoplastic; Chromosomes, Human, Pair 7; Culture Media; Epidermal Growth Factor; Humans; Immunoenzyme Techniques; Karyotyping; Mice; Trisomy; Tumor Cells, Cultured

1996
Activation of the epidermal growth factor receptor signal transduction pathway stimulates tyrosine phosphorylation of protein kinase C delta.
    The Journal of biological chemistry, 1996, Mar-08, Volume: 271, Issue:10

    The expression of an oncogenic rasHa gene in epidermal keratinocytes stimulates the tyrosine phosphorylation of protein kinase C delta and inhibits its enzymatic activity (Denning, M. F., Dlugosz, A. A., Howett, M. K., and Yuspa, S. H. (1993) J. Biol. Chem. 268, 26079-26081). Keratinocytes expressing an activated rasHa gene secrete transforming growth factor alpha (TGFalpha) and have an altered response to differentiation signals involving protein kinase C (PKC). Because the neoplastic phenotype of v-rasHa expressing keratinocytes can be partially mimicked in vitro by chronic treatment with TGF alpha and the G protein activator aluminum fluoride (AlF4-), we determined if TGF alpha or AlF4- could induce tyrosine phosphorylation of PKCdelta. Treatment of primary keratinocyte cultures for 4 days with TGFalpha induced tyrosine phosphorylation of PKCdelta, whereas AlF4- only slightly stimulated PKCdelta tyrosine phosphorylation. The PKCdelta that was tyrosine-phosphorylated in response to TGFalpha had reduced activity compared with the nontyrosine-phosphorylated PKCdelta. Treatment of keratinocytes expressing a normal epidermal growth factor receptor (EGFR) with TGFalpha or epidermal growth factor for 5 min induced PKCdelta tyrosine phosphorylation. This acute epidermal growth factor treatment did not induce tyrosine phosphorylation of PKCdelta in keratinocytes isolated from waved-2 mice that have a defective epidermal growth factor receptor. In addition, the level of PKCdelta tyrosine phosphorylation in v-rasHa-transduced keratinocytes from EGFR null mice was substantially lower than in v-rasHa transduced wild type cells, suggesting that activation of the EGFR is important for PKC delta tyrosine phosphorylation in ras transformation. However, purified EGFR did not phosphorylate recombinant PKC delta in vitro, whereas members of the Src family (c-Src, c-Fyn) and membrane preparations from keratinocytes did. Furthermore, clearing c-Src or c-Fyn from keratinocyte membrane lysates decreased PKCdelta tyrosine phosphorylation, and c-Src and c-Fyn isolated from keratinocytes treated with TGFalpha had increased kinase activity. Acute or chronic treatment with TGFalpha did not induce significant PKCdelta translocation in contrast to the phorbol ester 12-O-tetradecanoylphorbol-13-acetate, which induced both translocation and tyrosine phosphorylation of PKCdelta. This suggests that TGFalpha-induced tyrosine phosphorylation of PKC delta results from the activation of a t

    Topics: Alleles; Aluminum Compounds; Animals; Animals, Newborn; Cell Line; Cell Transformation, Neoplastic; Epidermal Growth Factor; ErbB Receptors; Fluorides; Gene Expression; Genes, ras; GTP-Binding Proteins; Isoenzymes; Keratinocytes; Mice; Mice, Inbred BALB C; Mice, Mutant Strains; Mutagenesis; Phosphorylation; Phosphotyrosine; Protein Kinase C; Protein-Tyrosine Kinases; Proto-Oncogene Proteins; Proto-Oncogene Proteins c-fyn; Proto-Oncogene Proteins pp60(c-src); Recombinant Proteins; Signal Transduction; Spodoptera; Transfection; Transforming Growth Factor alpha; Tyrosine

1996
The multiple endocrine neoplasia type 2B point mutation alters long-term regulation and enhances the transforming capacity of the epidermal growth factor receptor.
    The Journal of biological chemistry, 1996, Mar-08, Volume: 271, Issue:10

    The RET proto-oncogene encodes a member of the receptor tyrosine kinase family. Multiple endocrine neoplasia type 2B (MEN 2B) is caused by the mutation of a conserved methionine to a threonine in the catalytic domain of the RET kinase. When the MEN 2B point mutation was introduced into the epidermal growth factor (EGF) receptor (M857T EGFR), the intrinsic tyrosine kinase activity of the mutant receptor was similar to that of wild-type EGF receptor and remained ligand-dependent. However, the mutant receptor showed an enhanced transforming capacity compared to the wild-type receptor as judged by its ability to mediate the growth of NIH 3T3 cells in soft agar. Using the oriented peptide library approach to examine substrate specificity, the M857T mutation was found to be associated with a decrease in the selectivity of the receptor for Phe and an increase in the selectivity for acidic residues at the P + 1 position as compared to wild-type EGF receptor. Short-term responses to EGF were similar in cells expressing wild-type and M857T EGF receptors. However, significant differences in receptor down-regulation were observed between the two receptors. These data demonstrate that the MEN 2B point mutation alters the substrate specificity of receptor tyrosine kinases and suggest that the enhanced oncogenesis associated with the MEN 2B mutation may be due in part to alterations in receptor regulation.

    Topics: 3T3 Cells; Amino Acid Sequence; Animals; Base Sequence; Cell Division; Cell Line; Cell Transformation, Neoplastic; DNA Primers; Down-Regulation; Drosophila Proteins; Epidermal Growth Factor; ErbB Receptors; Gene Expression Regulation, Neoplastic; Gene Library; Humans; Kinetics; Mice; Molecular Sequence Data; Multiple Endocrine Neoplasia Type 2b; Mutagenesis, Site-Directed; Peptides; Phosphorylation; Plasmids; Point Mutation; Proto-Oncogene Mas; Proto-Oncogene Proteins; Proto-Oncogene Proteins c-ret; Proto-Oncogenes; Receptor Protein-Tyrosine Kinases; Recombinant Proteins; Substrate Specificity; Tumor Cells, Cultured

1996
Transformation of NIH 3T3 cells by HER3 or HER4 receptors requires the presence of HER1 or HER2.
    The Journal of biological chemistry, 1996, Feb-16, Volume: 271, Issue:7

    Members of the epidermal growth factor receptor (EGFR) subfamily of receptor protein tyrosine kinases have been implicated in the pathogenesis of various malignancies. The ability of one EGFR subfamily member to influence, or function synergistically with, another is likely to be a general feature of these receptors. To assess the role of receptor heterodimerization, we analyzed the ability of Neu differentiation factor (NDF) to induce cell growth and transformation of NIH 3T3 cells transfected with different combinations of the EGFR subfamily of receptors. NDF induced mitogenesis, but not transformation, of cells expressing either HER3 or HER4 alone. However, NDF-induced cell transformation was observed when either HER1 or HER2 was coexpressed with HER3 or HER4. In analogous receptor phosphorylation experiments, NDF-induced transphosphorylation appears to be correlated with synergistic transformation of NIH 3T3 cells. Interestingly, transphosphorylation between HER1 and HER4 can be stimulated by either EGF or NDF.

    Topics: 3T3 Cells; Animals; Cell Division; Cell Transformation, Neoplastic; Epidermal Growth Factor; ErbB Receptors; Glycoproteins; Kinetics; Mice; Nerve Growth Factors; Neuregulins; Phosphorylation; Proto-Oncogene Proteins; Receptor, ErbB-2; Receptor, ErbB-3; Receptor, ErbB-4; Transfection

1996
Transfection with a CRIPTO anti-sense plasmid suppresses endogenous CRIPTO expression and inhibits transformation in a human embryonal carcinoma cell line.
    International journal of cancer, 1996, May-16, Volume: 66, Issue:4

    CRIPTO is a member of the epidermal growth factor (EGF) gene family originally isolated from undifferentiated human NTERA2 clone D1 (NT2D1) multipotent embryonal carcinoma cells. Retinoic acid (RA) treatment of NT2D1 cells leads to a neuronal differentiation program and to concomitant loss of CRIPTO mRNA expression. To assess the role of CRIPTO in the control of NT2D1 cell growth or differentiation, these cells were treated with 3 anti-sense oligodeoxynucleotides complementary to the 5' end of the human CRIPTO mRNA. A dose-dependent inhibition of monolayer and soft agar growth was observed with each of these CRIPTO anti-sense oligodeoxynucleotides but not with a control oligodeoxynucleotide of random sequence or with the 3 corresponding CRIPTO sense oligodeoxynucleotides. In addition, NT2D1 cells were transfected with a recombinant expression vector containing a 918-bp coding fragment of the human CRIPTO cDNA in the 3' to 5' orientation. NT2D1 CRIPTO anti-sense transfectants exhibited a significantly reduced endogenous CRIPTO mRNA and protein, a 4- to 5-fold decrease in growth rate in monolayer and a 50-70% reduction in cloning efficiency in soft agar as compared with NT2D1 parental cells or with NT2D1 cells transfected with a plasmid containing the neomycin-resistance gene alone (NT2D1 neo cells). Finally, we examined the expression of immunophenotypic markers that are modulated during the differentiation of NT2D1 cells following RA treatment. The globoseries stage-specific embryonic antigen-3 recognized by the monoclonal antibody (MAb) SSEA-3 was expressed in 60% of undifferentiated parental NT2D1 or NT2D1 neo cells and in only 20% of NT2D1 CRIPTO anti-sense transfectants, whereas it was down-regulated in all cell lines following RA treatment. A neuroectodermal antigen recognized by the A2B5 MAb, which was not expressed in parental NT2D1, in NT2D1 neo or in CRIPTO anti-sense NT2D1 cells, was induced by RA treatment in all cell lines. Taken together, our results show that inhibition of endogenous CRIPTO expression in human embryonal carcinoma cells interferes with both transformation and differentiation.

    Topics: Base Sequence; Carcinoma, Embryonal; Cell Adhesion; Cell Differentiation; Cell Division; Cell Transformation, Neoplastic; DNA Primers; Epidermal Growth Factor; GPI-Linked Proteins; Growth Substances; Humans; Intercellular Signaling Peptides and Proteins; Membrane Glycoproteins; Molecular Sequence Data; Neoplasm Proteins; Plasmids; RNA, Antisense; Transfection; Tumor Cells, Cultured

1996
Role of a signal transduction pathway which controls disassembly of microfilament bundles and suppression of high-molecular-weight tropomyosin expression in oncogenic transformation of NRK cells.
    Oncogene, 1996, May-16, Volume: 12, Issue:10

    Role of disassembly of microfilament bundles and suppression of high-molecular-weight tropomyosin (TM) expression in growth factor- and various oncogene-induced transformation was studied by using NRK cells and its transformation-deficient mutants. In NRK cells which show a transformed phenotype by treatment with EGF and TGF-beta, cellular stress fibers became dissociated by EGF or EGF and TGF-beta combination, whereas TGF-beta alone caused thicker appearance of stress fibers. Accompanying these changes, the expression of TM isoforms 1 and 2 was suppressed by treatment with EGF or EGF and TGF-beta, but elevated by TGF-beta with similar time courses. On the other hand, the transformation-deficient mutant cell lines, 39-1 and 39-3, did not show the transformed phenotypes by treatment with EGF and TGF-beta. Neither EGF nor EGF and TGF-beta combination affected cellular stress fibers and expression of TM isoforms 1 and 2 in both mutant lines. The relationship between the formation of stress fibers and the expression of TM isoforms was consistent in NRK cells, the mutant lines and their various oncogene-expressing sublines under various culture conditions. NRK cells overexpressing exogenous mouse TM isoform 2 showed markedly decreased susceptibility to EGF-induced dissociation of stress fibers and decreased anchorage-independent growth potential in the presence of EGF and TGF-beta. These results indicate that the transformation-deficient NRK mutant lines, 39-1 and 39-3 have defects in an EGF signal transduction pathway which induces suppression of high-molecular-weight TM expression and disassembly of microfilament bundles and suggested that the activation of the pathway is important for morphological transformation and oncogenic growth in growth factors- and various oncogene-induced transformation of NRK cells.

    Topics: Actin Cytoskeleton; Actins; Animals; Cell Transformation, Neoplastic; Epidermal Growth Factor; Gene Expression; Isomerism; Mice; Mutation; Oncogenes; Signal Transduction; Tropomyosin

1996
Drug-induced reversion of progression phenotype is accompanied by reversion of AP-1 phenotype in JB6 cells.
    In vitro cellular & developmental biology. Animal, 1996, Volume: 32, Issue:4

    Transformed JB6 cells can be stably reverted to nontransformed phenotype by AP-1 inhibiting gluccorticoid fluocinolone (FA) and cAMP elevator forskolin (FN), yielding stable revertants of promotion resistant (P-) and promotion sensitive (P+) phenotypes. AP-1 activity of nontransformed P- and P+ revertant clones was decreased under a variety of experimental conditions compared with their transformed counterparts. Moreover, AP-1 activity in P+ cells under anchorage-independent conditions was induced by 12-0-tetradecanoyl-phorbol-13-acetate (TPA) while AP-1 activity in the reverted P- cells was not induced, just as observed for the original P+ and P- variants. Taken together these data suggest that changes in AP-1 activity may be one key mediator not only of forward progression but also of reversion of tumor cells to nontransformed phenotype. In addition, the higher transfection efficiency of the new reverted P- and P+ cells renders them useful for studying the role of transcription factors in tumor promotion.

    Topics: 3T3 Cells; Animals; Cell Division; Cell Line; Cell Transformation, Neoplastic; Clone Cells; Colforsin; Epidermal Growth Factor; Fluocinolone Acetonide; Glucocorticoids; Mice; Phenotype; Tetradecanoylphorbol Acetate; Transcription Factor AP-1; Transfection

1996
Differential effect of polyunsaturated fatty acids on cell proliferation during human epithelial in vitro carcinogenesis: involvement of epidermal growth factor receptor tyrosine kinase.
    British journal of cancer, 1996, Volume: 74, Issue:4

    Polyunsaturated fatty acids (PUFAs) have been implicated in tumour development and have been shown to influence cell proliferation in vitro. We report here that n-3 and n-6 PUFAs at concentration > 10 microM inhibited the proliferation of a human kidney epithelial cell line (21HKE), which has retained phenotypic characteristics of normal kidney epithelial cells. In contrast, the proliferation was stimulated by n-3 and n-6 PUFAs at concentrations < 10 microM under defined growth conditions. The stimulatory effect of n-3 and n-6 PUFAs was even more profound in the presence of EGF. In human kidney epithelial cell lines reflecting different stages of transformation (11HKE and 1THKEras), the stimulatory effect was abrogated both in the presence and absence of EGF. Saturated fatty acids did not show any stimulatory effect on cell growth. The tyrosine kinase inhibitors genistein and tyrphostin-47 inhibited EGF-induced protein tyrosine phosphorylation dose-dependently in the 21HKE cells, and abolished the growth stimulatory effect of docosahexaenoic acid (DHA). This indicates the involvement of EGF receptor tyrosine kinase activity in the observed increase in cell proliferation.

    Topics: Cell Division; Cell Line; Cell Transformation, Neoplastic; DNA; Enzyme Inhibitors; Epidermal Growth Factor; Epithelium; ErbB Receptors; Fatty Acids, Nonesterified; Fatty Acids, Omega-3; Fatty Acids, Omega-6; Fatty Acids, Unsaturated; Genes, ras; Genistein; Humans; Isoflavones; Kidney; Kinetics; Nitriles; Phenols; Thymidine; Tyrphostins

1996
Requirement for phosphatidylinositol 3-kinase in epidermal growth factor-induced AP-1 transactivation and transformation in JB6 P+ cells.
    Molecular and cellular biology, 1996, Volume: 16, Issue:11

    Phosphatidylinositol 3-kinase (PI 3-kinase) plays a role in a variety of biological processes, including regulation of gene expression, cell growth, and differentiation. However, little is known about its role in the cytoplasmic events involved in epidermal growth factor (EGF)-induced transduction of signals to the transcriptional machinery of the nucleus and in EGF-induced cell transformation. In this study, we examined whether PI 3-kinase is a mediator for the activation of AP-1 and neoplastic transformation by EGF in the murine epidermal cell line JB6. The results showed the following. (i) EGF not only induced a high level of PI 3-kinase activity by itself but also enhanced insulin-induced PI 3-kinase activity in JB6 P+ cells, the EGF-induced PI-3 kinase activity could be blocked by constitutive overexpression of a dominant negative P85 subunit of PI 3-kinase (deltaP85), and insulin could markedly promote EGF-induced AP-1 activity in a dose-dependent manner in JB6 P+ cells as well as promote EGF-induced JB6 P+ cell transformation. (ii) Inhibition of PI-3 kinase with wortmannin or LY294002 markedly decreased the AP-1 activity induced by insulin, EGF, or EGF and insulin in a dose-dependent manner, while wortmannin did not block UVB-induced AP-1 activity. (iii) AP-1 activation by insulin, EGF, or EGF and insulin could be completely inhibited by overexpression of deltaP85 in all the dose and time courses studied. (iv) Inhibitors of PI 3-kinase (wortmannin and LY294002) and stable overexpression of deltaP85 inhibited EGF-induced transformation but had no significant inhibitory effect on cell proliferation induced by EGF or EGF and insulin. These results demonstrate for the first time that PI 3-kinase appears to be required for EGF- or insulin-induced AP-1 transactivation and cell transformation but not cell proliferation in JB6 cells.

    Topics: Androstadienes; Animals; Cell Line; Cell Transformation, Neoplastic; Chromones; Enzyme Induction; Enzyme Inhibitors; Epidermal Growth Factor; Genes, Reporter; Insulin; Kinetics; Luciferases; Mice; Morpholines; Phosphatidylinositol 3-Kinases; Phosphotransferases (Alcohol Group Acceptor); Skin; Transcription Factor AP-1; Transcriptional Activation; Transfection; Wortmannin

1996
The role of transforming growth factor alpha in the enterochromaffin-like cell tumor autonomy in an African rodent mastomys.
    Gastroenterology, 1996, Volume: 111, Issue:5

    Gastric carcinoids evolved from enterochromaffin-like (ECL) cell hyperplasia are usually associated with high pH and hypergastrinemia. The Mastomys species exhibits a genetic propensity to gastric carcinoid formation that can be accelerated by acid inhibition-induced hypergastrinemia. Although gastrin is critical in the initiation of the ECL cell transformation, the role of other growth factors involved in the evolution of the tumor autonomy has not been established. The aim of this study was to evaluate the role of transforming growth factor (TGF) alpha in the regulation of ECL cell transformation.. Mastomys were orally administered an irreversible H2-receptor antagonist loxtidine for 0, 8, and 16 weeks, and ECL cell transformation was monitored by assessing gastrin levels, mucosal histamine content, and chromogranin immunoreactivity. The ECL cells were purified, and cell proliferation at each stage in response to gastrin and TGF alpha was measured by bromodeoxyuridine uptake. TGF-alpha expression was evaluated by radioimmunoassay and Northern blot, and epidermal growth factor (EGF) receptor expression was determined by Western blot, immunoprecipitation, and immunocytochemistry.. Although the response to gastrin decreased during hypergastrinemia, the proliferative effect of TGF-alpha on ECL cells was specifically amplified during the development of hyperplasia. TGF-alpha and EGF receptor expression increased steadily in the transformed cells.. During low acid-induced hypergastrinemia, the expression of TGF-alpha and EGF receptor may constitute an autocrine regulatory mechanism in ECL cell tumor transformation.

    Topics: Animals; Carcinoid Tumor; Cell Division; Cell Transformation, Neoplastic; Cells, Cultured; Enterochromaffin Cells; Epidermal Growth Factor; ErbB Receptors; Gastrins; Histamine; Muridae; Stomach Neoplasms; Transforming Growth Factor alpha

1996
The relationship between human epidermal growth-like factor receptor expression and cellular transformation in NIH3T3 cells.
    The Journal of biological chemistry, 1996, Nov-29, Volume: 271, Issue:48

    A collection of cell lines expressing each human epidermal growth factor receptor (HER) family member alone or in all pairwise combinations in a clone of NIH3T3 cells (3T3-7d) devoid of detectable epidermal growth factor receptor family members has been generated. Transformation, as measured by growth in soft agar, occurred only in the presence of appropriate ligand and only in cells expressing two different HER family members. Transfection of oncogenic neu (Tneu), conferred ligand-independent transformation only in cells which co-expressed HER1, HER3, or HER4, but not when expressed alone or with HER2. Cell lines were also tested for their ability to form tumors in animals. None of the cell lines expressing single HER family members was able to form tumors in animals with the exception of HER1, which was weakly tumorigenic. Although unable to form tumors when expressed alone, HER2 was tumorigenic when expressed with HER1 or HER3, but not HER4. Of all complexes analyzed, cells expressing HER1 + HER2 were the most aggressive. The relationship between HER1 activation, intracellular calcium fluxes, and phospholipase Cgamma1 activation is well established. We found that activation of HER1 was required for the induction of a calcium flux and the phosphorylation of phospholipase Cgamma1. These activities were independent of, and unaffected by, the co-expression of any other family member. Further, heregulin stimulation of all cell lines including those containing HER1 did not demonstrate any effect on intracellular calcium levels or phospholipase Cgamma1 phosphorylation. This demonstrates that heregulin induced cellular transformation by activating HER3- and HER4-containing complexes does not require the activation of either phospholipase Cgamma1 or the mobilization of intracellular calcium.

    Topics: 3T3 Cells; Animals; Calcium; Carrier Proteins; Cell Transformation, Neoplastic; Epidermal Growth Factor; ErbB Receptors; Glycoproteins; Humans; Isoenzymes; Mice; Neuregulin-1; Phospholipase C gamma; Phosphotyrosine; Proto-Oncogene Proteins; Receptor, ErbB-2; Receptor, ErbB-3; Receptor, ErbB-4; Signal Transduction; Type C Phospholipases

1996
Brk, a breast tumor-derived non-receptor protein-tyrosine kinase, sensitizes mammary epithelial cells to epidermal growth factor.
    The Journal of biological chemistry, 1996, Nov-29, Volume: 271, Issue:48

    brk (breast tumor kinase) shows homology to the src family of non-receptor protein-tyrosine kinases and is expressed in breast carcinomas. In order to investigate the role of brk in breast tumor development, we have examined the growth and transformation properties of human mammary epithelial cells engineered to overexpress Brk. Interestingly, like c-Src, overexpression of Brk leads to sensitization to EGF, and also results in a partially transformed phenotype. Further investigation of the latter activity was attempted by mutational analysis, targeting key residues known to affect tyrosine kinase activity in Src-like kinases. Mutation of amino acid residue Lys-219 to Met, by analogy to Src, abolished both kinase activity and transformation capacity. Mutation of amino acid residue Tyr-447 to Phe, however, resulted in a decrease in transforming potential without affecting kinase activity. These results suggest that while Src and Brk share some functional properties, they act differently during transformation. These differences are discussed in the context of the mechanisms underlying breast cancer development.

    Topics: 3T3 Cells; Animals; Breast; Cell Division; Cell Transformation, Neoplastic; Cells, Cultured; Epidermal Growth Factor; Epithelium; ErbB Receptors; Female; Humans; Mice; Neoplasm Proteins; Protein-Tyrosine Kinases

1996
Epidermal growth factor and glucocorticoid effects on blood prostaglandin E2 level in DENA-induced carcinogenesis.
    Prostaglandins, leukotrienes, and essential fatty acids, 1996, Volume: 55, Issue:4

    This paper describes results on carcinogenesis in rats induced with diethylnitrosamine (DENA; 280 mg/kg single dose, i.p.). After inducing chemical carcinogenesis, the animals were treated with different compounds that affect cell proliferation and the effects on the blood prostaglandin E2 (PGE2) levels were observed. These compounds were epidermal growth factor (EGF, 10 micrograms/kg/24 h i.p., 7 days) and dexamethasone (4 mg/kg/24 h i.p., 7 days). The blood PGE2 levels of rats were measured by RIA (Radioimmunoassay). The blood PGE2 levels increased significantly following DENA and EGF administrations alone or together. Dexamethasone administration decreased the blood PGE2 levels. In conclusion the data show that the increase in the plasma PGE2 level by DENA treatment was enhanced by EGF but opposed by dexamethasone. This suggests that EGF may be effective as a promoter in chemical carcinogenesis.

    Topics: Animals; Carcinogens; Cell Transformation, Neoplastic; Dexamethasone; Diethylnitrosamine; Dinoprostone; Epidermal Growth Factor; Glucocorticoids; Male; Rats; Rats, Wistar

1996
[Proliferation and differentiation of human salivary gland adenocarcinoma cell line HSG].
    Human cell, 1996, Volume: 9, Issue:1

    The adenocarcinoma cell line derived from an intercalated ductal epithelium of a human salivary gland (HSG) proliferates autonomously mediated by an epidermal growth factor-(EGF)-like molecule with a molecular weight of 46 kDa and an EGF receptor (EGFR). The c-erbB2 protein, a member of EGFR family was also expressed in HSG cells and was involved in the growth signal pathway of HSG cells as well as EGFR. The autocrine growth is regulated by glucocorticoid and retinoic acid (RA) via their receptors. Retinoic acid receptor (RAR) of HSG cells revealed a transcriptional activity in vivo, and the heterodimerization between RAR and 9-cis retinoic acid receptor (RXR) is requisite for the binding with a specific DNA element termed RA response element in vitro. RXR alpha and RXR beta were cloned from HSG cells, and these RXRs, together with RAR, seemed to play a physiological role in RA signaling in vivo.

    Topics: Adenocarcinoma; Bucladesine; Cell Division; Cell Transformation, Neoplastic; Epidermal Growth Factor; Humans; Receptors, Retinoic Acid; Salivary Gland Neoplasms; Signal Transduction; Transcription, Genetic; Tretinoin; Tumor Cells, Cultured

1996
Activation of the EGF receptor by insertional mutations in its juxtamembrane regions.
    Oncogene, 1995, Oct-19, Volume: 11, Issue:8

    Ligand dependent activation of receptor tyrosine kinases is mediated by an allosteric dimerization process that is responsible for the stimulation of protein tyrosine kinase activity and receptor autophosphorylation. In order to gain further insight into the processes which control this process, we have generated EGF receptor mutants that contain inserts of 20-40 amino acids in their juxtamembrane regions, on each side of the receptor's single transmembrane domain. An EGF receptor mutant with an insertion on the cytoplasmic side of the transmembrane domain exhibited typical EGF binding characteristics, ligand-dependent tyrosine autophosphorylation, as well as ligand-induced DNA synthesis. However, an EGF receptor mutant with an insertion on both sides of the transmembrane domain was found to be constitutively activated. This mutant also exhibited dramatically reduced EGF binding, but dimerized and had enhanced tyrosine kinase activity even in the absence of ligand. Moreover, NIH3T3 cells expressing this mutant receptor formed colonies in soft agar in the absence of EGF. This represents a novel example of a constitutively activated receptor, and provides further support for receptor dimerization as a mechanism for activation of EGFR and other receptor tyrosine kinases.

    Topics: 3T3 Cells; Amino Acid Sequence; Animals; Base Sequence; Cell Division; Cell Transformation, Neoplastic; Cells, Cultured; Enzyme Activation; Epidermal Growth Factor; ErbB Receptors; Humans; Membrane Proteins; Mice; Molecular Sequence Data; Mutagenesis, Insertional; Oligodeoxyribonucleotides; Phosphorylation; Protein Binding; Signal Transduction; Structure-Activity Relationship

1995
Effect of human papillomavirus type 16 oncogenes on MAP kinase activity.
    Journal of virology, 1995, Volume: 69, Issue:12

    The mitogen-activated protein (MAP) kinase signal transduction pathway is an intracellular signaling cascade which mediates cellular responses to growth and differentiation factors. The MAP kinase pathway can be activated by a wide range of stimuli dependent on the cell types, and this is normally a transient response. Oncogenes such as ras, src, raf, and mos have been proposed to transform cells in part by prolonging the activated stage of components within this signaling pathway. The human papillomavirus (HPV) oncogenes E6 and E7 play an essential role in the in vitro transformation of primary human keratinocytes and rodent cells. The HPV type 16 E5 gene has also been shown to have weak transforming activity and may enhance the epidermal growth factor (EGF)-mediated signal transduction to the nucleus. In the present study, we have investigated the effects of the oncogenic HPV type 16 E5, E6, and E7 genes on the induction of the MAP kinase signaling pathway. The E5 gene induced an increase in the MAP kinase activity both in the absence and in the presence of EGF. In comparison, the E6 and E7 oncoproteins do not alter the MAP kinase activity or prolong the MAP kinase activity induced with EGF. These findings suggest that E5 may function, at least in part, to enhance the cell response through the MAP kinase pathway. However, the transforming activity of E6 and E7 is not associated with alterations in the MAP kinase pathway. These findings are consistent with E5 enhancing the response to growth factor stimulation.

    Topics: Animals; Base Sequence; Blotting, Northern; Calcium-Calmodulin-Dependent Protein Kinases; Cell Line; Cell Nucleus; Cell Transformation, Neoplastic; Chlorocebus aethiops; DNA Primers; Enzyme Activation; Epidermal Growth Factor; Gene Expression; Humans; Keratinocytes; Molecular Sequence Data; Oncogene Proteins, Viral; Oncogenes; Papillomaviridae; Polymerase Chain Reaction; Recombinant Proteins; Signal Transduction; Transfection

1995
Effect of Clostridium perfringens-derived wound healing substance as compared with epidermal growth factor on the growth and morphological transformation of BALB/3T3 A31-1-1 cells.
    Mutation research, 1995, Volume: 341, Issue:3

    Clostridium perfringens-derived wound-healing substance (WHS), having growth-stimulating activity, was examined to determine its effect on the growth and morphological transformation of BALB/3T3 A31-1-1 cells. WHS accelerated the cell growth at the exponential growth phase, shortening the doubling time by 8-18%. The maximum cell density of the treated cultures was slightly higher than that of the control culture, and the cell number decreased in the same way as the control cells did. On the other hand, the cells treated with epidermal growth factor (EGF) or insulin showed growth rates similar to that of the control cells during the exponential growth phase, and after the control cells attained the maximum cell number, the number of the treated cells continued to increase gradually for more than 4 days and then decreased. Under the experimental conditions of the two-stage transformation assay, application of WHS at the tumor-initiation or promotion stage did not accelerate the formation of transformed foci. Although treatment with EGF at the initiation stage induced no enhancement, marked enhancement of morphological transformation was observed in the treatment at the promotion stage. These results indicate that the mode of action between WHS and EGF or insulin is different on the growth-stimulating activity and morphological transformation of BALB/3T3 A31-1-1 cells.

    Topics: 3T3 Cells; Animals; Cell Division; Cell Transformation, Neoplastic; Epidermal Growth Factor; Growth Substances; Mice; Mice, Inbred BALB C; Peptides; Wound Healing

1995
Rapid induction of heparin-binding epidermal growth factor/diphtheria toxin receptor expression by Raf and Ras oncogenes.
    Genes & development, 1995, Aug-15, Volume: 9, Issue:16

    We have used differential display PCR to search for mRNAs induced by delta Raf-1:ER, an estradiol-dependent form of Raf-1 kinase. Through this approach the gene encoding heparin-binding epidermal growth factor (HB-EGF) was identified as an immediate-early transcriptional target of oncogenic Raf kinases. Activation of delta Raf-1:ER and a conditional oncogenic form of B-Raf, delta B-RAF:ER, resulted in rapid and sustained induction of HB-EGF mRNA expression and secretion of mature HB-EGF from cells. Neutralizing anti-HB-EGF antisera prevented the delayed activation of the c-Jun amino-terminal kinases that is observed in cells transformed by delta Raf-1:ER. These results demonstrate that distinct signaling pathways can cross talk via the secretion of polypeptide growth factors. Furthermore, cells transformed by oncogenic Ras, which also induced HB-EGF expression, demonstrated a marked increase in sensitivity to the cytotoxic action of diphtheria toxin, for which the membrane anchored HB-EGF precursor acts as a cell-surface receptor.

    Topics: 3T3 Cells; Animals; Base Sequence; Calcium-Calmodulin-Dependent Protein Kinases; Cell Line; Cell Line, Transformed; Cell Transformation, Neoplastic; Diphtheria Toxin; DNA Primers; Epidermal Growth Factor; Estradiol; Gene Expression; Genes, ras; Heparin; Heparin-binding EGF-like Growth Factor; Humans; Intercellular Signaling Peptides and Proteins; JNK Mitogen-Activated Protein Kinases; Mice; Mitogen-Activated Protein Kinases; Molecular Sequence Data; Oncogene Proteins v-raf; Oncogenes; Polymerase Chain Reaction; Protein Serine-Threonine Kinases; Protein-Tyrosine Kinases; Proto-Oncogene Proteins; Proto-Oncogene Proteins c-jun; Proto-Oncogene Proteins c-raf; Receptors, Cell Surface; Retroviridae Proteins, Oncogenic; RNA, Messenger

1995
Alterations in receptor-mediated kinases and phosphatases during carcinogenesis.
    Journal of cancer research and clinical oncology, 1995, Volume: 121, Issue:3

    Increased phosphorylation in cancers can stimulate growth and up-regulate certain receptors. To test whether the functional response of phosphatase receptors is up-regulated during carcinogenesis, we examined the effects of ligands on net phosphorylation in isolated membranes derived from hamster cheek-pouch tissues undergoing malignant transformation. The buccal mucosa of groups of Syrian golden hamsters was exposed thrice weekly to 0.5% dimethylbenzanthracene (DMBA) in acetone for 2-12 weeks to produce premalignant and malignant tissues. Homogenates of these tissues were then incubated with [32P]ATP in the presence of epidermal growth factor (EGF), agonist of somatostatin analogue RC-160, luteinizing-hormone-releasing hormone (LH-RH) [D-Trp6]LH-RH, or combinations of EGF, RC-160, and [D-Trp6]LH-RH. Changes compared to controls in phosphorylation in response to ligands provided estimates of kinase or phosphatase activity. Phosphorylation increased continuously, from the first application of DMBA in a linear fashion, and independently of EGF stimulation. RC-160 and [D-Trp6]LH-RH reduced phosphorylation in vitro. This response occurred in premalignant (weeks 6-10 after DMBA application) as well as malignant tissues (week 12 after DMBA application), but was not significant in normal tissues. The results show a continuous augmentation in phosphatase activity prior to the appearance of cancers, but with a delay in expression following the primary event of increased kinase activity. Significantly less phosphorylation of substrates was induced by both RC-160 and [D-Trp6]LH-RH after in vitro activation by EGF than in the absence of EGF. This suggests that EGF activates latent systems of hormonal receptors. Collectively, these results support the hypothesis that the enhancement of the hormonally stimulated phosphatase in cancers occurs secondarily to the increased kinase activity.

    Topics: 9,10-Dimethyl-1,2-benzanthracene; Animals; Carcinoma, Squamous Cell; Cell Transformation, Neoplastic; Cricetinae; Epidermal Growth Factor; Gonadotropin-Releasing Hormone; Mesocricetus; Mouth Mucosa; Phosphorylation; Precancerous Conditions; Protein Tyrosine Phosphatases; Receptor Protein-Tyrosine Kinases; Somatostatin; Time Factors; Up-Regulation

1995
The human papillomavirus (HPV)-6 and HPV-16 E5 proteins co-operate with HPV-16 E7 in the transformation of primary rodent cells.
    The Journal of general virology, 1995, Volume: 76 ( Pt 5)

    E5 is the smallest transforming protein encoded by the human papillomaviruses (HPVs). It has been shown to promote anchorage-independent growth in established NIH 3T3 cells, an activity that is enhanced in the presence of epidermal growth factor (EGF). It is thought that this activity of E5 is brought about by an increase in the half-life of stimulated EGF receptors, possibly through the perturbation of receptor processing. Recent studies have also shown that E5 can co-operate with HPV-16 E7 to stimulate proliferation of primary rodent cells. Using haemagglutinin I epitope-tagged E5 proteins, we have compared the mitogenic activity of HPV-6 and HPV-16 E5. Both tagged proteins retain the ability to bind to the cellular 16 kDa H(+)-ATPase protein. In addition, both HPV-6 and HPV-16 E5 retain the ability to co-operate with E7 in primary rodent cells, although HPV-16 E5 is considerably more active than HPV-6 E5 in these mitogenic assays. Interestingly, transfection of a plasmid over-expressing c-Raf appears to be capable of functionally substituting for E5 in the co-mitogen assays. Polyclonal cell lines derived from baby rat kidney cells co-transfected with E7 and E5 genes continue to express both the E5 and E7 mRNA, although the level of E5 expression is very low and protein cannot be detected. These polyclonal lines appear to be immortal and in some cases demonstrate anchorage-independent growth, an activity which is enhanced by the addition of EGF.

    Topics: Animals; Cell Line; Cell Transformation, Neoplastic; Epidermal Growth Factor; Oncogene Proteins, Viral; Papillomaviridae; Papillomavirus E7 Proteins; Rats

1995
Constitutive phosphorylation of eps8 in tumor cell lines: relevance to malignant transformation.
    Molecular and cellular biology, 1995, Volume: 15, Issue:7

    eps8, a recently identified tyrosine kinase substrate, has been shown to augment epidermal growth factor (EGF) responsiveness, implicating it in EGF receptor (EGFR)-mediated mitogenic signaling. We investigated the status of eps8 phosphorylation in normal and transformed cells and the role of eps8 in transformation. In NIH 3T3 cells overexpressing EGFR (NIH-EGFR), eps8 becomes rapidly phosphorylated upon EGF stimulation. At receptor-saturating doses of EGF, approximately 30% of the eps8 pool is tyrosine phosphorylated. Under physiological conditions of activation (i.e., at low receptor occupancy), corresponding to the 50% effective dose of EGF for mitogenesis, approximately 3 to 4% of the eps8 contains phosphotyrosine. In human tumor cell lines, we detected constitutive tyrosine phosphorylation of eps8, with a stoichiometry (approximately 5%) similar to that associated with potent mitogenic response in NIH-EGFR cells. Overexpression of eps8 was able to transform NIH 3T3 cells under limiting conditions of activation of the EGFR pathway. Concomitant tyrosine phosphorylation of eps8 and shc, but not of rasGAP, phospholipase C-gamma, and eps15, was frequently detected in tumor cells. This suggested that eps8 and shc might be part of a pathway which is preferentially selected in some tumors. Cooperation between these two transducers was further indicated by the finding of their in vivo association. This association was, at least in part, dependent on recognition of shc by the SH3 domain of eps8. Our results indicate that eps8 is physiologically part of the EGFR-activated signaling and that its alterations can contribute to the malignant phenotype.

    Topics: Adaptor Proteins, Signal Transducing; Animals; Cell Transformation, Neoplastic; Cytoskeletal Proteins; Epidermal Growth Factor; ErbB Receptors; Humans; Intracellular Signaling Peptides and Proteins; Mice; Mitogens; Phosphorylation; Precipitin Tests; Protein-Tyrosine Kinases; Proteins; Recombinant Proteins; Signal Transduction; Tumor Cells, Cultured

1995
Epidermal growth factor ligand-independent, unregulated, cell-transforming potential of a naturally occurring human mutant EGFRvIII gene.
    Cell growth & differentiation : the molecular biology journal of the American Association for Cancer Research, 1995, Volume: 6, Issue:10

    The type III deletion-mutant gene for the epidermal growth factor receptor (EGFRvIII) is frequently expressed in glioblastomas and in breast and non-small cell lung carcinomas. To understand its contribution to the malignant phenotype in humans, we transfected NR6 cells with the mammalian vector pH beta APr-1-neo containing cDNA for either EGFRvIII or wild-type EGFR. Western blot analyses showed that NR6 transfected with wild-type EGFR (NR6W) contained a normal-sized protein (170 kilodaltons); cells transfected with EGFRvIII (NR6M) contained a truncated protein (145 kilodaltons). NR6W cells demonstrated a saturation binding curve with 125I-labeled EGF (affinity, 1.8 x 10(8); r2 = 0.96). NR6M cells, however, showed a low but consistent level of 125I-labeled EGF binding (affinity, 4 x 10(7); r2 = 0.99) compared with NR6, which lacked binding. The population doubling time was shorter for NR6M (0.64 days) than for NR6W (1.1 days) and NR6V (2.27 days). Soft agar focus formation assay by NR6M was 4- to 5-fold higher than that by NR6W. In nude mice, NR6M (1 x 10(7) cells), without exogenous ligand, formed tumors within 12 days; no tumors were observed over 90 days in mice receiving identical doses of NR6W, NR6V, or NR6 cells. EGF stimulated autophosphorylation of receptor in NR6W (4- to 9-fold) but caused only slight (1.8- to 1.9-fold) to no enhancement in NR6M. Further, there was no difference in constitutive tyrosine kinase activity between NR6M and NR6W. Our results clearly indicate that EGFRvIII functions as an oncoprotein, but its intrinsic tyrosine kinase activity may not be responsible for its biological function.

    Topics: 3T3 Cells; Animals; Cell Division; Cell Transformation, Neoplastic; Epidermal Growth Factor; ErbB Receptors; Humans; Kinetics; Mice; Mice, Nude; Oncogene Proteins; Phosphorylation; Protein-Tyrosine Kinases; RNA, Messenger; Sequence Deletion; Transfection

1995
Tyrosine phosphorylation of protein phosphatase 2A in response to growth stimulation and v-src transformation of fibroblasts.
    The Journal of biological chemistry, 1994, Mar-18, Volume: 269, Issue:11

    The catalytic subunit of protein phosphatase 2A (PP2A) is inactivated by in vitro phosphorylation of Tyr307 by receptor and nonreceptor protein tyrosine kinases (Chen, J., Martin, B. L., and Brautigan, D. L. (1992) Science 257, 1261-1264). Here we show the phosphorylation of PP2A in cells under different growth conditions. In lysates of nontransformed murine 10T1/2 fibroblasts, there were two forms of PP2A at 36 kDa detected after two-dimensional gel electrophoresis and immunoblotting with anti-PP2A peptide antibody. These two forms exactly comigrated with unphosphorylated purified PP2A and the PP2A 32P-labeled by in vitro phosphorylation with p60v-src kinase. The phosphorylated form of PP2A recovered from red blood cells or produced by in vitro phosphorylation was eliminated by incubation with tyrosine-specific phosphatase (PTP1B). Transformation of 10T1/2 cells by expression of p60v-src resulted in most of the PP2A in the cells being converted to a phosphorylated form that was reactive with anti-phosphotyrosine antibody. Serum starvation of cells reduced the amount of phosphorylated PP2A, whereas serum stimulation of quiescent cells caused an increase to the same relative amount of phosphorylated PP2A as in src-transformed cells. Addition of epidermal growth factor to quiescent NeoR cells (10T1/2 fibroblasts overexpressing epidermal growth factor receptors) temporarily increased the level of phosphorylation of PP2A, with a peak at 5-15 min and a return to basal level within 60 min. The results show that PP2A is phosphorylated in intact cells, and the extent of this modification is increased by growth factors or cell transformation, providing evidence for a physiological mechanism of PP2A regulation.

    Topics: Amino Acid Sequence; Animals; Antibodies; Cell Line; Cell Transformation, Neoplastic; Culture Media; Electrophoresis, Gel, Two-Dimensional; Epidermal Growth Factor; ErbB Receptors; Fibroblasts; Genes, src; Immunoblotting; Kinetics; Macromolecular Substances; Mice; Molecular Sequence Data; Peptides; Phosphoprotein Phosphatases; Phosphorylation; Phosphotyrosine; Protein Phosphatase 2; Tyrosine

1994
Autonomous growth in serum-free medium and production of hepatocellular carcinomas by differentiated hepatocyte lines that overexpress transforming growth factor alpha 1.
    Cancer research, 1994, Nov-15, Volume: 54, Issue:22

    Transforming growth factor alpha (TGF-alpha) is a polypeptide closely associated with hepatocyte proliferation in vivo and in vitro. In order to investigate the mechanisms by which TGF-alpha contributes to hepatocyte replication and transformation, we isolated hepatocytes from mice bearing a human TGF-alpha transgene and examined their growth properties and gene expression in defined, serum-free culture. The transgenic hepatocytes continued to overexpress human TGF-alpha mRNA and peptide, and were able to proliferate without exogenous growth factors in primary culture, in contrast to nontransgenic mouse hepatocytes. In short-term culture the transgenic hepatocytes underwent 1 wave of DNA replication at 72-96 h in culture before senescing, similar to nontransgenic hepatocytes supplemented with epidermal growth factor. Constitutive expression of TGF-alpha rendered the transgenic hepatocytes unresponsive to further growth stimulation by exogenous TGF-alpha, as well as other mitogens such as epidermal growth factor and hepatocyte growth factor. However, it did not alter their sensitivity to growth inhibition by TGF beta 1, 2 and 3. The addition of nicotinamide to the culture medium enabled both transgenic and epidermal growth factor-supplemented normal hepatocytes to replicate repeatedly and survive for > or = 2 months in primary culture while maintaining differentiated traits. From these long-term primary cultures of transgenic and nontransgenic hepatocytes, we established immortalized cell lines (designated TAMH and NMH lines, respectively). Both lines continued to express differentiated adult hepatocytic markers such as albumin, alpha-1-antitrypsin, transferrin, and connexin 26 and 32 mRNAs, but also expressed mRNAs for the oncofetal markers alpha-fetoprotein and insulin-like growth factor II. Unlike the near-diploid NMH hepatocyte line, the transgenic TAMH hepatocyte line was quasi-tetraploid, strongly expressed human TGF-alpha mRNA, and was highly tumorigenic in nude mice. Well-differentiated hepatocellular carcinomas developed in nude mice given injections of the TAMH line, and these appeared similar to the primary liver tumors seen in TGF-alpha transgenic mice with regard to histology and strong expression of mouse and human TGF-alpha, insulin-like growth factor II, and alpha-fetoprotein mRNAs. Our data show that TGF-alpha overexpression causes autonomous hepatocyte proliferation and contributes to neoplasia but that additional cellular alterations mus

    Topics: alpha-Fetoproteins; Animals; Carcinoma, Hepatocellular; Cell Division; Cell Line; Cell Transformation, Neoplastic; Culture Media, Serum-Free; DNA Replication; Epidermal Growth Factor; Insulin-Like Growth Factor II; Liver; Male; Mice; Mice, Nude; Mice, Transgenic; Niacinamide; Ploidies; RNA, Messenger; Time Factors; Transforming Growth Factor alpha

1994
Frequent immunohistochemical detection of EGF supergene family members in ovarian carcinogenesis.
    International journal of gynecological pathology : official journal of the International Society of Gynecological Pathologists, 1994, Volume: 13, Issue:4

    Primary and metastatic ovarian cystadenocarcinomas, carcinomas of low malignant potential (borderline tumors), benign ovarian cystadenomas, and normal ovaries were compared for immunoperoxidase detection of the ligands epidermal growth factor (EGF), transforming growth factor-alpha (TGF-alpha), amphiregulin (AR), cripto, and the receptors, epidermal growth factor receptor (EGF-R), and c-erbB-2. This matrix analysis of these EGF family members indicated no specific pattern of ligand or receptor expression with a specific ovarian histologic category except in the case of AR and TGF-alpha. AR was detected almost exclusively in borderline tumors, suggesting that these tumors may not arise as a pathological continuum between benign cystadenomas and invasive cystadenocarcinomas. Second, the presence of TGF-alpha immunoreactivity in the absence of coexpression of cripto or EGF appeared to be associated only with adenocarcinomas of high grade and stage.

    Topics: Amphiregulin; Cell Transformation, Neoplastic; EGF Family of Proteins; Epidermal Growth Factor; ErbB Receptors; Female; Glycoproteins; GPI-Linked Proteins; Growth Substances; Humans; Immunoenzyme Techniques; Intercellular Signaling Peptides and Proteins; Membrane Glycoproteins; Multigene Family; Neoplasm Proteins; Ovarian Neoplasms; Receptor, ErbB-2; Transforming Growth Factor alpha

1994
Amphiregulin as an autocrine growth factor for c-Ha-ras- and c-erbB-2-transformed human mammary epithelial cells.
    Proceedings of the National Academy of Sciences of the United States of America, 1994, Mar-29, Volume: 91, Issue:7

    Amphiregulin (AR), a member of the epidermal growth factor (EGF) family, was found to be as potent as EGF in stimulating the anchorage-dependent growth (ADG) of immortalized, nontransformed human mammary epithelial MCF-10A cells. MCF-10A cells transformed by either an activated human c-Ha-ras protooncogene (MCF-10A ras) or by overexpression of a nonactivated rat c-neu gene (MCF-10A neu) exhibited a 35% reduction in the response to AR in ADG when compared to MCF-10A cells, but AR was still as potent as EGF in these transformants. Exogenous AR exhibited only 15-20% of the activity of EGF in stimulating the anchorage-independent growth, a response that is normally dependent upon exogenous EGF, of the oncogene-transformed MCF-10A cells. MCF-10A cells express low levels of a 1.4-kb AR mRNA transcript, while MCF-10A ras and MCF-10A neu cells display a 15- to 30-fold increase in the levels of AR mRNA and endogenous AR protein as determined by Western blot analysis. Exogenous EGF was found to induced both the AR mRNA and protein in the MCF-10A parental and transformed cells. A 20-mer phosphorothioate antisense deoxyoligonucleotide complementary to the 5' sequence of AR mRNA was able to significantly reduce the levels of endogenous AR protein and to inhibit the EGF-stimulated ADG and anchorage-independent growth of MCF-10A ras and MCF-10A neu cells. These data suggest that AR may function as an EGF-dependent autocrine growth factor in mammary epithelial cells that have been transformed by either a point-mutated c-Ha-ras or c-neu.

    Topics: Amphiregulin; Cell Division; Cell Line, Transformed; Cell Transformation, Neoplastic; EGF Family of Proteins; Epidermal Growth Factor; Epithelium; Genes, ras; Glycoproteins; Growth Substances; Humans; Intercellular Signaling Peptides and Proteins; Oligonucleotides, Antisense; Oncogene Proteins, Viral; Receptor, ErbB-2; RNA, Messenger; Thionucleotides

1994
Kinase-deficient neu proteins suppress epidermal growth factor receptor function and abolish cell transformation.
    Oncogene, 1994, Volume: 9, Issue:5

    p185c-neu and epidermal growth factor receptor (EGFR) associate into an active heterodimer, and overexpression of these two receptors leads to a transformed phenotype. However, the association of EGFR and kinase-deficient Neu proteins (point mutant N757 or cytoplasmic domain deletion mutant N691stop) results in a defective or inactive heterodimeric complex. In this report we explore the biological consequences of heterodimerization between EGFR and wild-type (WT) or kinase-deficient mutant Neu proteins in living cells. We show that co-expression of EGFR and kinase-deficient Neu proteins abolished the synergistic transformation and tumorigenicity. Moreover, the normal responses of EGFR to ligand were significantly suppressed, e.g., loss of EGF-dependent transformation, reduced rate of receptor endocytosis and turnover, diminished DNA synthesis, and decreased EGF binding affinity. These results provide the first evidence that kinase-deficient Neu proteins suppress normal EGFR function and display a dominant negative mutant phenotype. Together with the stimulatory effects observed in cells forming active heterodimers, these studies provide a role for heterodimerization of EGFR and Neu/c-erbB2 in interreceptor activation and synergistic signaling which may be responsible for the transition from normal receptor function into oncogenesis.

    Topics: Animals; Cell Line; Cell Transformation, Neoplastic; DNA; Dose-Response Relationship, Drug; Down-Regulation; Epidermal Growth Factor; ErbB Receptors; Fibroblasts; Half-Life; Mice; Mice, Nude; Phenotype; Point Mutation; Proto-Oncogene Proteins; Receptor, ErbB-2

1994
Preferential primary-response gene expression in promotion-resistant versus promotion-sensitive JB6 cells.
    Molecular carcinogenesis, 1994, Volume: 11, Issue:2

    The 12-O-tetradecanoylphorbol-13-acetate (TPA)-inducible sequence (TIS) genes are a set of primary response genes induced in Swiss 3T3 cells by TPA. They include three transcription factors, a prostaglandin synthase, and three proteins of unknown function. To ascertain which, if any TIS genes might be involved in tumor promotion, we examined the expression of these genes in response to tumor promoters in transformation promotion-sensitive (P+) and -resistant (P-) JB6 murine epidermal cells, a model used to identify events relevant to promotion. A subset of TIS genes (TIS1, TIS10, and TIS21) was preferentially induced by TPA in P-cells. In addition, TIS1 and TIS21 mRNAs were preferentially induced in P-cells by epidermal growth factor, another transformation promoter that distinguishes P+ from P-cells. TIS1 and TIS21 protein levels were also greater in TPA-treated P-cells than P+ cells. Forskolin, a cAMP-elevating anti-promoter, increased TPA-induced levels of TIS1, TIS10, and TIS21 mRNAs in P+ cells, ruling in potential roles for these genes in modulating tumor promotion. The anti-promoters fluocinolone acetonide, retinoic acid, and superoxide dismutase did not enhance TPA-induced levels of TIS1 and TIS21 mRNAs in P+ cells, suggesting that these inhibitors may act on other promotion-relevant genes. TIS1 encodes a member of the steroid receptor superfamily. TIS1 encodes a protein of unknown function with strong sequence similarity to BTG1, a proposed "anti-proliferative gene" (Rouault JP, Rimokh R, Tessa C, et al., EMBO J 11:1663-1670, 1992). Preferential induction by multiple promoters of these TIS genes in P-cells and enhancement of their induction in P+ cells by the anti-promoter forskolin make TIS1 and TIS21 candidates for promotion suppressor genes.

    Topics: 3T3 Cells; Animals; Carcinogens; Cell Transformation, Neoplastic; DNA-Binding Proteins; Epidermal Growth Factor; Gene Expression; Genes; Immediate-Early Proteins; Mice; Mice, Inbred BALB C; Multigene Family; Proteins; Tetradecanoylphorbol Acetate; Tristetraprolin

1994
c-Ha-rasEJ transfection of rat aortic smooth muscle cells induces epidermal growth factor responsiveness and characteristics of a malignant phenotype.
    Journal of cellular physiology, 1994, Volume: 161, Issue:3

    Although the role of several protooncogenes, including sis, myc, and myb in the regulation of growth and differentiation of vascular cells has been examined in some detail, limited information is available on the contribution of ras genes to these processes. In the present studies the influence of oncogenic ras transfection on the phenotypic expression of rat aortic smooth muscle cells (SMCs) was examined. Cultured rat aortic SMCs during early passage (P4) were transfected by lipofection with c-Ha-rasEJ in a pSV2 neo vector or with pSV2 neo vector alone. Stable transfectants were selected in G418 over a 6-week period. Oncogene-transfected cells (ras-LF-1) exhibited differences in morphology and growth pattern relative to vector controls (neo-LF-1), or naive SMCs, including the development of prominent processes and the appearance of focal cellular arrangements giving rise to latticelike structures. Southern analysis revealed multiple integration of oncogenic ras in ras LF-1 cells. Transfection of c-Ha-rasEJ was associated with a twofold increase in p21 levels relative to pSV2 vector controls demonstrating that exogenous ras was expressed in these cells. Overexpression of ras p21 afforded SMCs a lower serum requirement for growth compared to vector controls, anchorage independent growth on soft agar, and acquisition of epidermal growth factor (EGF) responsiveness. Stimulation of serum-deprived SMCs with 5% fetal bovine serum (FBS) increased steady-state levels of c-Ha-ras mRNA in both ras-LF-1 and neo-LF-1 but ras induction was more pronounced in ras-transfected cells. alpha-smooth muscle (SM) actin gene expression was markedly reduced in ras-transfected cells relative to vector controls. These results show that transfection of c-Ha-rasEJ into aortic SMCs induces an altered phenotypic state characterized by alterations in growth factor-related signal transduction and tumorigenic potential.

    Topics: Actins; Animals; Cell Division; Cell Transformation, Neoplastic; Epidermal Growth Factor; Gene Expression; Genes, myc; Muscle, Smooth, Vascular; Proto-Oncogene Proteins p21(ras); Rats; Transfection; Tubulin

1994
Effects of antiepidermal growth factor receptor antibody 528 on the proliferation and differentiation of head and neck cancer.
    Otolaryngology--head and neck surgery : official journal of American Academy of Otolaryngology-Head and Neck Surgery, 1994, Volume: 111, Issue:5

    Antibodies directed against the epidermal growth factor receptor may impede proliferation and induce differentiation of head and neck squamous cell carcinoma. To test this hypothesis, we examined the effect of monoclonal antibody 528 directed against epidermal growth factor receptor on the proliferation and differentiation of monolayer cells and multicenter tumor spheroids from three head and neck squamous cell carcinoma cell lines (1483, MDA 686Ln, and MDA 886Ln) and the epidermal growth factor-responsive vulvar carcinoma A431. All head and neck squamous cell carcinoma lines were shown to express high levels of epidermal growth factor receptor by Scatchard analyses. Epidermal growth factor inhibited the growth of monolayer cells but stimulated the growth of 886 and A431 multicellular tumor spheroids. Epidermal growth factor modulated the differentiation of A431 and 686 with respect to involucrin immunohistochemistry and cornified enveloped competency. Monoclonal antibody 528 directed against epidermal growth factor receptor inhibited cellular proliferation as measured by cell number, thymidine incorporation, and multicellular tumor spheroid volume. A mild promotion of differentiation was observed in the epidermal growth factor-responsive cells. In conclusion, monoclonal antibody 528 directed against epidermal growth factor receptor inhibits growth of head and neck squamous cell carcinoma cells bearing high levels of epidermal growth factor receptors and promotes differentiation in some tumors. The use of a multicellular tumor spheroid model to evaluate growth factor responsiveness and inhibition of proliferation may more accurately reflect in vivo tumor growth than monolayer cells. Antibodies against epidermal growth factor receptor may prove effective in modulating disease progression in patients with head and neck squamous cell carcinoma.

    Topics: Animals; Antibodies, Monoclonal; Carcinoma, Squamous Cell; Cell Division; Cell Transformation, Neoplastic; Epidermal Growth Factor; ErbB Receptors; Head and Neck Neoplasms; Humans; Mice; Tumor Cells, Cultured

1994
The peroxisome proliferator class of non-genotoxic hepatocarcinogens synergize with epidermal growth factor to promote clonal expansion of initiated rat hepatocytes.
    Carcinogenesis, 1994, Volume: 15, Issue:12

    The mechanisms by which the peroxisome proliferator (PP) class of non-genotoxic carcinogens perturb growth regulation and cause rodent liver cancer are unknown. Using a soft agar cloning assay, we have demonstrated that PPs synergize with the physiological liver mitogen epidermal growth factor (EGF) to cause the clonal expansion of rat hepatocytes associated with the early stages of tumourigenesis. In the presence of EGF (25 ng/ml), the PP nafenopin (100 microM) was able to stimulate a 5-fold increase in the number of colonies (35 colonies/50,000 hepatocytes compared to seven in the control). EGF alone or nafenopin alone gave 11 and 14 colonies respectively. TGF alpha, which acts through the EGF receptor, also synergized with nafenopin, whereas HGF was inactive, despite its potency as an hepatocyte growth factor. The ability to promote colony formation was shared by the potent PP Wyeth-14,643 but not by the less potent compounds methylclofenapate or trichloroacetic acid. TGF beta, a physiological negative regulator of liver growth, was able to inhibit the nafenopin/EGF-stimulated colony formation at 0.25 ng/ml, a concentration below that required for TGF beta-induced hepatocyte apoptosis. The colonies formed are derived from and consist of hepatocytes, since they express the hepatocyte-specific marker albumin, although the majority are negative for the PP-induced cytochrome, P4504A1. Pre-treatment in vivo with the genotoxic carcinogen dimethylhydrazine hydrochloride (150 mg/kg) caused a doubling in the number of colonies from 35 to 75/50,000 hepatocytes. Taken together, these data suggest that some PPs act as hepatocarcinogens by synergizing with EGF and/or TGF alpha to promote the clonal expansion of spontaneously initiated hepatocytes. This clonal expansion may be inhibited by TGF beta. Such a synergy may provide a mechanistic basis for the hepatocarcinogenicity of this class of non-genotoxic carcinogens.

    Topics: Albumins; Animals; Biomarkers; Carcinogens; Cell Division; Cell Transformation, Neoplastic; Cells, Cultured; Clone Cells; Cocarcinogenesis; Colony-Forming Units Assay; Cytochrome P-450 CYP4A; Cytochrome P-450 Enzyme System; DNA Replication; Drug Synergism; Epidermal Growth Factor; Hyperplasia; Liver; Liver Neoplasms, Experimental; Male; Microbodies; Mixed Function Oxygenases; Nafenopin; Rats; Rats, Wistar; Transforming Growth Factor alpha

1994
Growth factor responsiveness and alterations in growth factor homeostasis in Syrian hamster embryo cells during in vitro transformation.
    Carcinogenesis, 1994, Volume: 15, Issue:6

    Syrian hamster embryo (SHE) cells were investigated for their growth factor responsiveness as well as changes in growth factor homeostasis, including alterations in autocrine growth factor production and growth factor responsiveness, during in vitro transformation. For wild-type SHE cells, fetal bovine serum (FBS), epidermal growth factor (EGF) family members, platelet derived growth factor (PDGF) family members, fibroblast growth factor family members, interleukin-4, interleukin-9, oncostatin M, hepatocyte growth factor, erythropoietin and pituitary extract were found to be mitogenic. SHE cell mitogenesis was inhibited in response to transforming growth factor beta (TGF-beta) family members, interleukin-1 alpha, interleukin-1 beta and nerve growth factor. Additional experiments were conducted to study alterations in growth factor responsiveness to three SHE cell mitogens (FBS, EGF and PDGF) and one inhibitor of mitogenesis (TGF-beta) during SHE cell in vitro transformation. Alterations in either EGF, PDGF or TGF-beta responsiveness were observed in 7/8 SHE transformed lineages during the stepwise transformation process. Finally, 6/8 lineages underwent alterations which resulted in the production of autocrine growth factors during the transformation process. These results indicate that multiple alterations in growth factor homeostasis occur during the in vitro transformation process.

    Topics: Animals; Cell Division; Cell Line, Transformed; Cell Transformation, Neoplastic; Cells, Cultured; Cricetinae; Epidermal Growth Factor; Growth Substances; Homeostasis; Mesocricetus; Platelet-Derived Growth Factor; Transforming Growth Factor alpha; Transforming Growth Factor beta

1994
A specific inhibitor of the epidermal growth factor receptor tyrosine kinase.
    Science (New York, N.Y.), 1994, Aug-19, Volume: 265, Issue:5175

    A small molecule called PD 153035 inhibited the epidermal growth factor (EGF) receptor tyrosine kinase with a 5-pM inhibition constant. The inhibitor was specific for the EGF receptor tyrosine kinase and inhibited other purified tyrosine kinases only at micromolar or higher concentrations. PD 153035 rapidly suppressed autophosphorylation of the EGF receptor at low nanomolar concentrations in fibroblasts or in human epidermoid carcinoma cells and selectively blocked EGF-mediated cellular processes including mitogenesis, early gene expression, and oncogenic transformation. PD 153035 demonstrates an increase in potency over that of other tyrosine kinase inhibitors of four to five orders of magnitude for inhibition of isolated EGF receptor tyrosine kinase and three to four orders of magnitude for inhibition of cellular phosphorylation.

    Topics: 3T3 Cells; Animals; Cell Transformation, Neoplastic; Epidermal Growth Factor; ErbB Receptors; Fibroblast Growth Factor 2; Gene Expression; Humans; Kinetics; Mice; Mitosis; Phosphorylation; Platelet-Derived Growth Factor; Protein-Tyrosine Kinases; Quinazolines; Tumor Cells, Cultured; Tyrosine

1994
Tumorigenicity of SV40 T antigen immortalized human prostate epithelial cells: association with decreased epidermal growth factor receptor (EGFR) expression.
    International journal of cancer, 1994, Sep-01, Volume: 58, Issue:5

    Our primary objectives were to: 1) develop a system for the study of prostatic tumor evolution; and 2) examine the role of the epidermal growth factor/epidermal growth factor receptor (EGF/EGFR) pathway in prostate tumor progression. Adult human prostate epithelial cells previously immortalized by transfection with the SV40 T antigen gene (P69SV40T) produced tumors in only 2/18 mice with a 6 month latency period. Reinjection of cells recovered from these tumors after 1 or 2 cycles of growth in nude mice produced tumors in 2/4 and 2/3 mice with markedly decreased latent intervals of 12, 25, 25 and 25 days each. The chromosomal complement of each tumor was human, consistently pseudodiploid, and retained the Y chromosome. In both anchorage-independent and adherent cell growth assays, EGF stimulated proliferation by approximately 2-fold in both the parental P69SV40T line and the tumor sublines. The tumor sublines expressed less EGFR protein than the parental line, as assessed by Western immunoblotting and flow cytometric analysis. Immunoprecipitation revealed increased production of the 18 and 25 kDa TGF-alpha precursors parallel to decreases in detectable EGFR. The growth of both the parental P69SV40T line and the tumor sublines was inhibited by a neutralizing antibody to TGF-alpha under serum-free defined conditions. Inclusion of the TGF-alpha neutralizing antibody consistently inhibited the proliferation of the tumor sublines more than P69SV40T in both proliferation and [3H]thymidine incorporation assays. This finding suggests that the increased tumorigenicity and decreased latent interval observed among the human prostate tumor cells is partially due to activation of the TGF-alpha/EGFR autocrine network.

    Topics: Animals; Antigens, Polyomavirus Transforming; Cell Adhesion; Cell Division; Cell Transformation, Neoplastic; Epidermal Growth Factor; ErbB Receptors; Humans; Karyotyping; Male; Mice; Mice, Nude; Neoplasm Transplantation; Prostatic Neoplasms; Transforming Growth Factor alpha; Tumor Cells, Cultured

1994
Identification of a cell-surface glycoprotein associated with normal mammary and extramammary epithelial cells.
    British journal of cancer, 1994, Volume: 69, Issue:3

    The goal of the study was to identify any normal genes that may become inactivated in malignant cells, with associated modifications or loss of gene products. Consequently, attempts were made to identify such products by generating monoclonal antibodies using an immune tolerisation-immunisation procedure. Using such a technique, a plasma membrane-associated glycoprotein with an apparent molecular weight of 92 kDa was identified. The glycoprotein was termed luminal epithelial antigen (LEA.92). The pattern of expression of LEA.92 was demonstrated by an indirect immunostaining technique. Using an in vitro model system representing various stages of breast oncogenesis, LEA.92 was detected on normal or immortalised mammary epithelial cell (MEC) lines which were dependent on epidermal growth factor (EGF) and anchorage formation for growth and non-tumorigenic in nude mice. In contrast, LEA.92 was undetectable on oncogenically transformed or established lines of mammary carcinoma cell lines which were independent of EGF or anchorage formation for growth and were highly tumorigenic. The results appear to suggest a correlation between the down-regulation of LEA.92 and the development of tumorigenicity in malignant MEC lines. Furthermore, the patterns of expression of LEA.92 on breast cells in tissue mirrored those of breast epithelial cells in cell cultures. LEA.92 was detected on the surface of normal but not malignant epithelial cells, which included breast, cervix, colon, lung, pancreas and stomach. LEA.92 appeared to be distinct from receptor for epidermal growth factor, antigens associated with milk fat globule membrane and the family of epithelium-specific keratins.

    Topics: Adult; Animals; Antibodies, Monoclonal; Breast; Breast Neoplasms; Cell Transformation, Neoplastic; Electrophoresis, Polyacrylamide Gel; Epidermal Growth Factor; Epithelial Cells; Epithelium; Female; Humans; Immunohistochemistry; Membrane Glycoproteins; Mice; Mice, Nude; Molecular Weight; Transplantation, Heterologous

1994
Expression of cripto in human pancreatic tumors.
    Japanese journal of cancer research : Gann, 1994, Volume: 85, Issue:2

    The expression of cripto gene product was examined immunohistochemically in 45 surgically resected pancreatic tumors, including 32 invasive ductal carcinomas, 4 intraductal papillary adenocarcinomas, 4 intraductal papillary adenomas, 2 mucinous cystadenomas, 2 islet cell tumors, and one solid and cystic tumor, and compared with that in 32 areas of accompanying chronic pancreatitis present in the cases of invasive ductal carcinomas and 5 non-tumorous areas of pancreas without pancreatitis. All pancreatic ductal tumors including adenomas and carcinomas showed positive staining with no difference in terms of staining intensity among intraductal tumors and invasive carcinomas with or without mucin hypersecretion. Islet cell tumors were positively stained but the solid and cystic tumor was negative. Duct epithelial cells and acinar cells were negative but islet cells were positive in the pancreas tissues without pancreatitis. Cells arranged in duct-like structures in areas of accompanying chronic pancreatitis were positively stained. The results suggest that cripto expression might be associated with a growth advantage of tumor cells and also with differentiation to form duct-like structures.

    Topics: Biomarkers, Tumor; Carcinoma; Carcinoma, Islet Cell; Cell Transformation, Neoplastic; Cystadenoma, Mucinous; Epidermal Growth Factor; Gene Expression; GPI-Linked Proteins; Humans; Intercellular Signaling Peptides and Proteins; Islets of Langerhans; Membrane Glycoproteins; Neoplasm Proteins; Pancreatic Ducts; Pancreatic Neoplasms; Pancreatitis

1994
Regulated expression of Wnt family members during proliferation of C57mg mammary cells.
    Cell growth & differentiation : the molecular biology journal of the American Association for Cancer Research, 1994, Volume: 5, Issue:2

    At least six members of the Wnt gene family are expressed in the murine mammary gland during growth and differentiation, whereas several other Wnt family members participate in malignant transformation of this tissue. We have used the C57mg mammary cell line, which naturally expresses the Wnt-4 and Wnt-5a genes, to examine Wnt gene expression during proliferation. The data show that the growth factors basic fibroblast growth factor, transforming growth factor beta 1, and epidermal growth factor are mitogenic for C57mg cells, and partial transformation by Wnt-1 can substitute for the proliferative signal provided by these factors. Several different mitogenic stimuli selectively down-modulate the levels of endogenous Wnt-4 and Wnt-5a RNA in C57mg cells. Partial transformation by either Wnt-1 or Wnt-2 is accompanied by a dramatic decrease in Wnt-4 RNA and a small decrease in Wnt-5a RNA. Mitogenic stimulation by basic fibroblast growth factor or partial transformation by Int-2, a fibroblast growth factor family member, also leads to a selective decrease in the levels of endogenous Wnt RNA. No expression of the Wnt-4 and Wnt-5a genes is detectable in C57mg cells that are fully transformed by the activated tyrosine kinase oncogene Neu. In contrast, overexpression of Wnt-5a in C57mg cells does not lead to a transformed phenotype and is not accompanied by a decrease in endogenous Wnt-4 RNA levels. Overexpression of Wnt-5a does lead to a small decrease in endogenous Wnt-5a levels, perhaps through autoregulation. These data indicate that Wnt-4 and Wnt-5a expression in mammary cells is responsive to growth regulatory signals, and the down-modulation of expression of either or both genes correlates with cell proliferation. The inverse correlation between expression of the endogenous Wnt genes and cell proliferation suggests that Wnt-4 and Wnt-5a may participate in restricting the proliferation of C57mg cells.

    Topics: Animals; Cell Division; Cell Line; Cell Line, Transformed; Cell Transformation, Neoplastic; DNA; DNA Replication; Epidermal Growth Factor; Female; Fibroblast Growth Factor 2; Gene Expression Regulation; Kinetics; Mammary Glands, Animal; Mice; Multigene Family; Phenotype; Protein-Tyrosine Kinases; Proto-Oncogene Proteins; Thymidine; Transforming Growth Factor beta; Wnt Proteins; Wnt-5a Protein; Wnt1 Protein; Wnt4 Protein; Zebrafish Proteins

1994
NIH 3T3 cells transfected with a yeast H(+)-ATPase have altered sensitivity to insulin, insulin growth factor-I, and platelet-derived growth factor-AA.
    Journal of cellular physiology, 1994, Volume: 159, Issue:3

    The role of intracellular pH (pHin) in the regulation of cell growth in both normal and transformed cells is a topic of considerable controversy. In an effort to study this relationship NIH 3T3 cells were stably transfected with the gene for the yeast H(+)-ATPase, constitutively elevating their pHin. The resulting cell line, RN1a, has a transformed phenotype: The cells are serum independent for growth, clone in soft agar, and form tumors in nude mice. In the present study, we further characterize this system in order to understand how transfection with this proton pump leads to serum-independent growth, using defined media to investigate the effects of specific growth factors on the transfected and parental NIH 3T3 cells. While both cell lines show similar growth increases in response to platelet-derived growth factor (PDGF)-BB and epidermal growth factor (EGF), they respond differently to insulin, insulin-like growth factor-I (IGF-I) and PDGF-AA. RN1a cells exhibit increased growth at nanomolar concentrations of insulin but the parental cells had only a relatively minor response to insulin at 10 microM. Both cell lines showed some response to IGF-I in the nanomolar range but the response of RN1a cells was much larger. Differences in insulin and IGF-I receptor number alone could not explain these results. The two cell lines also respond differently to PDGF-AA. RN1a cells are relatively insensitive to stimulation by PDGF-AA and express fewer PDGF alpha receptors as shown by Northern blots and receptor-binding studies. We propose a unifying hypothesis in which the H(+)-ATPase activates a downstream element in the PDGF-AA signal transduction pathway that complements insulin and IGF-I signals, while leading to downregulation of the PDGF alpha receptor.

    Topics: 3T3 Cells; Animals; Becaplermin; Cell Division; Cell Line; Cell Line, Transformed; Cell Transformation, Neoplastic; Culture Media, Serum-Free; Epidermal Growth Factor; Hydrogen-Ion Concentration; Insulin; Insulin-Like Growth Factor I; Kidney; Kinetics; Mice; Mice, Nude; Platelet-Derived Growth Factor; Point Mutation; Proto-Oncogene Proteins c-sis; Proton-Translocating ATPases; Rats; Receptor, IGF Type 1; Receptor, Insulin; Receptors, Platelet-Derived Growth Factor; Recombinant Proteins; Saccharomyces cerevisiae; Transfection

1994
Effect of a null mutation of the insulin-like growth factor I receptor gene on growth and transformation of mouse embryo fibroblasts.
    Molecular and cellular biology, 1994, Volume: 14, Issue:6

    Fibroblast cell lines, designated R- and W cells, were generated, respectively, from mouse embryos homozygous for a targeted disruption of the Igf1r gene, encoding the type 1 insulin-like growth factor receptor, and from their wild-type littermates. W cells grow normally in serum-free medium supplemented with various combinations of purified growth factors, while pre- and postcrisis R- cells cannot grow, as they are arrested before entering the S phase. R- cells are able to grow in 10% serum, albeit more slowly than W cells, and with all phases of the cell cycle being elongated. An activated Ha-ras expressed from a stably transfected plasmid is unable to overcome the inability of R- cells to grow in serum-free medium supplemented with purified clones. Nevertheless, even in the presence of serum, R- cells stably transfected with Ha-ras, alone or in combination with simian virus 40 large T antigen, fail to form colonies in soft agar. Reintroduction into R- cells (or their derivatives) of a plasmid expressing the human insulin-like growth factor I receptor RNA and protein restores their ability to grow with purified growth factors or in soft agar. The signaling pathways participating in cell growth and transformation are discussed on the basis of these results.

    Topics: Animals; Cell Cycle; Cell Division; Cell Line, Transformed; Cell Transformation, Neoplastic; Cells, Cultured; Embryo, Mammalian; Epidermal Growth Factor; Genes, ras; Growth Substances; Insulin; Insulin-Like Growth Factor I; Kinetics; Mice; Mutation; Phenotype; Platelet-Derived Growth Factor; Receptor, IGF Type 1; Signal Transduction; Transfection

1994
Epidermal growth factor (EGF) modulation of feline sarcoma virus fms tyrosine kinase activity, internalization, degradation, and transforming potential in an EGF receptor/v-fms chimera.
    Journal of virology, 1994, Volume: 68, Issue:1

    The feline sarcoma virus oncogene v-fms has significantly contributed to the dissection of peptide growth factor action since it encodes the transmembrane tyrosine kinase gp140v-fms, a transforming version of colony-stimulating factor 1 receptor, a member of the growth factor receptor tyrosine kinase family. In this study, the functional significance of structural differences between distinct tyrosine kinase types, in particular between cellular receptors and viral transforming proteins of distinct structural types, has been further investigated, and their functional compatibility has been addressed. For this purpose, major functional domains of three structurally distinct tyrosine kinases were combined into two chimeric receptors. The cytoplasmic gp140v-fms kinase domain and the kinase domain of Rous sarcoma virus pp60v-src were each fused to the extracellular ligand-binding domain of the epidermal growth factor (EGF) receptor to create chimeras EFR and ESR, respectively, which were studied upon stable expression in NIH 3T3 fibroblasts. Both chimeras were faithfully synthesized and routed to the cell surface, where they displayed EGF-specific, low-affinity ligand-binding domains in contrast to the high- and low-affinity EGF-binding sites of normal EGF receptors. While the EFR kinase was EGF controlled for autophosphorylation and substrate phosphorylation in vitro, in vivo, and in digitonin-treated cells, the ESR kinase was not responsive to EGF. While ESR appeared to recycle to the cell surface upon endocytosis, EGF induced efficient EFR internalization and degradation, and phorbol esters stimulated protein kinase C-mediated downmodulation of EFR. Despite its ligand-inducible kinase activity, EFR was partly EGF independent in mediating mitogenesis and cell transformation, while ESR appeared biologically inactive.

    Topics: 3T3 Cells; Animals; Base Sequence; Biological Transport; Cell Adhesion; Cell Division; Cell Transformation, Neoplastic; Down-Regulation; Enzyme Activation; Epidermal Growth Factor; ErbB Receptors; Gene Expression Regulation, Enzymologic; Gene Expression Regulation, Viral; Growth Substances; Mice; Molecular Sequence Data; Oncogene Protein gp140(v-fms); Oncogene Protein pp60(v-src); Protein-Tyrosine Kinases; Recombinant Fusion Proteins; Sarcoma Viruses, Feline; Transfection

1994
Blocking of tumor promoter-induced AP-1 activity inhibits induced transformation in JB6 mouse epidermal cells.
    Proceedings of the National Academy of Sciences of the United States of America, 1994, Jan-18, Volume: 91, Issue:2

    AP-1 transcriptional activity is stimulated by the transformation promoters phorbol 12-myristate 13-acetate ("12-O-tetradecanoylphorbol 13-acetate," TPA) and epidermal growth factor (EGF) in promotion-sensitive (P+) but not in promotion-resistant (P-) JB6 mouse epidermal cell lines. Although TPA stimulates expression of the jun and fos family genes, only c-jun expression shows higher elevation in P+ cells than in P- cells. The present study tests the hypothesis that induced AP-1 activity is required for tumor promoter-induced transformation in JB6 P+ cells. Both retinoic acid and the glucocorticoid fluocinolone acetonide inhibited basal and TPA-induced AP-1 activities that were tested with a stromelysin promoter-chloramphenicol acetyltransferase reporter gene in P+ cells. Since both retinoic acid and fluocinolone acetonide are active in inhibiting TPA-induced anchorage-independent transformation of P+ cells in the dose range that blocks TPA-induced AP-1 activity, their antipromoting effects may occur through inhibition of AP-1 activity. To test the hypothesis with a more specific inhibitor, stable clonal transfectants of P+ cells expressing dominant negative c-jun mutant encoding a transcriptionally inactive product were analyzed. All transfectants showed a block in TPA and EGF induction of AP-1 activity. All transfectants also showed inhibition of TPA-induced transformation, and most transfectants showed a block in EGF-induced transformation. These results indicate that AP-1 activity is required for TPA- or EGF-induced transformation. This work demonstrates that a specific block in induced AP-1 activity inhibits tumor promoter-induced transformation.

    Topics: Animals; Base Sequence; Binding Sites; Cell Line; Cell Transformation, Neoplastic; DNA; Epidermal Growth Factor; Fluocinolone Acetonide; Gene Expression; Genes, fos; Genes, jun; Genes, Reporter; Mice; Molecular Sequence Data; Mutagenesis, Site-Directed; Point Mutation; Proto-Oncogene Proteins c-jun; Skin; Tetradecanoylphorbol Acetate; Transfection; Tretinoin

1994
Growth-regulatory effects of sensory neuropeptides, epidermal growth factor, insulin, and somatostatin on the non-transformed intestinal epithelial cell line IEC-6 and the colon cancer cell line HT 29.
    Scandinavian journal of gastroenterology, 1993, Volume: 28, Issue:10

    A non-transformed small-intestinal cell line from the rat (IEC-6) and a human colon cancer cell line (HT 29) were examined for their trophic response to sensory neuropeptides. Substance P, neurokinin A (NKA), calcitonin gene-related peptide (CGRP), vasoactive intestinal peptide (VIP), and peptide YY (PYY) were tested. Epidermal growth factor (EGF), insulin, and somatostatin-14 were also used. Interaction studies were performed on IEC-6 cells by combining EGF or insulin with somatostatin-14. The sensory neuropeptides had no effect either on IEC-6 cell growth and DNA synthesis or on HT29 cell growth. EGF and insulin stimulated cell growth and DNA synthesis in IEC-6 cells and cell growth in HT 29 cells in a dose-dependent fashion. Somatostatin-14 had no effect either alone or in combination with EGF or insulin on IEC-6 cell growth and DNA synthesis. HT 29 cell growth was inhibited by somatostatin-14 only in the presence of serum with a maximal and significant response at 10(-7) M. Our observations suggest that the sensory neuropeptides do not exert a direct growth-regulatory effect either on IEC-6 cells or on HT 29 cells. Somatostatin, however, inhibits serum-induced HT 29 cell growth but does not interfere directly with the proliferative effect of serum, EGF, or insulin on IEC-6 cells in this model.

    Topics: Animals; Calcitonin Gene-Related Peptide; Cell Division; Cell Line, Transformed; Cell Transformation, Neoplastic; Cells, Cultured; Colonic Neoplasms; DNA, Neoplasm; Dose-Response Relationship, Drug; Epidermal Growth Factor; Epithelium; Gastrointestinal Hormones; Humans; Insulin; Intestinal Mucosa; Intestines; Neurokinin A; Neurons, Afferent; Neuropeptides; Peptide YY; Peptides; Rats; Somatostatin; Substance P; Transforming Growth Factor beta; Vasoactive Intestinal Peptide

1993
Inactivation of a tumor suppressor function in immortal Syrian hamster cells by N-methyl-N'-nitro-N-nitrosoguanidine and by 5-aza-2'-deoxycytidine.
    Carcinogenesis, 1993, Volume: 14, Issue:2

    Clonal lines of immortal Syrian hamster cells were previously isolated that either suppressed (supB+) tumorigenicity in hybrids with a malignant hamster cell line (BP6T) or had lost this suppression ability (supB-). Neither line was tumorigenic or showed anchorage-independent growth in normal growth medium. SupB- cells, but not supB+ cells, grew in agar supplemented with the growth factors EGF, PDGF and insulin (EPI), providing a selective assay for the supB- phenotype. After treatment of supB+ cells with either N-methyl-N'-nitro-N-nitrosoguanidine (10-300 ng/ml) or 5-aza-2'-deoxycytidine (25-250 ng/ml), and an expression period of 4-8 weeks, a dose-dependent increase in altered cells that grew in agar supplemented with EPI was observed. Cell lines derived from colonies in agar showed persistent EPI-stimulated growth in agar, and decreased suppression of growth in agar for hybrids with BP6T cells. Thus, carcinogen-induced loss of the tumor suppressor phenotype has been demonstrated.

    Topics: Animals; Antineoplastic Agents; Azacitidine; Cell Adhesion; Cell Division; Cell Line; Cell Transformation, Neoplastic; Cells, Cultured; Cricetinae; Decitabine; Embryo, Mammalian; Epidermal Growth Factor; Gene Expression Regulation, Neoplastic; Genes, Tumor Suppressor; Insulin; Mesocricetus; Methylnitronitrosoguanidine; Mice; Mice, Nude; Phenotype; Platelet-Derived Growth Factor

1993
Decreased expression of epidermal growth factor and its receptor in the malignant transformation of trophoblasts.
    Cancer, 1993, Jun-15, Volume: 71, Issue:12

    Gestational trophoblastic neoplasms comprise the neoplastic spectrum of nonmalignant hydatidiform mole, invasive hydatidiform mole, and truly malignant choriocarcinoma. Increasing evidence indicates that epidermal growth factor (EGF) acts as an enhancer of trophoblast function to produce human chorionic gonadotropin and that EGF and its receptor may provide a growth advantage to certain carcinoma cells. The current study was undertaken to evaluate a possible link between malignant transformation of trophoblast and expression of EGF and EGF receptor.. Cytologic localization and cellular levels of expression of EGF and EGF receptor in hydatidiform mole, invasive hydatidiform mole, and choriocarcinoma tissue specimens were examined by the avidin-biotin immunoperoxidase techniques with monoclonal antibodies against EGF and EGF receptor.. EGF in hydatidiform mole and invasive mole was localized in syncytiotrophoblasts, whereas cytologic localization of EGF receptor in hydatidiform mole and invasive mole was observed in both cytotrophoblasts and syncytiotrophoblasts. By contrast, EGF and EGF receptor in choriocarcinoma were exhibited in cytotrophoblastic and syncytiotrophoblastic elements. Most (72%) hydatidiform moles immunostained intensely for EGF and EGF receptor, whereas most (78%) choriocarcinomas immunostained slightly for EGF and EGF receptor. Invasive mole occupied the middle position in the staining intensity for EGF and EGF receptor, between hydatidiform mole and choriocarcinoma, with 50% of the cases exhibiting moderate staining.. The simultaneous expression of EGF and EGF receptor in the neoplastic trophoblasts implies that EGF may act in an autocrine-paracrine manner in trophoblastic neoplasms. Furthermore, the results obtained suggest that cytologic expression of EGF and EGF receptor in trophoblastic neoplasms decreases in the malignant transformation of trophoblast.

    Topics: Cell Transformation, Neoplastic; Epidermal Growth Factor; ErbB Receptors; Female; Gene Expression Regulation, Neoplastic; Humans; Hydatidiform Mole; Hydatidiform Mole, Invasive; Immunoenzyme Techniques; Pregnancy; Staining and Labeling; Trophoblasts; Uterine Neoplasms

1993
SH2-containing phosphotyrosine phosphatase as a target of protein-tyrosine kinases.
    Science (New York, N.Y.), 1993, Mar-12, Volume: 259, Issue:5101

    A mouse phosphotyrosine phosphatase containing two Src homology 2 (SH2) domains, Syp, was identified. Syp bound to autophosphorylated epidermal growth factor (EGF) and platelet-derived growth factor (PDGF) receptors through its SH2 domains and was rapidly phosphorylated on tyrosine in PDGF- and EGF-stimulated cells. Furthermore, Syp was constitutively phosphorylated on tyrosine in cells transformed by v-src. This mammalian phosphatase is most closely related, especially in its SH2 domains, to the corkscrew (csw) gene product of Drosophila, which is required for signal transduction downstream of the Torso receptor tyrosine kinase. The Syp gene is widely expressed throughout embryonic mouse development and in adult tissues. Thus, Syp may function in mammalian embryonic development and as a common target of both receptor and nonreceptor tyrosine kinases.

    Topics: Amino Acid Sequence; Animals; Base Sequence; Cell Line, Transformed; Cell Transformation, Neoplastic; Embryo, Mammalian; Embryonic and Fetal Development; Epidermal Growth Factor; ErbB Receptors; Genes, src; Humans; Intracellular Signaling Peptides and Proteins; Kinetics; Mice; Molecular Sequence Data; Oligodeoxyribonucleotides; Phosphorylation; Platelet-Derived Growth Factor; Poly A; Polymerase Chain Reaction; Protein Tyrosine Phosphatase, Non-Receptor Type 11; Protein Tyrosine Phosphatase, Non-Receptor Type 6; Protein Tyrosine Phosphatases; Protein-Tyrosine Kinases; Rats; Receptors, Platelet-Derived Growth Factor; RNA, Messenger; Sequence Homology, Amino Acid; Transfection

1993
The epidermal growth factor family in pulmonary carcinoids: immunohistochemical evidence of growth-promoting circuits.
    Modern pathology : an official journal of the United States and Canadian Academy of Pathology, Inc, 1993, Volume: 6, Issue:2

    Autocrine neoplastic growth circuits are based on excess synthesis of growth factors and/or cognate membrane receptors. We analyzed by immunohistochemistry 19 typical lung carcinoids for the expression of the epidermal growth factor (EGF), transforming growth factor alpha (TGF alpha), EGF receptor (EGFr), and EGFr-related c-erbB-2 protein (p185). Thirteen tumors (68%) were positive for TGF alpha, 11 for EGFr (58%), three for EGF (16%), and four for p185 (21%). Six carcinoids (32%) were consistently negative for these gene products. The following patterns of coexpression could be documented: TGF alpha, EGFr, EGF, and p185: two cases (11%); TGF alpha, EGFr, and EGF: one case (5%); TGF alpha, EGFr, and p185: two cases (11%); TGF alpha and EGFr: six cases (32%); TGF alpha by itself: two cases (11%). Thus, EGFr was coexpressed with its ligands, TGF alpha and EGF, and the receptor encoded by c-erbB-2 was detected in carcinoids positive for EGFr and TGF alpha. Therefore, alterations of EGF/EGFr-related growth control pathways may be implicated in the pathogenesis of pulmonary carcinoids via the establishment of autocrine growth promoting circuits, as documented in adenocarcinomas and squamous cell carcinomas of the lung.

    Topics: Adenocarcinoma; Carcinoma, Squamous Cell; Cell Transformation, Neoplastic; Epidermal Growth Factor; ErbB Receptors; Humans; Immunohistochemistry; Lung Neoplasms; Proto-Oncogene Proteins; Receptor, ErbB-2; Transforming Growth Factor alpha

1993
Mitogenic activation of the Ras guanine nucleotide exchange factor in NIH 3T3 cells involves protein tyrosine phosphorylation.
    Proceedings of the National Academy of Sciences of the United States of America, 1993, Sep-15, Volume: 90, Issue:18

    We report biochemical evidence that epidermal growth factor and platelet-derived growth factor stimulate the Ras guanine nucleotide exchange factor activity in quiescent NIH 3T3 cells. Moreover, the exchange activity is constitutively enhanced in NIH 3T3 cells transformed by Src and ErbB2 oncogenic tyrosine protein kinases (TPKs), whereas transformation by oncogenic Mos and Raf does not alter the activity. GTPase-activating protein activity was not affected under these conditions. Overexpression of pp60c-Src mutants containing activated and suppressor TPK mutations resulted in stimulation and inhibition of the exchange factor activity, respectively. A TPK inhibitor, genistein, prevented the activation of the exchange factor in epidermal growth factor/platelet-derived growth factor-treated cells and src-transformed cells. Furthermore, the exchange factor activity bound to an anti-phosphotyrosine antibody immunoaffinity column. These findings suggest that the guanine nucleotide exchange factor, but not GTPase-activating protein, plays a major role in the Ras activation in cell proliferation initiated by growth factor receptor TPKs and malignant transformation by oncogenic TPKs and that tyrosine phosphorylation of either the exchange factor or a tightly bound protein(s) may mediate the activation of the exchange factor by these TPKs.

    Topics: 3T3 Cells; Amino Acid Sequence; Animals; Cell Line, Transformed; Cell Transformation, Neoplastic; Chromatography, Affinity; Chromatography, Ion Exchange; Epidermal Growth Factor; ErbB Receptors; Genes, src; Genistein; GTPase-Activating Proteins; Guanine Nucleotide Exchange Factors; Guanosine Diphosphate; Isoflavones; Mice; Mutagenesis, Site-Directed; Phosphorylation; Platelet-Derived Growth Factor; Point Mutation; Protein-Tyrosine Kinases; Proteins; Proto-Oncogene Proteins; Proto-Oncogenes; ras GTPase-Activating Proteins; ras Guanine Nucleotide Exchange Factors; Receptor, ErbB-2; Receptors, Platelet-Derived Growth Factor

1993
Synthesis and assembly of the hyaluronan-containing coats around normal human mesothelial cells.
    Experimental cell research, 1993, Volume: 208, Issue:2

    In this study we examined the capacity of normal human mesothelial (NHM) cells and human malignant mesothelioma cells to form hyaluronan-containing pericellular matrices or "coats." The assembly of the pericellular coats was visualized by a particle exclusion assay. We found that large hyaluronan-containing coats were formed around NHM cells whereas their transformed counterparts had no or very limited coats. The coats were removed by treatment with Streptomyces hyaluronidase, which specifically degrades hyaluronan. NHM cells exhibited hyaluronan-containing pericellular matrix within 5 h after seeding. The formation of the coats was stimulated by platelet-derived growth factor and epidermal growth factor. Interestingly, the assembly of the hyaluronan-dependent pericellular matrices was inhibited by the addition of hyaluronan dodecasaccharides. The inhibitory effect on the formation of the coats was due to a destabilization of pericellular matrix and not due to an inhibitory effect of hyaluronan dodecasaccharides on hyaluronan synthesis. In contrast, hyaluronan hexasaccharides, an inhibitor of the interaction between polymeric hyaluronan and its cell surface receptors, had no effect on the size of the coat. Thus, our results are compatible with the possibility that the pericellular matrix surrounding NHM cells consists of newly synthesized hyaluronan which is extruded from the cell and independent of hyaluronan receptors on the cell surface. The coat seems to be stabilized by interactions (hyaluronan-hyaluronan or hyaluronan-protein bridges) which can be prevented by hyaluronan dodecasaccharides.

    Topics: Cell Transformation, Neoplastic; Epidermal Growth Factor; Epithelium; Extracellular Matrix; Humans; Hyaluronic Acid; In Vitro Techniques; Mesothelioma; Platelet-Derived Growth Factor

1993
The E5 oncoprotein of human papillomavirus type 16 transforms fibroblasts and effects the downregulation of the epidermal growth factor receptor in keratinocytes.
    Journal of virology, 1993, Volume: 67, Issue:8

    To determine the function of the E5 open reading frame (ORF) of the human papillomaviruses (HPVs), rodent fibroblast cell lines were transfected with the E5 ORF of HPV type 6 (HPV-6) and HPV-16 expressed from an exogenous promoter. Transfected fibroblasts were transformed to colony formation in soft agar, and the transformation frequency was increased by epidermal growth factor (EGF) but not by platelet-derived growth factor. In a transitory assay, the E5 ORFs from both HPV-6 and HPV-16 were mitogenic in primary human foreskin epithelial cells (keratinocytes) and acted synergistically with EGF. Investigation of keratinocytes expressing HPV-16 E5 showed that the number of endogenous EGF receptors (EGFRs) per cell was increased two- to fivefold. Immunofluorescence microscopy of HPV-16 E5-expressing keratinocytes indicated that there was an apparent delay in the internalization and degradation of EGFRs compared with controls. Kinetic studies with [125I]EGF showed that the ligand underwent normal internalization and degradation in both HPV-16 E5-expressing and control keratinocytes, but in E5-expressing cells, a greater number of receptors recycled back to the cell surface within 1 to 6 h of ligand binding. Finally, ligand-stimulated phosphorylation of the EGFR on tyrosine, an indication of receptor kinase activity, was of greater magnitude in the HPV-16 E5-expressing keratinocytes than in control cells, although the basal level of receptor phosphorylation was similar.

    Topics: Animals; Cell Line; Cell Transformation, Neoplastic; Cells, Cultured; Down-Regulation; Epidermal Growth Factor; ErbB Receptors; Fibroblasts; Fluorescent Antibody Technique; Humans; Keratinocytes; Kinetics; Oncogene Proteins, Viral; Open Reading Frames; Papillomaviridae; Phosphorylation; Plasmids; Promoter Regions, Genetic; Repressor Proteins; Transfection

1993
Negative regulation of mitogen-stimulated, anchorage-independent cell growth by a tumor-suppressor gene function.
    Molecular carcinogenesis, 1993, Volume: 7, Issue:4

    Immortal, nontumorigenic cell lines of Syrian hamster embryo (SHE) cells with different tumor-suppressing activity were isolated. Subclones from the parental cells were isolated that either had retained (supB+) or lost (supB-) the ability to suppress tumorigenicity after hybridization with tumor cells. The growth properties of these cells were studied to determine how this tumor-suppressor gene function influences cell growth. When the cells were grown on plastic, their growth properties were similar, and neither cell type grew in soft agar containing 10% serum, which supported the growth of tumorigenic cells. However, in agar supplemented with growth factors and 10% serum, supB- cells formed colonies whereas supB+ cells did not. Efficient growth (colony-forming efficiencies greater than 20%) of supB- cells was obtained in agar supplemented with serum and a combination of epidermal growth factor (EGF), platelet-derived growth factor (PDGF), and insulin (EPI) or with serum and basic fibroblast growth factor (bFGF). The effect of EPI and bFGF together was additive. supB+ cells failed to grow under any of these conditions, suggesting that the suppressor gene function blocked the growth response of the cells to multiple growth factors when the cells were suspended in agar. In SupB- cells, transforming growth factor-beta 1 and retinoic acid inhibited anchorage-independent growth response to EPI but not the growth response to bFGF. These observations are consistent with the hypothesis that bFGF stimulates the growth of supB- cells by a signal transduction pathway that differs from the pathway stimulated by EGF or PDGF. Thus, this suppressor gene function may regulate anchorage-independent growth at some common point in signal transduction for multiple mitogens.

    Topics: Animals; Cell Adhesion; Cell Division; Cell Line; Cell Transformation, Neoplastic; Cricetinae; Drug Interactions; Epidermal Growth Factor; ErbB Receptors; Fetus; Fibroblast Growth Factor 2; Genes, Tumor Suppressor; Growth Substances; Insulin; Interferon-gamma; Mesocricetus; Platelet-Derived Growth Factor; Receptor, Insulin; Receptors, Cell Surface; Receptors, Platelet-Derived Growth Factor; Transforming Growth Factor beta; Tumor Necrosis Factor-alpha

1993
Autocrine transforming growth factor-alpha is associated with progression of transformed properties in human colon cancer cells.
    The Journal of biological chemistry, 1993, Jan-05, Volume: 268, Issue:1

    The GEO colon carcinoma cell line is weakly tumorigenic in athymic mice and shows differentiated properties both in tissue culture and in xenografts. Proliferating monolayer cultures of GEO cells which normally require exogenous epidermal growth factor (EGF) for optimal growth displayed a marked inhibition in growth upon addition of antibodies that block binding to the EGF receptor or neutralize TGF-alpha. These results indicated that GEO cells utilize TGF-alpha in a weak autocrine loop. The availability of a weakly malignant model system in which TGF-alpha had demonstrable, but low level autocrine activity, permitted the investigation of the role of TGF-alpha in tumorigenesis by generating a stronger autocrine loop through the overexpression of the polypeptide. GEO cells were electroporated with an expression vector containing the human TGF-alpha cDNA, and stable clones were isolated that constitutively expressed the TGF-alpha cDNA in a strong autocrine loop. However, the growth rate of the parental cells in EGF-supplemented medium was the same as that of transfected cells with or without growth factor-supplemented medium. Thus, any biological changes generated by the overexpression of TGF-alpha were due to the autocrine nature of the growth mechanism rather than due to any decrease in doubling time leading to a faster growth rate. Transfected GEO cells showed an increase in anchorage-independent growth and formed tumors more readily in athymic nude mice indicating that TGF-alpha plays a role in progression of transformed properties.

    Topics: Animals; Blotting, Northern; Blotting, Southern; Cell Division; Cell Transformation, Neoplastic; Clone Cells; Colonic Neoplasms; DNA; Epidermal Growth Factor; ErbB Receptors; Female; Humans; Kinetics; Mice; Mice, Nude; RNA, Messenger; Transfection; Transforming Growth Factor alpha; Transplantation, Heterologous; Tumor Cells, Cultured

1993
Identification of a nerve growth factor- and epidermal growth factor-regulated protein kinase that phosphorylates the protooncogene product c-Fos.
    Proceedings of the National Academy of Sciences of the United States of America, 1993, Jan-15, Volume: 90, Issue:2

    Nerve growth factor (NGF) treatment of rat pheochromocytoma (PC12) cells induces the synthesis of the transcription factor c-Fos, which becomes highly phosphorylated relative to that produced as a result of depolarization of the cell. A peptide derived from the carboxyl terminus of c-Fos (residues 359-370, RKGSSSNEPSSD) containing putative phosphorylation sites was used to detect a NGF-stimulated Fos kinase. NGF treatment of PC12 cells resulted in a rapid activation of a protein kinase which phosphorylated both the c-Fos peptide and authentic c-Fos at its carboxyl terminus. The kinase was selectively activated by NGF and epidermal growth factor but was not induced by depolarization or other agents. The c-Fos peptide was phosphorylated at a serine corresponding to Ser362, a site critically implicated in the capacity of c-Fos to exhibit transrepressive activity [Ofir, R., Dwarki, V. J., Rashid, D. & Verma, I. M. (1990) Nature (London) 348, 80-82)]. The NGF-stimulated Fos kinase may play an important role in regulating the expression and transforming potential of c-Fos.

    Topics: Amino Acid Sequence; Animals; Cell Transformation, Neoplastic; Enzyme Activation; Epidermal Growth Factor; Fibroblast Growth Factors; Ligands; Molecular Sequence Data; Nerve Growth Factors; PC12 Cells; Peptide Fragments; Protein Serine-Threonine Kinases; Proto-Oncogene Proteins c-fos; Ribosomal Protein S6 Kinases, 90-kDa; Serine; Signal Transduction

1993
Effects of five phorbol esters on gap junctional intercellular communication, morphological transformation and epidermal growth factor binding in Syrian hamster embryo cells.
    Carcinogenesis, 1993, Volume: 14, Issue:1

    The effects of 12-O-tetradecanoylphorbol-13-acetate (TPA), 12-deoxyphorbol-13-phenylacetate (DOPP), 12-deoxyphorbol-13-phenylacetate-20-acetate (DOPP A), sapintoxin D (SAP D) and sapintoxin A (SAP A) on the decrease in [125I]epidermal growth factor (EGF) binding (indicating protein kinase C activation), suppression of gap junctional intercellular communication (GJIC) and induction of morphological cell transformation (MCT) in Syrian hamster embryo (SHE) cells were investigated. All five phorbol esters were found to reduce [125I]EGF binding in early passage SHE cells at comparable concentrations. DOPP A was approximately 10-fold less potent in decreasing GJIC compared to the other phorbol esters in early passage SHE cells, while the compounds showed less difference in suppressing GJIC in the phorbol ester sensitive SHE cell line BPNi. The decreases in [125I]EGF binding and GJIC were found to be transient in the continuous presence of phorbol esters. All phorbol esters induced MCT in early passage SHE cells, but DOPP and DOPP A were approximately 10-fold less potent than TPA, SAP D and SAP A. Thus, there seems to be some degree of correlation, but not to a full extent, between the ability of the phorbol esters to activate PKC, decrease GJIC and to induce MCT. The results do not suggest a simple relationship between PKC activation, inhibition of GJIC and the reported tumor-promoting activities of the compounds.

    Topics: Animals; Cell Communication; Cell Line, Transformed; Cell Transformation, Neoplastic; Cells, Cultured; Cricetinae; Enzyme Activation; Epidermal Growth Factor; Intercellular Junctions; Mesocricetus; Phorbol Esters; Protein Kinase C

1993
Orthovanadate both mimics and antagonizes the transforming growth factor beta action on normal rat kidney cells.
    Journal of cellular physiology, 1993, Volume: 154, Issue:2

    Normal rat kidney [NRK] cells grown in the presence of epidermal growth factor (EGF) or platelet-derived growth factor (PDGF) have a normal phenotype and undergo density-dependent growth inhibition, whereas in the presence of multiple growth factors, density arrest is lost and the cells become phenotypically transformed. We studied the influence of the protein tyrosine phosphatase (PTPase) inhibitor sodium orthovanadate on the mitogenic stimulation of NRK cells by growth factors and on transformation-linked properties as loss of density-dependent growth inhibition and anchorage-independent growth. The fraction of cells in serum-deprived monolayer cultures that is induced to proliferate upon mitogenic stimulation by EGF or PDGF is only slightly enhanced upon addition of low concentrations (25-50 microM) of vanadate. Addition of vanadate per se induces proliferation of only a very limited amount of cells, but results in a shift of the dose-response curves for other growth factors to lower concentrations. Vanadate added in combination with EGF or PDGF is able to mimic the effect of transforming growth factor beta (TGF beta) in inducing phenotypic transformation. In monolayer cultures density-dependent growth inhibition is lost and anchorage-independent proliferation is observed on dishes coated with poly(2-hydroxy-ethyl methacrylate) (polyHEMA). The extent of these changes is similar to that induced by TGF beta. However, the morphology of the obtained colonies in polyHEMA-coated dishes is quite different. Cells transformed by TGF beta in the presence of EGF form rather amorphous colonies, whereas in the presence of orthovanadate colonies are formed that tend to fall apart in loose cells. The effect of vanadate on cell transformation is dependent on the growth factor conditions in a bimodal way. When a suboptimal dose of growth factor(s) is used, 25 microM vanadate is very effective in preventing density-induced growth inhibition and stimulating anchorage-independent proliferation. However, the same concentration of vanadate is inhibitory when cells are maximally stimulated and antagonizes the transforming effect of TGF beta added in combination with other growth factors. It is hypothesized that vanadate acts on a set of different protein tyrosine phosphatases. Some of these are positive and others negative regulators of growth.

    Topics: Animals; Cell Adhesion; Cell Transformation, Neoplastic; Cells, Cultured; Drug Interactions; Epidermal Growth Factor; Kidney; Platelet-Derived Growth Factor; Rats; Transforming Growth Factor beta; Vanadates

1993
Single point mutations in the SH2 domain impair the transforming potential of vav and fail to activate proto-vav.
    Oncogene, 1993, Volume: 8, Issue:7

    The importance of an intact Src Homology 2 (SH2) domain for transformation by members of the tyrosine kinase family, including v-src, c-src, c-abl, fps and fyn is well documented. To determine the role of the SH2 domain in transformation by a protein which is not a member of this family, we employed site directed mutagenesis to change four highly conserved residues in the SH2 domain of the vav oncogene and the vav proto-oncogene (proto-vav). Proto-vav encodes a protein that contains one SH2 domain and two Src Homology 3 (SH3) domains, in addition to a number of other motifs usually found in transcriptional factors and guanine nucleotide exchange factors. Substitution of arginine 629 to glycine (R629G) and arginine 647 to leucine (R647L) in vav did not impair its transforming potential in NIH3T3 fibroblasts. By contrast, substitutions of tryptophan 622 to arginine (W622R) and glycine 642 to valine (G642V) in the vavSH2, greatly reduced its transforming potential. Similar point mutations introduced in the SH2 domain of proto-vav did not activate the transforming potential of the normal gene. Interestingly, although all the vav SH2 mutant proteins were constitutively phosphorylated on tyrosine when expressed in NIH3T3 cells, they fail to bind to a phosphorylated epidermal growth factor receptor (EGFR), regardless of their transforming potential.

    Topics: 3T3 Cells; Amino Acid Sequence; Animals; Base Sequence; Cell Cycle Proteins; Cell Transformation, Neoplastic; Epidermal Growth Factor; ErbB Receptors; Gene Expression Regulation, Neoplastic; Mice; Molecular Sequence Data; Mutagenesis, Site-Directed; Oncogenes; Phosphorylation; Point Mutation; Proto-Oncogene Proteins; Proto-Oncogene Proteins c-vav; Proto-Oncogenes; Structure-Activity Relationship

1993
Expression of c-fos and c-myc protooncogenes in an immortalized hepatocyte line harbouring SV40 T antigen and hGH as transgenes.
    Transgenic research, 1993, Volume: 2, Issue:2

    A clonal hepatocyte line (FMH-202-2), derived from livers of fetal transgenic mice harbouring human growth hormone (hGH) and SV40 T antigen as transgenes, was used in the investigation of protooncogene expression involved in liver-specific growth control and/or in hepatocellular transformation. In this model system, representing an immortalized, yet untransformed phenotype, the transgenes hGH and SV40 T antigen were expressed constitutively. The c-fos protooncogene was induced by incubation with insulin, epidermal growth factor (EGF) and insulin-like growth factor (IGF-I) in a transient manner comparable to its expression in primary murine hepatocytes. Elucidation of second messenger mechanisms demonstrated that c-fos induction by hepatotrophic growth factors was not mediated by protein kinase C. In contrast to primary hepatocytes, the c-myc protooncogene exhibited a constitutive expression pattern which was independent of growth factor stimulation. These results indicate that apart from hGH and SV40 T antigen, c-myc may play a role in cellular immortalization, but that constitutive expression of these genes, even in combined coexpression, does not suffice to induce the transformed phenotype.

    Topics: Animals; Antigens, Polyomavirus Transforming; Cell Line, Transformed; Cell Transformation, Neoplastic; Epidermal Growth Factor; Gene Expression; Genes, fos; Genes, myc; Growth Hormone; Humans; Insulin; Insulin-Like Growth Factor I; Liver; Mice; Mice, Transgenic; RNA, Messenger

1993
Effects of 5-HT1C-receptor expression on cell proliferation control in hamster fibroblasts: serotonin fails to induce a transformed phenotype.
    Experimental cell research, 1992, Volume: 200, Issue:2

    5-HT1c receptors have been shown to act as protooncogenes in NIH 3T3 cells, inducing ligand-dependent focus formation. In order to assess their mitogenic and oncogenic potential in a different cell system, we transfected these receptors into CCL39 hamster fibroblasts, a well-characterized growth factor-dependent cell line. Cell clones expressing functional receptors were isolated and tested for (a) growth factor dependence of proliferation measuring thymidine incorporation in response to varying doses of serum, (b) the response to serotonin alone or in combination with other growth factors, and (c) the capacity for anchorage-independent proliferation. In the absence or presence of serotonin, the large majority of the clones isolated showed normal morphology and normal growth factor dependence and was unable to grow in soft agar. None of the clones showed a significant response to serotonin alone in DNA synthesis reinitiation experiments, but synergy was observed between serotonin and the tyrosine kinase activating growth factors EGF and FGF. However, the major part of this effect could be abolished by an antagonist of 5-HT1b receptors, which are endogenous in CCL39 cells. The same receptor was found to mediate a significant mitogenic response to the neurotransmitter in Ha-ras-transfected cells. The fact that 5-HT1c receptors do not readily induce a transformed phenotype in CCL39 cells clearly distinguishes them from strong dominantly acting oncogene products like RAS, SRC, or FMS.

    Topics: Animals; Cell Division; Cell Line; Cell Transformation, Neoplastic; Cricetinae; Cricetulus; DNA; Epidermal Growth Factor; Fibroblast Growth Factors; In Vitro Techniques; Phosphatidylinositols; Receptors, Serotonin; Serotonin

1992
Relative resistance to 1,25-dihydroxyvitamin D3 in a keratinocyte model of tumor progression.
    The Journal of biological chemistry, 1992, Jun-15, Volume: 267, Issue:17

    We have examined the effect of 1,25-dihydroxyvitamin D3 (1,25-(OH)2D3) on mitogen-stimulated growth and on c-myc proto-oncogene expression in a keratinocyte model of tumor progression. A dose-dependent inhibition of cell growth by 1,25-(OH)2D3 was demonstrated in both established (HPK1A) and malignant (HPK1A-ras) cells. However, this inhibition was observed with the addition of 1,25-(OH)2D3 at a higher concentration in HPK1A-ras cells than in HPK1A cells. Cell cycle analysis revealed a blockage of the normal progression of the cell cycle from G0 to S phase in the presence of 1,25-(OH)2D3. A higher concentration of 1,25-(OH)2D3 was required in HPK1A-ras cells to overcome the mitogen-stimulated progression into S phase, when compared with HPK1A cells. Analysis of c-myc messenger RNA revealed a strong inhibition of its expression at early time points with higher concentrations of 1,25-(OH)2D3 being required to obtain an inhibition in HPK1A-ras cells similar to that obtained in HPK1A cells. 1,25-(OH)2D3 receptor characterization by sucrose gradient analysis and equilibrium binding demonstrated the presence of a single 3.7 S protein with similar receptor numbers and affinity in both cell lines. These observations therefore demonstrate that an alteration of the growth inhibitory response to 1,25-(OH)2D3 occurs when keratinocytes acquire the malignant phenotype and suggest that the alteration lies beyond the interaction of the ligand with its receptor. In addition, relative resistance to 1,25-(OH)2D3 was also observed in the expression of the cell-cycle associated oncogene c-myc. These studies may therefore have important implications in vivo in the development and growth of epithelial cell cancers.

    Topics: Animals; Blotting, Northern; Calcitriol; Cell Division; Cell Line; Cell Transformation, Neoplastic; Epidermal Growth Factor; Flow Cytometry; Genes, myc; Humans; Keratinocytes; Mice; Mice, Nude; Proto-Oncogene Mas; Receptors, Calcitriol; Receptors, Steroid; RNA, Messenger

1992
Progressive abrogation of TGF-beta 1 and EGF growth control is associated with tumour progression in ras-transfected human keratinocytes.
    International journal of cancer, 1992, Sep-30, Volume: 52, Issue:3

    This study examined the response of human keratinocytes in different stages of transformation to exogenous TGF-beta 1 and EGF as well as their receptor and growth-factor expression. Cells of the spontaneously immortalized HaCaT cell line and c-Ha-ras transfected clones (I-6, I-7, II-3, II-4) exhibited different tumorigenic potentials when transplanted to athymic mice. HaCaT- and I-6 cells were non-tumorigenic, I-7 cells formed persisting epidermal cysts (benign tumours) and II-3 and II-4 cells developed into invasive squamous-cell carcinomas. TGF-beta 1 inhibited thymidine uptake in a dose-dependent manner, a progressive decrease in response being associated with an increasing malignant potential (HaCaT greater than I-6 greater than I-7 = II-4). HaCaT-cells and ras-clones expressed TGF-beta 1 mRNA at similar levels, but cells of increasing malignant potential secreted markedly less receptor-binding TGF-beta (HaCaT greater than I-6 = I-7 greater than II-3 greater than II-4) into the culture medium. Whilst ras-transfected cells expressed fewer TGF-beta receptors than HaCaT cells, there was little difference between TGF-beta receptor number or affinity between the 4 transfected cell clones. The same was true for the TGF-beta receptor types, but Type-II receptors were expressed at lower levels by the malignant clones II-3 and II-4. When HaCaT and ras-transfected cells were investigated for their response to exogenous EGF, cells were refractory (I-7, II-4), partially stimulated (I-6) or fully stimulated (HaCaT). Cells with increasing malignant potential produced increasing amounts of endogenous TGF-alpha (II-4 = II-3 greater than I-7 = I-6 greater than HaCaT). All tumorigenic ras clones expressed higher mRNA levels than HaCaT-cells. Ras-transfected clones expressed fewer high- and low-affinity EGF receptors than HaCaT cells with a tendency toward increased numbers of high-affinity EGF receptors associated with increasing malignant potential (II-4 = II-3 greater than I-7 greater than I-6) but these changes were associated with a progressive decrease in receptor affinity. The results indicate that tumour progression in human epidermal keratinocytes transfected with c-Ha-ras is associated with a progressive abrogation of TGF-beta 1 and EGF growth control. They suggest that the increased autonomous growth potential associated with advanced stages of epithelial tumour progression can be defined more closely using a cellular profile of TGF-beta and EGF.

    Topics: Cell Division; Cell Transformation, Neoplastic; Epidermal Growth Factor; ErbB Receptors; Genes, ras; Humans; Keratinocytes; Receptors, Cell Surface; Receptors, Transforming Growth Factor beta; Transfection; Transforming Growth Factor alpha; Transforming Growth Factor beta

1992
The SH2 and SH3 domain-containing Nck protein is oncogenic and a common target for phosphorylation by different surface receptors.
    Molecular and cellular biology, 1992, Volume: 12, Issue:12

    Signalling proteins such as phospholipase C-gamma (PLC-gamma) or GTPase-activating protein (GAP) of ras contain conserved regions of approximately 100 amino acids termed src homology 2 (SH2) domains. SH2 domains were shown to be responsible for mediating association between signalling proteins and tyrosine-phosphorylated proteins, including growth factor receptors. Nck is an ubiquitously expressed protein consisting exclusively of one SH2 and three SH3 domains. Here we show that epidermal growth factor or platelet-derived growth factor stimulation of intact human or murine cells leads to phosphorylation of Nck protein on tyrosine, serine, and threonine residues. Similar stimulation of Nck phosphorylation was detected upon activation of rat basophilic leukemia RBL-2H3 cells by cross-linking of the high-affinity immunoglobulin E receptors (Fc epsilon RI). Ligand-activated, tyrosine-autophosphorylated platelet-derived growth factor or epidermal growth factor receptors were coimmunoprecipitated with anti-Nck antibodies, and the association with either receptor molecule was mediated by the SH2 domain of Nck. Addition of phorbol ester was also able to stimulate Nck phosphorylation on serine residues. However, growth factor-induced serine/threonine phosphorylation of Nck was not mediated by protein kinase C. Interestingly, approximately fivefold overexpression of Nck in NIH 3T3 cells resulted in formation of oncogenic foci. These results show that Nck is an oncogenic protein and a common target for the action of different surface receptors. Nck probably functions as an adaptor protein which links surface receptors with tyrosine kinase activity to downstream signalling pathways involved in the control of cell proliferation.

    Topics: 3T3 Cells; Adaptor Proteins, Signal Transducing; Animals; Cell Line; Cell Transformation, Neoplastic; Epidermal Growth Factor; Humans; Mice; Oncogene Proteins; Organ Specificity; Phosphorylation; Platelet-Derived Growth Factor; Precipitin Tests; Protein-Tyrosine Kinases; Rats; Receptors, Cell Surface; Signal Transduction; Sulfhydryl Compounds; Tetradecanoylphorbol Acetate; Transfection; Vanadates

1992
Elongation factor-1 alpha gene determines susceptibility to transformation.
    Nature, 1992, Sep-24, Volume: 359, Issue:6393

    Elongation factor-1 alpha (EF-1 alpha), an essential component of the eukaryotic translational apparatus, is a GTP-binding protein that catalyses the binding of aminoacyl-transfer RNAs to the ribosome. Expression of the EF-1 alpha gene decreases towards the end of the lifespans of mouse and human fibroblasts, but forced expression of EF-1 alpha prolongs the lifespan of Drosophila melanogaster. Eukaryotic initiation factor-4E, another component of the translational machinery, is mitogenic or oncogenic when constitutively expressed in some mammalian cells. Thus, components of the protein synthesis apparatus seem to be involved in the control of cell proliferation. Using expression cloning, we have isolated a complementary DNA clone from a BALB/c 3T3 mouse fibroblast variant, A31-I-13 (ref. 10), which specifies a factor determining the susceptibility of BALB/c3T3 to chemically and physically induced transformation. Here we report that the factor is EF-1 alpha and that its constitutive expression causes BALB/c 3T3 A31-I-1 (ref. 10), C3H10T1/2 (ref. 11) and Syrian hamster SHOK fibroblasts to become highly susceptible to transformation induced by 3-methylcholanthrene and ultraviolet light. EF-1 alpha messenger RNA is also constitutively expressed in a quiescent culture of the highly susceptible variant A31-I-13. We conclude that the removal of regulation of the expression of these components of the translational machinery may predispose cells to become more susceptible to malignant transformation.

    Topics: 3T3 Cells; Animals; Blotting, Northern; Cell Division; Cell Transformation, Neoplastic; DNA Replication; Epidermal Growth Factor; Methylcholanthrene; Mice; Mice, Inbred BALB C; Mice, Inbred C3H; Peptide Elongation Factor 1; Peptide Elongation Factors; Platelet-Derived Growth Factor; RNA; Thymidine; Ultraviolet Rays

1992
Raf-1 protein kinase is an integral component of the oncogenic signal cascade shared by epidermal growth factor and platelet-derived growth factor.
    Molecular and cellular biology, 1992, Volume: 12, Issue:11

    Our recent studies with cell mutants indicate that a cascade shared by the epidermal growth factor (EGF) and platelet-derived growth factor (PDGF) signals exists in NRK cells and mediates oncogenic signals induced by many oncogenes (A. Masuda, S. Kizaka-Kondoh, H. Miwatani, Y. Terada, H. Nojima, and H. Okayama, New Biol. 4:489-503, 1992). We have employed the antisense RNA technique to investigate possible involvement of Raf-1 kinase in this signal transduction cascade. NRK cell clones highly reduced in the Raf-1 production are generated by the expression of a c-raf-1 antisense RNA. They have no apparent growth defects and retain proper mitotic responses to growth factors but are refractory to transformation by EGF or PDGF plus transforming growth factor beta, v-erbB, v-fms, v-K-ras, v-mos, v-fos, v-src, simian virus 40 large T, and polyomavirus middle T but not by v-raf or adenovirus E1A. These results not only support our model for the oncogenic signal cascade but also lead to the conclusion that Raf-1 protein kinase is a downstream component of this oncogenic signal cascade shared by EGF and PDGF.

    Topics: Blotting, Western; Cell Division; Cell Line; Cell Transformation, Neoplastic; Cloning, Molecular; Epidermal Growth Factor; Oncogenes; Platelet-Derived Growth Factor; Protein Serine-Threonine Kinases; Proto-Oncogene Proteins; Proto-Oncogene Proteins c-raf; Signal Transduction; Transforming Growth Factor beta

1992
Polyoma middle T antigen or v-src desensitizes human epidermal growth factor receptor function and interference by a monensin-resistant mutation in mouse Balb/3T3 cells.
    Experimental cell research, 1992, Volume: 203, Issue:2

    Epidermal growth factor (EGF)-induced down-regulation of its receptor is an obligatory pathway for cellular regulation of EGF-specific receptor (EGF-R) in normal and malignant cells. BNER4 cells are mouse Balb/3T3 cells transfected with the human EGF-R complementary DNA (cDNA). Polyoma middle T antigen-transfectants of BNER4, B4/MT-2, B4/MT-13, B4/MT-23, and B4/MT-24, showed diminished down-regulation of cell surface human EGF-R in response to EGF relative to the parental BNER4 cells. Also, the v-src-transfectants B4/SRC-13 and B4/SRC-24 showed much less down-regulation than BNER4 cells, whereas H-ras-transfectants of BNER4, B4/RAS-24 and B4/RAS-25, showed EGF-induced down-regulation of the cell surface EGF-R similar to that of BNER4. EGF induced DNA synthesis more than 20-fold in BNER4, but induced only about a 1.5- to 6-fold increase in the middle T antigen- and v-src-transfectants. EGF-Rs of the middle T antigen-transfectants were metabolically stable in the presence of EGF in comparison with their parental BNER4 cells. EGF-Rs of BNER4 cells degraded with half-lives of about 2 h in the presence of EGF, but those of the middle T antigen transformants were found to be highly stabilized in the presence of EGF. On the other hand, transfection with polyoma middle T antigen (MTAg) cDNA causes malignant transformation of Balb/3T3 cells, but not its monensin (an ionophoric antibiotic)-resistant mutant MO-5 cells, which have no significant EGF binding activity. Transfection of human EGF-R cDNA into MO-5 leads to the expression of high levels of human EGF-R in MNER31. Unlike the polyoma MTAg transfectants of BNER4, EGF-R in polyoma MTAg cDNA-transfectants into MNER31, M31/MT-13 and M31/MT-14, were down-regulated to levels similar to those of their parental MNER31. Exposure to EGF induced a more than 10-fold increase in DNA synthesis of quiescent BNER4, MNER31, M31/MT-13, and M31/MT-14 cells. Polyoma middle T antigen or v-src appears to modulate EGF-induced down-regulation of EGF-R, possibly through interaction of the receptor with the viral oncogenes, and this interaction may be altered in the mutant.

    Topics: 3T3 Cells; Animals; Antigens, Polyomavirus Transforming; Base Sequence; Cell Division; Cell Transformation, Neoplastic; Cell Transformation, Viral; DNA; Down-Regulation; Drug Resistance; Epidermal Growth Factor; ErbB Receptors; Genes, src; Mice; Molecular Sequence Data; Monensin; Mutation; Phosphorylation; Transfection

1992
Epigenetic activation of Gi-2 protein, the product of a putative protooncogene, mediates tumor promotion in vitro.
    Carcinogenesis, 1992, Volume: 13, Issue:12

    Promotion of 'initiated' JB6 epidermal cells to the tumor phenotype can be effected by 12-O-tetradecanoylphorbol-13-acetate treatment, by stimulation of epidermal growth factor (EGF) receptor activity with EGF or transforming growth factor alpha and by exposure to the isoquinoline derivative H7. When these cells were incubated with pertussis toxin (PTX), induction of anchorage-independent growth by all four promoting substances was suppressed. The inhibition is specific since cell proliferation is not affected, suggesting that activation of a Gi protein is essential for promotion of the epidermal cells. This interpretation is strongly supported by the observation that the wasp poison mastoparan, which is known to mimic receptor-mediated activation of certain Gi proteins, also promoted anchorage independence. Immunological data and partial amino acid sequence analysis of ADP-ribosyl alpha i isolated from PTX-treated JB6 cells indicate that a Gi-2 protein is a mediator to tumor promotion in this system. The inhibitory action of 4-bromophenacyl bromide may point to a coupling of the Gi protein to phospholipase A2. From our data we infer that promoters induce the tumor phenotype in 'initiated' JB6 epidermal cells by activating epigenetically the same Gi protein that in a number of adrenal and ovarian tumors appears to be persistently activated by mutational events.

    Topics: Adrenal Gland Neoplasms; Amino Acid Sequence; Animals; Cell Transformation, Neoplastic; Cells, Cultured; Epidermal Growth Factor; Female; GTP-Binding Proteins; Mice; Molecular Sequence Data; Mutation; Ovarian Neoplasms; Pertussis Toxin; Phenotype; Phospholipases A; Phospholipases A2; Proto-Oncogene Proteins; Rats; Sequence Homology, Amino Acid; Tetradecanoylphorbol Acetate; Transforming Growth Factor alpha; Virulence Factors, Bordetella

1992
Signal transduction cascade shared by epidermal growth factor and platelet-derived growth factor is a major pathway for oncogenic transformation in NRK cells.
    The New biologist, 1992, Volume: 4, Issue:5

    We have isolated two recessive, mutually complementary NRK cell mutants that are refractory to transformation by epidermal growth factor (EGF) and transforming growth factor-beta. Both mutants are defective in a signal transduction cascade shared by EGF and platelet-derived growth factor (PDGF). Analysis of the mutants suggests that transformation of NRK cells by the v-fms, v-erbB, activated erbB-2, v-ras, v-fos, v-mos, v-fes, v-src, SV40 large T, polyomavirus middle T, and human papillomavirus type 16 E6,E7 oncogenes is mediated by the EGF/PDGF signal cascade. The data also suggest that the EGF/PDGF cascade branches into mitogenic and oncogenic signals, the latter of which is required for soft agar growth and focus formation.

    Topics: Animals; Cell Division; Cell Line; Cell Transformation, Neoplastic; Epidermal Growth Factor; ErbB Receptors; Gene Expression; Mutation; Oncogenes; Platelet-Derived Growth Factor; Signal Transduction; Transfection

1992
Response of FR3T3 cells transformed by Ha-ras oncogene and epidermal growth factor gene to differentiation induction by N,N-dimethylformamide.
    International journal of cancer, 1992, Feb-20, Volume: 50, Issue:4

    Treatment of chemically transformed fibroblasts with N,N-dimethylformamide (DMF) results in the restoration of a nontransformed phenotype. In an attempt to identify more precisely the mechanisms by which DMF reverses the transformed phenotype, the effects of DMF on fibroblasts which were transformed by a single gene--specifically a synthetic epidermal growth factor (EGF) gene or the Ha-ras oncogene--were examined. The constitutive expression of either the Ha-ras oncogene or the EGF gene in FR3T3 fibroblasts resulted in cellular transformation. The effect of the differentiation-inducing agent DMF on several properties of these transformed cell lines was examined. The EGF-transfected cells were much more responsive to DMF than the ras-transfected cells. DMF treatment of the EGF-transfected cells resulted in the inhibition of anchorage-independent growth, the restoration of a normal cellular morphology and growth rate in monolayer culture, and the down-regulation of the proliferation-associated nucleolar protein B23. DMF treatment had a much slighter effect on growth of the ras-transfected cells in monolayer culture or under anchorage-independent growth conditions. The high proliferation rate of the ras-3 cells was associated with elevated expression of protein B23. The 19-3 cells, but not the ras-3 cells, expressed cell-surface fibronectin. Treatment of the ras-3 cells with DMF did not restore fibronectin expression. The binding of EGF was increased 3-fold in the EGF-transfected cells and decreased 20-fold in ras-transfected cells, but in neither case did DMF alter EGF binding. DMF treatment increased the secretion of EGF in the 2 transfected lines as well as in control cells. These results suggest that the aberrant-growth control in the EGF-transfected cells, but not in the ras-transfected cells, could be modulated by DMF and that the aberrant-growth control mechanisms were different in these 2 cell types.

    Topics: Animals; Cell Differentiation; Cell Division; Cell Transformation, Neoplastic; Dimethylformamide; Epidermal Growth Factor; ErbB Receptors; Fibronectins; Genes, ras; In Vitro Techniques; Mice; Nuclear Proteins; Nucleophosmin; Proto-Oncogene Proteins p21(ras)

1992
Differential c-jun expression in response to tumor promoters in JB6 cells sensitive or resistant to neoplastic transformation.
    Molecular carcinogenesis, 1992, Volume: 5, Issue:1

    The activity of AP-1, a trans-acting transcription factor, is stimulated by 12-O-tetradecanoylphorbol-13-acetate (TPA) and epidermal growth factor (EGF) in promotion-sensitive (P+) but not in promotion-resistant (P-) JB6 mouse epidermal cell lines. TPA and EGF also promote neoplastic transformation only in P+ cells. Thus, it has been proposed that AP-1-dependent gene expression is involved in determining sensitivity to tumor promotion. This paper explores the possible basis for the differential inducibility of AP-1 activity in P+ and P- JB6 cells, focusing in particular on the regulation of expression of the components of the AP-1 complex at the mRNA level. The expression of jun and fos gene family members, which make up the AP-1 complex, can be stimulated by serum and a number of growth factors, including EGF, and by TPA. Therefore, the possibility that differential expression of one or more forms of jun or fos contributes to the differential AP-1 activity was considered. The data presented here demonstrate both similarities and differences in the basal and TPA- or EGF-induced levels of fos and jun family members between P+ and P- cells. The most striking observation was that the overall TPA- and EGF-induced levels of jun but not fos expression were higher in P+ cells. This suggests that tumor promoter-regulated c-jun expression may contribute to the differential AP-1 activation observed in these cells and may be important in determining sensitivity to promotion of neoplastic transformation.

    Topics: Animals; Carcinogens; Cell Transformation, Neoplastic; Cells, Cultured; Epidermal Growth Factor; Gene Expression; Genes, fos; Genes, jun; In Vitro Techniques; Mice; Neoplasm Transplantation; Proto-Oncogene Proteins c-jun; RNA, Messenger; Tetradecanoylphorbol Acetate

1992
Growth in serum-free medium of NIH3T3 cells transformed by the EJ-H-ras oncogene: evidence for multiple autocrine growth factors.
    International journal of cancer, 1992, Jul-30, Volume: 51, Issue:6

    Rodent fibroblastic cells transformed by ras oncogenes can grow in serum-free (S-) medium. We have studied clonal lines of mouse NIH3T3 fibroblasts transfected with the EJ-H-ras oncogene, and observed that practically all become independent of exogenous growth and attachment factors shortly after transfection. Moreover, all the clones tested soon form anchorage-independent (AI) colonies in S- medium, and most give rise to spheroids able to grow in suspension. The cell-conditioned S- medium of the transformed (TR) cells stimulates autocrinally the AI and anchored growth of these cells, in the absence of serum, and it contains growth factors related to TGF-alpha (or EGF), PDGF and bFGF, and other uncharacterized factors. Some of these factors are not found, or are found only in very small amounts, in the S- medium of non-transformed NIH3T3 cells, which also stimulates the growth of the TR cells, in the absence of serum. In addition, the TR cells contain 4-6 times more cell-associated bFGF than the non-transformed cells and release more latent TGF-beta activatable by acid treatments. However, no active TGF-beta is secreted by either cell type. Activated TGF-beta and pure TGF-beta 1 stimulate the growth of the anchored TR and NIH3T3 cells, but inhibit the AI growth of the TR cells. Another inhibitor of this growth is also found in the concentrated medium of the NIH3T3 cells.

    Topics: 3T3 Cells; Animals; Cell Division; Cell Line, Transformed; Cell Transformation, Neoplastic; Clone Cells; Culture Media, Serum-Free; Epidermal Growth Factor; Genes, ras; Growth Substances; Humans; Mice; Platelet-Derived Growth Factor; Transfection; Transforming Growth Factor alpha; Urinary Bladder Neoplasms

1992
The effects of continuous exposure to epidermal growth factor on the spontaneous transformation of cultured rat liver epithelial cells.
    The American journal of pathology, 1992, Volume: 140, Issue:1

    A long-term continuous exposure to epidermal growth factor (EGF) enhanced the tumorigenicity of spontaneously transformed cells arising in a clonal population of normal cultured rat liver epithelial cells propagated in a selective growth condition. Lengthy EGF exposure also induced the expression of several phenotypes that differed from the phenotypes of rat liver epithelial cells transformed spontaneously in the absence of EGF. Epidermal growth factor treatment caused consistently an enhancement of the constitutive mRNA expression of transforming growth factor-alpha (TGF-alpha), but not of the EGF receptor and transforming growth factor-beta. The overexpression of TGF-alpha persisted in cell lines derived from tumors formed by the EGF-treated transformed cells. These tumors also exhibited high metastatic incidence and ductal cell differentiation. In contrast, untreated spontaneously transformed cells formed non-metastatic tumors with hepatocellular differentiation. These results suggest that long-term, continuous exposure to EGF/TGF-alpha may modulate the phenotypic expressions of neoplastic transformation.

    Topics: Animals; Blotting, Northern; Cell Line, Transformed; Cell Transformation, Neoplastic; Cells, Cultured; Epidermal Growth Factor; Epithelial Cells; Epithelium; ErbB Receptors; Gene Expression Regulation, Neoplastic; Liver; Liver Neoplasms; Neoplasm Metastasis; Phenotype; Proto-Oncogene Proteins c-myc; Proto-Oncogene Proteins p21(ras); Rats; Rats, Inbred F344; RNA, Messenger; Transforming Growth Factor alpha; Transforming Growth Factor beta

1992
Highly purified human alpha-thrombin promotes morphological transformation of BALB/c 3T3 cells.
    Carcinogenesis, 1992, Volume: 13, Issue:1

    Purified human alpha-thrombin stimulates phosphoinositide turnover as a necessary step in its stimulation of fibroblast cell proliferation. Since phosphoinositide turnover releases diacylglycerol, which activates protein kinase C, we postulated that long-term exposure to thrombin might promote cellular transformation in a manner similar to long-term exposure to phorbol esters, which also activate protein kinase C. The present studies show that chronic exposure of BALB/c 3T3 cells (subclone A31-1-13) to thrombin (5 micrograms/ml) led to a 4- to 20-fold increase in the frequency of morphological transformation over controls as determined by induced foci in monolayer cultures. The foci appeared to represent true transformants as cells from randomly selected foci grew in soft agar and had saturation densities 2- to 3-fold higher than control cells. Acute thrombin treatment for 24 h resulted in small but statistically significant (P less than 0.05) increases in morphological transformation with or without promotion by phorbol myristate acetate, indicating that thrombin can act as a weak initiator or complete carcinogen in this test system. Initiation of cells with low levels of 3-methylcholanthrene followed by promotion with thrombin caused a greater enhancement of morphological transformation (P less than 0.005). Thus, it appears that most of the stimulation of in vitro cell transformation by thrombin may be due to its promotional activity. These results raise the possibility that thrombin released locally following tissue injury or chronic irritation may play a role in cellular transformation and tumor development, especially in tissues sensitized by exposure to initiating carcinogens.

    Topics: 3T3 Cells; Animals; Cell Transformation, Neoplastic; Epidermal Growth Factor; Mice; Tetradecanoylphorbol Acetate; Thrombin

1992
EGF-R antisense RNA blocks expression of the epidermal growth factor receptor and suppresses the transforming phenotype of a human carcinoma cell line.
    The Journal of biological chemistry, 1992, Feb-05, Volume: 267, Issue:4

    We have used an antisense approach to investigate the role of overexpression of the normal human epidermal growth factor (EGF) receptor in the transformed phenotype of KB cells, which are a tumor derived human cell line. Initial experiments performed in vitro, showed that antisense RNA complementary to the entire coding region (AS-FL) or to parts of the EGF-R mRNA (AS-3', AS-5', and AS-K) effectively blocked translation of EGF-R mRNA. In addition, upon microinjection into KB cells, the in vitro synthesized antisense RNAs were able to inhibit transiently the synthesis of EGF-R. Inhibition was concentration-dependent, both in vitro and in cells, and the most effective constructs were those complementary to the entire coding region (AS-FL) or to the 3'-coding end of the mRNA (AS-3'). Transfection of the same EGF-R antisense RNA constructs into the human epidermoid carcinoma KB cell line gave rise to several clones stably expressing elevated levels of antisense RNA and resulting in low residual levels of EGF receptor. The most reduced clones exhibited a totally restored serum-dependent growth and were severely impaired in colony formation and growth in agar. In addition the severity of the phenotype was directly proportional to the residual amount of EGF-R expressed. We conclude that over-expression of normal EGF-R plays a direct primary role in the development of the transformed phenotype of this human cancer cell line.

    Topics: Carcinoma, Squamous Cell; Cell Transformation, Neoplastic; Culture Media, Serum-Free; Epidermal Growth Factor; ErbB Receptors; Gene Expression; Humans; Microscopy, Fluorescence; Phenotype; Plasmids; Precipitin Tests; Protein Biosynthesis; RNA, Antisense; RNA, Messenger; Transcription, Genetic; Transfection; Tumor Cells, Cultured

1992
Susceptibility to ras oncogene transformation is coregulated with signal transduction through growth factor receptors.
    Oncogene, 1991, Volume: 6, Issue:5

    The human teratocarcinoma cell line PA-1 was derived from culturing ascites fluid cells from a patient with an ovarian germ line tumor. We previously described a non-neoplastic variant cloned from the PA-1 human teratocarcinoma cell line, clone 6, which at passage 40 was resistant to transformation by activated ras oncogenes. However, these cells could be transformed by a plasmid containing both myc and ras. Another PA-1 cell variant, clone 1, isolated at passage 63 and used 50 passages later becomes tumorigenic in nude mice after transfection with an activated ras oncogene (Tainsky et al., Anticancer Res., 8, 899-914, 1988). We report here that the progression from ras resistance to ras susceptibility occurs in both clone 1 and clone 6 cells during 25 passages in culture. In the presence of epidermal growth factor, transforming growth factor-alpha, and basic fibroblast growth factor, the ras-transformable cells exhibit anchorage independent growth, whereas the ras-resistant cells can not be growth stimulated by these growth factors. Similarly, ornithine decarboxylase (ODC) activity was inducible in ras susceptible and ras transformed cells by these growth factors, but not in the ras resistant cells. These differences are not due to the level and activity of epidermal growth factor receptor or to the level of expression of 25 proto-oncogenes.

    Topics: Blotting, Northern; Cell Line; Cell Transformation, Neoplastic; Clone Cells; Enzyme Induction; Epidermal Growth Factor; ErbB Receptors; Fibroblast Growth Factor 2; Genes, ras; Humans; Immunoblotting; Oncogenes; Ornithine Decarboxylase; Plasmids; Protein-Tyrosine Kinases; Proto-Oncogene Proteins p21(ras); Receptors, Cell Surface; Receptors, Fibroblast Growth Factor; Signal Transduction; Teratoma

1991
Loss of p53 protein in human papillomavirus type 16 E6-immortalized human mammary epithelial cells.
    Journal of virology, 1991, Volume: 65, Issue:12

    We have shown previously that introduction of the human papillomavirus type 16 (HPV16) or HPV18 genome into human mammary epithelial cells induces their immortalization. These immortalized cells have reduced growth factor requirements. We report here that transfection with a single HPV16 gene E6 is sufficient to immortalize these cells and reduce their growth factor requirements. The RB protein is normal in these cells, but the p53 protein is sharply reduced, as shown by immunoprecipitation with anti-p53 antibody (pAB 421). We infer that the E6 protein reduces the p53 protein perhaps by signalling its destruction by the ubiquitin system. The HPV-transforming gene E7 was unable to immortalize human mammary epithelial cells. Thus, cell-specific factors may determine which viral oncogene plays a major role in oncogenesis.

    Topics: Blotting, Western; Breast; Cell Division; Cell Line; Cell Line, Transformed; Cell Transformation, Neoplastic; Epidermal Growth Factor; Epithelial Cells; Epithelium; Female; Humans; Hydrocortisone; Insulin; Oncogene Proteins, Viral; Papillomaviridae; Phosphorylation; Repressor Proteins; Retinoblastoma Protein; RNA, Messenger; Transfection; Tumor Suppressor Protein p53

1991
Lack of effects of viral sialoadenitis and depletion of epidermal growth factor on initiation of hepatic carcinogenesis in the rat.
    Toxicologic pathology, 1991, Volume: 19, Issue:2

    Sialodacryoadenitis (SDA) is a commonly-encountered coronaviral infection in laboratory rats that causes acute destruction of submandibular salivary glands. SDA results in depletion of salivary epidermal growth factor (EGF) and may thereby affect EGF-dependent cell growth processes. The purpose of this study was to determine the effects of SDA virus (SDAV) infection on the growth factor-dependent stages of experimental liver carcinogenesis. Rats were injected ip with the carcinogen diethylnitrosamine (DENA) at 1, 2, or 3 weeks following inoculation with SDAV. Uninfected control rats were treated only with DENA. The salivary glands of SDAV-inoculated and control rats were stained using the immunoperoxidase method for the detection of EGF. Residual submandibular salivary gland lesions and focal depletion of EGF were still evident in affected submandibular glands for up to 42 days after SDAV infection. Serum EGF concentrations measured at 9, 28, and 42 days following SDAV inoculation were reduced, but were not significantly different in comparison with non-inoculated, DENA-treated control rats. Initiated hepatocytes were detected 21 days after DENA treatment in formalin-fixed sections by an immunoperoxidase stain for the P isoenzyme of the enzyme glutathione S-transferase (GST-P). There was no significant difference in the number of foci of GST-P positive cells in a comparison of initiated cells in SDAV-inoculated and non-inoculated rats. Based on this model, concurrent infection with SDAV does not appear to have any significant effects on the initial stages of chemical hepatocarcinogenesis in the rat.

    Topics: Animals; Antibodies, Viral; Cell Transformation, Neoplastic; Coronaviridae; Coronaviridae Infections; Diethylnitrosamine; Epidermal Growth Factor; Glutathione Transferase; Immunohistochemistry; Incidence; Liver; Liver Neoplasms, Experimental; Male; Rats; Rats, Inbred F344; Salivary Glands; Sialadenitis

1991
Phosphorylation and activation of epidermal growth factor receptors in cells transformed by the src oncogene.
    Molecular and cellular biology, 1991, Volume: 11, Issue:1

    Because functionally significant substrates for the tyrosyl protein kinase activity of pp60v-src are likely to include membrane-associated proteins involved in normal growth control, we have tested the hypothesis that pp60v-src could phosphorylate and alter the signaling activity of transmembrane growth factor receptors. We have found that the epidermal growth factor (EGF) receptor becomes constitutively phosphorylated on tyrosine in cells transformed by the src oncogene and in addition displays elevated levels of phosphoserine and phosphothreonine. High-performance liquid chromatography phosphopeptide mapping revealed two predominant sites of tyrosine phosphorylation, both of which differed from the major sites of receptor autophosphorylation; thus, the src-induced phosphorylation is unlikely to occur via an autocrine mechanism. To determine whether pp60v-src altered the signaling activity of the EGF receptor, we analyzed the tyrosine phosphorylation of phospholipase C-gamma, since phosphorylation of this enzyme occurs in response to activation of the EGF receptor but not in response to pp60v-src alone. We found that in cells coexpressing pp60v-src and the EGF receptor, phospholipase C-gamma was constitutively phosphorylated, a result we interpret as indicating that the signaling activity of the EGF receptor was altered in the src-transformed cells. These findings suggest that pp60v-src-induced alterations in phosphorylation and function of growth regulatory receptors could play an important role in generating the phenotypic changes associated with malignant transformation.

    Topics: Animals; Blotting, Western; Cell Line; Cell Transformation, Neoplastic; Epidermal Growth Factor; ErbB Receptors; Humans; In Vitro Techniques; Oncogene Protein pp60(v-src); Peptide Mapping; Phosphorylation; Phosphotyrosine; Protein-Tyrosine Kinases; Rats; Signal Transduction; Transfection; Type C Phospholipases; Tyrosine

1991
Transformation by pp60src or stimulation of cells with epidermal growth factor induces the stable association of tyrosine-phosphorylated cellular proteins with GTPase-activating protein.
    Molecular and cellular biology, 1991, Volume: 11, Issue:2

    GTPase-activating protein (GAP) is a cytosolic protein that stimulates the rate of hydrolysis of GTP (GTP to GDP) bound to normal p21ras, but does not catalyze the hydrolysis of GTP bound to oncogenic, activated forms of the ras protein. Transformation of cells with v-src or activated transforming variants of c-src or stimulation of cells with epidermal growth factor resulted in the stable association of GAP with two tyrosine-phosphorylated cellular proteins of 64 kDa (p64) and 190 kDa (p190). Analysis of GAP immune complexes isolated from extracts of metabolically labeled src-transformed cells and epidermal growth factor-stimulated cells indicated that tyrosine phosphorylation of p64 and p190 appeared to be coincident with the stable association of these proteins with GAP. Quantitation of the amount of p64 associated with GAP in v-src-transformed cells, however, indicated that only 15 to 25% of tyrosine-phosphorylated p64 was found in complex with GAP. Mutations within the SH2 region of pp60src that render activated pp60src defective for transformation inhibited the efficient formation of complexes between GAP and the tyrosine-phosphorylated forms of p64 and p190. From these data, we suggest that tyrosine phosphorylation and stable association of p64 with GAP is an important step in mediating cellular signaling through the p21ras-GAP pathway.

    Topics: Animals; Avian Sarcoma Viruses; Cell Line; Cell Transformation, Neoplastic; Chick Embryo; Electrophoresis, Gel, Two-Dimensional; Epidermal Growth Factor; Genes, src; GTPase-Activating Proteins; Mutagenesis, Site-Directed; Peptide Mapping; Phosphopeptides; Phosphorylation; Proteins; Proto-Oncogene Proteins pp60(c-src); ras GTPase-Activating Proteins; Rats; Transfection; Tyrosine

1991
Characterization of trans-immortalized hepatic cell lines established from transgenic mice.
    Experimental cell research, 1991, Volume: 195, Issue:1

    Hepato-specific regulatory (promoter/enhancer) DNA sequences were used for targeting the expression of onc genes, such as murine c-myc and Simian Virus 40 T Antigen, to hepatocytes of transgenic mice which subsequently developed hepatocellular carcinomas after a variable period of time (depending on the type of onc gene employed). Several trans-immortalized cell lines were established and compared with respect to the expression of adult hepatic markers and response to growth factors. Despite the morphological differences observed between trans-hepatomas, owing to the expression of the two different onc genes, all tumor-derived cell lines behaved in a comparable fashion during long-term culture displaying an adult hepatic phenotype for at least 40 passages. They differed, however, in response to epidermal growth factor. When the gene coding for human alpha 1-antitrypsin was placed under the control of the same hepato-specific promoter/enhancer, high levels of the human recombinant protein could be harvested from the supernatants of trans-hepatoma-derived cell lines.

    Topics: Acute-Phase Proteins; Albumins; alpha 1-Antitrypsin; alpha-Fetoproteins; Animals; Antigens, Polyomavirus Transforming; Cell Differentiation; Cell Division; Cell Line; Cell Transformation, Neoplastic; Epidermal Growth Factor; Gene Expression; Liver; Liver Neoplasms, Experimental; Mice; Mice, Transgenic; Microscopy, Electron; Promoter Regions, Genetic; Proto-Oncogene Proteins c-myc; RNA, Messenger

1991
[Involvement of cellular responses to growth factors in angiogenesis and transformation malignant].
    Tanpakushitsu kakusan koso. Protein, nucleic acid, enzyme, 1991, Volume: 36, Issue:7

    Topics: Animals; Cell Division; Cell Transformation, Neoplastic; Endothelium, Vascular; Epidermal Growth Factor; Fibroblast Growth Factors; Humans; Neoplasms; Neovascularization, Pathologic; Signal Transduction; Transforming Growth Factor beta

1991
Altered response to growth factors or retinoic acid in phenotypic transformation of normal rat kidney cells expressing human c-fos gene.
    Biochemical and biophysical research communications, 1991, Dec-31, Volume: 181, Issue:3

    Anchorage-independent growth of normal rat kidney (NRK) fibroblast in soft agar depends on both transforming growth factor beta (TGF beta) and epidermal growth factor (EGF). To examine whether c-fos protein is involved in phenotypic transformation of NRK cells, we have transfected and isolated several NRK cell lines that carry the human c-fos gene fused to the metallothionein IIA promoter. A transfectant, Nf-1, had constitutive levels of the human c-fos expression. Anchorage-independent growth of Nf-1 was already stimulated by EGF alone, and the colony sizes of Nf-1 were comparable to those of the parental NRK in the presence of both EGF and TGF beta. Anchorage-independent growth of NRK could be observed in the presence of TGF beta or retinoic acid or platelet derived growth factor (PDGF) and EGF. No growth of NRK in soft agar appeared when basic fibroblast growth factor (bFGF) and EGF were present. By contrast, anchorage-independent growth of Nf-1 was surprisingly enhanced by EGF and TGF beta or retinoic acid or PDGF or bFGF. Expression of the human c-fos gene may compensate the signal to phenotypic transformation induced by TGF beta as well as retinoic acid or PDGF or bFGF.

    Topics: Animals; Blotting, Northern; Blotting, Southern; Blotting, Western; Cell Line; Cell Transformation, Neoplastic; Epidermal Growth Factor; Fibroblast Growth Factor 2; Genes, fos; Growth Substances; Humans; Kidney; Nuclear Proteins; Phenotype; Plasmids; Platelet-Derived Growth Factor; Rats; Recombinant Proteins; Transfection; Transforming Growth Factor beta; Tretinoin

1991
Immediate early gene responses of NIH 3T3 fibroblasts and NMuMG epithelial cells to TGF beta-1.
    Growth factors (Chur, Switzerland), 1991, Volume: 5, Issue:4

    Transforming growth factor beta has a wide range of physiological effects on cell growth and metabolism. We have previously reported on the rapid induction of jun transcription factors in TGF beta-treated cells. Here we show that the early genomic response to TGF beta-1 includes activation of a broad spectrum of serum-inducible genes both in NIH 3T3 fibroblasts and in NMuMG epithelial cells, which are growth-stimulated and growth-inhibited by TGF beta, respectively. Of particular interest is the presence of a putative nuclear DNA-binding receptor (N10) and zinc finger transcription factors (Krox 20 and Krox 24) among the TGF beta-induced genes. In addition to the stimulatory effects of TGF beta, expression of a few genes including c-myc is decreased in both types of cells. In cells transformed by neu or ras oncogenes the immediate early mRNA responses to TGF beta are deregulated. Our results suggest that certain transcription factors are required for both positive and negative regulation of cell proliferation by TGF beta, and that their relative concentrations may determine the subsequent cellular responses.

    Topics: 3T3 Cells; Animals; Blotting, Northern; Cell Division; Cell Transformation, Neoplastic; Cells, Cultured; Epidermal Growth Factor; Epithelial Cells; Epithelium; Gene Expression Regulation; Mice; Oncogenes; RNA, Messenger; Signal Transduction; Transforming Growth Factor beta

1991
Okadaic acid inhibits PDGF-induced proliferation and decreases PDGF receptor number in C3H/10T1/2 mouse fibroblasts.
    Carcinogenesis, 1991, Volume: 12, Issue:4

    Okadaic acid is both a potent inhibitor of protein serine/threonine phosphatases and a tumor promoter in the mouse skin model. We have previously shown that at non-toxic nanomolar concentrations okadaic acid reversibly inhibits induction (promotion) by PDGF of transformed cells by the 'complete' and 'two-stage' protocols in the C3H/10T1/2 mouse fibroblast transformation assay. In the present study we have demonstrated that treatment of confluent and proliferatively quiescent C3H/10T1/2 mouse fibroblasts with low doses of okadaic acid inhibits the platelet-derived growth factor (PDGF)-induced mitogenic response. This inhibition is accompanied by a loss of PDGF binding sites, a decreased PDGF-induced phosphatidylinositol turnover and a decrease in the PDGF-induced intracellular calcium signal. The decrease in the PDGF-generated intracellular signalling processes represents a mechanism by which okadaic acid inhibits PDGF-induced proliferation and the promotion of in vitro neoplastic transformation by PDGF.

    Topics: Animals; Calcium; Cell Division; Cell Transformation, Neoplastic; DNA; Epidermal Growth Factor; Ethers, Cyclic; Fibroblasts; Inositol Phosphates; Intracellular Fluid; Iodine Radioisotopes; Mice; Okadaic Acid; Platelet-Derived Growth Factor; Receptors, Cell Surface; Receptors, Platelet-Derived Growth Factor; Vasoconstrictor Agents

1991
PDGF, CSF-1, and EGF induce tyrosine phosphorylation of p120, a pp60src transformation-associated substrate.
    Oncogene, 1991, Volume: 6, Issue:4

    Transformation by activated pp60c-src has been correlated by genetic analysis with the tyrosine phosphorylation of a 120 kilodalton (kDa) protein, p120. We now demonstrate tyrosine phosphorylation of p120 following stimulation of cells by growth factors whose receptors have intrinsic tyrosine-specific protein kinase activity. Stimulation of quiescent NIH3T3 cells with platelet-derived growth factor (PDGF) resulted in the tyrosine phosphorylation of p120 that was maximal by 5 min and returned to background levels by 30 min. p120 was also phosphorylated on tyrosine after addition of colony-stimulating factor 1 (CSF-1) or epidermal growth factor (EGF) to NIH3T3 cells engineered to express high levels of their respective receptors. Two additional src substrates, p110 and p85, were analysed under identical assay conditions. PDGF, CSF-1, and EGF induced only a minimal increase in the tyrosine phosphorylation of p85 and no change in the phosphorylation of p110. Thus, the marked ligand-induced tyrosine phosphorylation of p120 was a property not shared by the other src substrates examined. Immunoblotting with antibodies to p120 and the ras GTPase activating protein, GAP, suggests that p120 and GAP are unrelated. In addition, the amino acid sequences of four cyanogen bromide peptides derived from p120 showed no homology to GAP or to sequences in either the PIR or Swiss-Prot databases. These data suggest that tyrosine phosphorylation of p120 may contribute to both signal transduction through growth factor receptors and pp60src induced transformation.

    Topics: Amino Acid Sequence; Animals; Avian Sarcoma Viruses; Cell Transformation, Neoplastic; Chromatography, Affinity; Electrophoresis, Polyacrylamide Gel; Epidermal Growth Factor; In Vitro Techniques; Macrophage Colony-Stimulating Factor; Mice; Molecular Sequence Data; Oncogene Protein pp60(v-src); Phosphorylation; Platelet-Derived Growth Factor; Tyrosine

1991
N-myc oncogene enhances mitogenic responsiveness of diploid human fibroblasts to growth factors but fails to immortalize.
    Oncogene, 1991, Volume: 6, Issue:7

    The N-myc gene is amplified in several types of human tumors. To assess the role of the N-myc gene in the transformation of normal human cells, we transfected an N-myc expression vector into diploid human fibroblasts. Transfected clones were isolated and found to express the N-myc gene at levels similar to those seen in a tumor cell line (neuroblastoma LA-N-1) which contains an amplified N-myc gene. The level of N-myc expression decreased as the N-myc clones senesced. Clones expressing N-myc had an increased saturation density and an altered morphology but did not have an extended lifespan. Under low serum conditions, neither the clones expressing N-myc nor the control cells showed anchorage-dependent growth. Clones expressing N-myc were compared to control cells to determine if different growth factors would affect the ability of cells to grow in soft agarose. Clones expressing N-myc and the control cells did not grow in soft agarose supplemented with epidermal growth factor (EGF) or transforming growth factor-alpha (TGF-alpha). However, compared to control cells, clones expressing N-myc grew in agarose 2.8- to 18-fold higher in response to basic fibroblast growth factor (bFGF) and 5.5- to 55-fold higher in response to platelet-derived growth factor B-chain homodimer (PDGF-BB).

    Topics: Animals; Cell Division; Cell Line; Cell Survival; Cell Transformation, Neoplastic; Diploidy; Epidermal Growth Factor; Fibroblast Growth Factor 2; Fibroblasts; Gene Expression; Genes, myc; Growth Substances; Humans; Mice; Mice, Nude; Neoplasm Transplantation; Neoplasms, Experimental; Platelet-Derived Growth Factor; Proto-Oncogene Proteins; Proto-Oncogene Proteins c-sis; Transfection; Transforming Growth Factor alpha

1991
Elevation of glucose transporter, c-myc, and transin RNA levels by Ha-rasT24 is independent of its effect on the cell cycle.
    Molecular carcinogenesis, 1991, Volume: 4, Issue:4

    Elevation of the steady-state mRNA levels of glucose transporter and c-myc are among the earliest changes in gene expression observed after Ha-rasT24 stimulation of Rat-1 fibroblasts to enter the cell cycle. Since the expression of these genes may be the result of either increased cell proliferation or a specific response to rasT24, we evaluated the expression of glucose transporter and c-myc and their induction during the cell cycle in both parental Rat-1 cells and cell lines bearing a metallothionein rasT24 fusion gene (MTrasT24). We showed that, although levels of glucose transporter and c-myc mRNAs in Rat-1 cells underwent a transient increase within hours of the addition of serum, epidermal growth factor, or 12-O-tetradecanoylphorbol-13-acetate to quiescent (G0) cells, the levels of glucose transporter and c-myc mRNA otherwise remained constant throughout the normal cell cycle. In cells carrying MTrasT24 (MR5 cells), induction of rasT24 expression by ZnSO4 led to a rapid induction of glucose transporter and c-myc mRNA expression in both quiescent (density-arrested) and G1/S-synchronized (aphidicolin-blocked) cells. These increases exceeded the constitutive levels expressed in rapidly proliferating Rat-1 cells, indicating that the ras oncogene has an effect on these genes that is independent of growth status. In addition, the transin gene, which is not expressed in proliferating Rat-1 cells in the continuous presence of serum growth factors, was also induced after increased expression of the mutant ras gene. These results suggest that the induction of glucose transporter, c-myc, and transin is the direct result of rasT24-mediated alterations in cellular gene expression and is distinct from normal cell-cycle events.

    Topics: Animals; Cell Cycle; Cell Line; Cell Transformation, Neoplastic; DNA Replication; Epidermal Growth Factor; Fibroblasts; G1 Phase; Gene Expression; Genes, myc; Genes, ras; Kinetics; Matrix Metalloproteinase 3; Metalloendopeptidases; Monosaccharide Transport Proteins; Neoplasm Proteins; Rats; RNA, Messenger; S Phase; Tetradecanoylphorbol Acetate; Transcription, Genetic; Transfection

1991
A comparison of the growth promoting properties of ascitic fluids, cyst fluids and peritoneal fluids from patients with ovarian tumours.
    British journal of cancer, 1991, Volume: 63, Issue:1

    The growth promoting properties of ascitic fluids, cyst fluids and peritoneal fluids from patients with ovarian malignancy, benign ovarian tumours and non-tumour related gynaecological conditions have been investigated using an ovarian carcinoma cell line (OAW 42), mesothelial cells (58MC) and rat kidney cells (NRK-49F). Colony stimulating activity (CSA) for tumour cells and transforming activity (TA) for mesothelial cells were weakly correlated, but whereas elevated TA was tumour-associated, CSA was not. However, TA was not cancer-associated and, although the difference between the mean TA values of benign and malignant cyst fluids was of borderline significance, some benign cyst fluids from cystadenomas showed high TA values. Higher levels of TA in the cystadenomas showed a significant correlation with the menopausal status of the patient and higher levels of TA in the malignant cyst fluid/peritoneal fluid groups were associated with more advanced disease. Results indicated that some fluids contained TGF-beta-like activity, but there was no direct evidence for the presence of TGF-alpha/EGF-like activity in the fluids. Heparin inhibited clonogenic growth of tumour cells but not mesothelial cells. The reduced CSA which was observed after treatment of fluids with both heparin and thrombin implicated coagulation factors in the manifestation of CSA. It was concluded that CSA in the fluids was due, at least partly, to fibrin coagulation, and TA was due to unknown growth factor(s) which may include TGF-beta-like activity. The results are discussed in the context of the aetiology of ovarian carcinoma, and the possible clinical significance of TA.

    Topics: Animals; Ascitic Fluid; Blood Coagulation Factors; Body Fluids; Cell Division; Cell Transformation, Neoplastic; Colony-Stimulating Factors; Cysts; Epidermal Growth Factor; Female; Growth Substances; Heparin; Humans; Mice; Ovarian Neoplasms; Thrombin; Transforming Growth Factor beta; Tumor Cells, Cultured

1991
Liver tumor promotion: effect of phenobarbital on EGF and protein kinase C signal transduction and transforming growth factor-beta 1 expression.
    Digestive diseases and sciences, 1991, Volume: 36, Issue:5

    Phenobarbital (PB) added to the medium of cultured rat hepatocytes alters epidermal growth factor (EGF) dependent mitogenesis in a biphasic manner; PB concentrations less than 1.5 mM are growth stimulatory but higher concentrations significantly inhibit normal hepatocyte proliferation. In contrast, the growth of putative preneoplastic cells is inhibited less by high concentrations of PB. Mechanistic studies designed to test the ability of PB to alter the early events of EGF signal transduction demonstrate that PB neither competes with EGF for binding to the EGF receptor nor alters EGF-induced receptor down-regulation. However, pretreatment with PB (greater than 1 mM) results in a transient inhibition of EGF binding to hepatocytes. The kinetics of this effect are similar to those obtained when hepatocytes are exposed to the phorbol ester 12-O-tetradecanoylphorbol-13-acetate (TPA), a skin tumor promoter and activator of Ca2+/phospholipid-dependent protein kinase C. However, several observations suggest that distinct mechanisms mediate the responses to these two tumor promoters. First, the inhibitory effects of PB and TPA on EGF binding are additive. Also down-regulation of EGF receptors in response to TPA occurs with hepatocytes, A431 epidermal carcinoma cells, HepG2 hepatoma cells, and rat liver epithelial cells, but only hepatocytes are sensitive to PB. Furthermore, translocation of protein kinase C to the membrane occurs in hepatocytes treated with TPA but not in those treated with PB. The chronic treatment of rats with PB further sensitizes hepatocytes to EGF receptor down-regulation by in vitro PB while desensitizing them to EGF receptor down-regulation by TPA. This latter effect is correlated with a decreased ability of TPA to induce translocation of protein kinase C to the membrane. PB significantly increases the intracellular concentration of TGF-beta 1 in periportal hepatocytes but not in putative preneoplastic cells. TGF-beta 1 may therefore have an important function in regulating early stages of cell cycle progression in proliferating hepatocytes.

    Topics: Animals; Binding, Competitive; Cell Transformation, Neoplastic; Diethylnitrosamine; Epidermal Growth Factor; Liver; Liver Neoplasms, Experimental; Phenobarbital; Protein Kinase C; Rats; Signal Transduction; Transforming Growth Factor beta; Tumor Cells, Cultured

1991
Phenotypic expression in spontaneously transformed cultured rat liver epithelial cells.
    Cancer research, 1990, Mar-15, Volume: 50, Issue:6

    Five clonal cell strains of an early-passage normal rat liver epithelial cell line were transformed spontaneously using the protocol of "selective culture" condition. Twelve cell lines were established from the tumors produced after injecting these transformed cells into 1-day-old syngeneic rats. The phenotypic expressions of these spontaneously transformed tumor cell lines were studied and compared to those of cell lines obtained from tumors produced by rat liver epithelial cells transformed by N-methyl-N'-nitro-N-nitrosoguanidine. Like the chemically induced tumor cells, spontaneously transformed tumor cells exhibited phenotypic heterogeneity in the expression of isoenzymes, proto-oncogenes, growth factors and their receptors, and cellular responses to the effect of growth factors. However, unlike the chemically induced tumor cells, these spontaneously induced tumor cells did not express the "resistant phenotypes" characteristic of chemically induced or promoted tumors. Although all the spontaneously induced tumor cell lines expressed variable amounts of transforming growth factor-alpha mRNA, it was not functionally coordinated with the expression of its receptor, the epidermal growth factor receptor. Thus, spontaneously transformed rat liver epithelial cells demonstrate both similarity and diversity in their phenotypic expression when compared to their chemically induced counterpart. This model of spontaneous transformation of cultured rat liver epithelial cells may be useful for the mechanistic study of non-chemically induced carcinogenesis.

    Topics: Animals; Blotting, Northern; Cell Division; Cell Line; Cell Transformation, Neoplastic; Epidermal Growth Factor; Epithelium; Liver; Liver Neoplasms; Methylnitronitrosoguanidine; Nucleic Acid Hybridization; Phenotype; Rats; RNA; RNA, Neoplasm; Transforming Growth Factors

1990
Differential phosphorylation of myelin-associated glycoprotein isoforms in cell culture.
    Journal of neurochemistry, 1990, Volume: 55, Issue:4

    The alternative splicing of myelin-associated glycoprotein (MAG) mRNA generates two isoforms that harbor distinct potential phosphorylation sites in their cytoplasmic tails. Here we characterize the in vivo phosphorylation of MAG isoforms in NIH 3T3 cells transfected with the cDNAs encoding the two isoforms of MAG. Our results demonstrate that the longer isoform, L-MAG, is phosphorylated constitutively mainly on serine, but also on threonine and tyrosine residues. This phosphorylation is subject to change by 12-O-tetradecanoylphorbol 13-acetate (TPA) and ammonium vanadate, but not by dibutyryl-cyclic AMP. The shorter isoform, S-MAG, is constitutively phosphorylated only on serine residues. While TPA and dibutyryl-cyclic AMP have no detectable effect, ammonium vanadate induces tyrosine and threonine phosphorylation in S-MAG. 32P labeling of v-src-transformed NIH 3T3 cells that express L-MAG also show that L-MAG is likely to be an in vivo substrate for pp60v-src tyrosine kinase activity. These results demonstrate that both MAG isoforms are phosphorylated in a heterologous cell system and that this phosphorylation is subject to pharmacological manipulation.

    Topics: Animals; Bucladesine; Cell Line; Cell Transformation, Neoplastic; Cyclic AMP; Epidermal Growth Factor; Insulin; Insulin-Like Growth Factor I; Mice; Myelin Proteins; Myelin-Associated Glycoprotein; Phosphorylation; Tetradecanoylphorbol Acetate; Transfection

1990
Similar early gene responses to ligand-activated EGFR and neu tyrosine kinases in NIH3T3 cells.
    Oncogene, 1990, Volume: 5, Issue:4

    We have compared the responses of serum-inducible immediate early genes to ligand activation of the epidermal growth factor receptor (EGFR) or a hybrid EGFR/neu receptor containing the intracellular domain of the neu proto-oncogene. Few differences in mRNA induction kinetics were found, emphasizing the functional similarity of these structurally related tyrosine kinases.

    Topics: Animals; Cell Transformation, Neoplastic; Cells, Cultured; Chimera; Epidermal Growth Factor; ErbB Receptors; Gene Expression; Kinetics; Mice; Protein-Tyrosine Kinases; Proto-Oncogene Proteins; Proto-Oncogenes; Receptor, ErbB-2; RNA, Messenger; Transcription, Genetic

1990
Intermolecular association of the p185neu protein and EGF receptor modulates EGF receptor function.
    Cell, 1990, Jun-29, Volume: 61, Issue:7

    We have used cross-linking reagents on cell lines expressing both p185neu and EGFR. The lysates of the cells were precipitated with anti-p185neu or anti-EGFR antibodies. These precipitates included a high molecular weight complex that was identified as an EGFR-p185neu heterodimer. Heterodimerization was found to be induced by exposure to EGR. The EGFR of these cells displayed three affinity states for EGF: low (Kd, approximately 10(-9) M), high (Kd, 10(-9) to 10(-10) M), and very high (Kd, 10(-11) M), as determined by Scatchard analyses. Relatively small levels of EGF had a dramatic biological effect on cells expressing very high affinity EGFR. The very high affinity EGFR disappeared after the cells were treated with anti-p185neu monoclonal antibodies that selectively down-regulated p185neu. EGF and TPA had differential effects on down-modulation of the EGFR in cells that express either one or both species of receptor proteins.

    Topics: Animals; Cell Division; Cell Line; Cell Transformation, Neoplastic; Cross-Linking Reagents; DNA Replication; Down-Regulation; Epidermal Growth Factor; ErbB Receptors; Fibroblasts; Gene Expression; Kinetics; Macromolecular Substances; Mice; Molecular Weight; Protein-Tyrosine Kinases; Proto-Oncogene Proteins; Proto-Oncogenes; Receptor, ErbB-2; Tetradecanoylphorbol Acetate; Thymidine

1990
Neoplastic transformation of mouse mammary epithelial cells by deregulated myc expression.
    Cell regulation, 1990, Volume: 1, Issue:11

    A spontaneously immortalized, nontumorigenic mouse mammary epithelial cell line (MMEC) was transfected with an activated myc construct by electroporation. Constitutive expression of myc in MMEC resulted in anchorage independence in soft agar and tumorigenicity in nude mice. The myc-expressing MMEC showed higher saturation density, faster growth rate, and partial abrogation of serum-derived growth factor(s) requirement compared with parent MMEC. Epidermal growth factor or transforming growth factor alpha stimulated the anchorage-independent growth, but not the anchorage-dependent growth, of MMEC-myc cells. Type 1 transforming growth factor beta, on the other hand, inhibited both the anchorage-independent and anchorage-dependent growth of MMEC-myc cells. These results demonstrate that deregulated expression of myc results in neoplastic transformation iin mammary epithelial cells. Accompanying the transformation is altered sensitivity to polypeptide growth factors.

    Topics: Animals; Cell Line; Cell Transformation, Neoplastic; Epidermal Growth Factor; Epithelium; Female; Gene Expression Regulation, Neoplastic; Genes, myc; Mammary Glands, Animal; Mice; Mice, Nude; Neoplasm Transplantation; Proto-Oncogene Proteins c-myc; Transfection; Transforming Growth Factor alpha; Transforming Growth Factor beta

1990
A Swiss 3T3 variant cell line resistant to the effects of tumor promoters cannot be transformed by src.
    Molecular and cellular biology, 1990, Volume: 10, Issue:8

    To study the relationship between oncogenesis by v-src and normal cellular signalling pathways, we determined the effects of v-src on 3T3-TNR9 cells, a Swiss 3T3 variant which does not respond mitogenically to tumor promoters such as 12-O-tetradecanoyl-phorbol-13-acetate (TPA). We found that src was unable to transform these variant cells, whether the oncogene was introduced by infection with a murine retrovirus vector or by transfection with plasmid DNA. 3T3-TNR9 cells were not inherently resistant to transformation, since infection with similar recombinant retroviruses containing either v-ras or v-abl did induce transformation. Further analysis of Swiss 3T3 and 3T3-TNR9 cell populations infected with the v-src-containing retrovirus revealed that although the amount of v-src DNA in each was approximately the same, the level of the v-src message and protein and the overall level of phosphotyrosine expressed in the infected variants was much less than in infected parental cells. Cotransfection experiments using separate v-src and neo plasmids revealed a decrease in the number of G418-resistant colonies when transfections of TNR9 cells occurred in the presence of the src-containing plasmid, suggesting a growth inhibitory effect of v-src on 3T3-TNR9 cells, as has also been found for TPA itself. Since v-src cannot transform this variant cell line, which does not respond mitogenically to the protein kinase C agonist TPA, we suggest that src makes use of the protein kinase C pathway as part of its signalling activities.

    Topics: Animals; Cell Transformation, Neoplastic; Cells, Cultured; Deoxyglucose; DNA Probes; DNA Replication; Drug Resistance; Epidermal Growth Factor; Mice; Oncogene Protein pp60(v-src); Oncogenes; Plasmids; Retroviridae; Tetradecanoylphorbol Acetate; Transfection

1990
Transforming growth factor-beta activities in 'in vivo' lines of hormone-dependent and independent mammary adenocarcinomas induced by medroxyprogesterone acetate in BALB/c mice.
    Breast cancer research and treatment, 1990, Volume: 16, Issue:1

    We have determined the presence of transforming growth factor-beta (TGF-beta)-like polypeptides in mammary adenocarcinomas induced by medroxyprogesterone acetate (MPA) in BALB/c mice. In hormone-dependent tumors (HD) from nontreated and MPA-treated mice a high molecular weight (43 kDa) transforming activity was purified by Bio-Gel P-60 chromatography. This TGF was able to confer the neoplastic phenotype on NRK-49F cells without the addition of epidermal growth factor (EGF), though its activity was potentiated by EGF. It did not compete for binding to the EGF receptor, had no mitogenic activity on monolayer cultures of NRK fibroblasts, and was a potent inhibitor of DNA synthesis induced in these cells by EGF and insulin. In HD and hormone-independent tumors (HI) another TGF with a Mr of 13 kDa was isolated. This transforming activity showed the same biological properties as 43 kDa TGF, with the exception that in the absence of EGF it did not stimulate soft agar growth of NRK-49F cells. The synthesis of both factors in 'in vivo' HD tumors seems to be under MPA control, since it is much lower in HD tumors from MPA-treated mice. Further purification of the 13 and 43 kDa TGFs by hydrophobic interaction HPLC demonstrated that each one eluted in a different position, and that their elution profile differed from the TGF-beta from human platelets. The biological activity of the 13 and 43 kDa TGFs was not neutralized by a specific anti-TGF-beta antibody.

    Topics: Adenocarcinoma; Animals; Cell Division; Cell Transformation, Neoplastic; Chromatography, High Pressure Liquid; DNA Replication; Drug Interactions; Epidermal Growth Factor; Fibroblasts; Gene Expression Regulation, Neoplastic; Insulin; Mammary Neoplasms, Experimental; Medroxyprogesterone; Medroxyprogesterone Acetate; Mice; Mice, Inbred BALB C; Neoplasm Proteins; Neoplasms, Hormone-Dependent; Rats; Transforming Growth Factor beta

1990
Accumulation of p21ras.GTP in response to stimulation with epidermal growth factor and oncogene products with tyrosine kinase activity.
    Proceedings of the National Academy of Sciences of the United States of America, 1990, Volume: 87, Issue:20

    The ras gene product (p21) is a GTP-binding protein and has been thought to transduce signals regulating proliferation or differentiation of cells. Like other GTP-binding proteins, p21.GTP is an active conformation, which can transduce the signals downstream, whereas p21.GDP is an inactive one. Recently, we have shown that p21.GTP levels increased in cells treated with fetal bovine serum or platelet-derived growth factor to initiate DNA synthesis. In this paper, we report that epidermal growth factor can also increase the amounts of p21.GTP in the cells. Effects of epidermal growth factor and platelet-derived growth factor are not additive. In contrast, mutant [Val12]p21, which has transforming activity, responded neither to platelet-derived growth factor nor to epidermal growth factor. We also found that the ratio of p21.GTP to p21.GDP increased 3- to 4-fold in transformants carrying activated erbB-2/neu or v-src oncogenes. These results strongly suggest an important role of p21 in transduction of signals for both normal proliferation and malignant transformation through growth factor receptors with tyrosine kinase activity or related oncogene products.

    Topics: Animals; Cell Division; Cell Line; Cell Transformation, Neoplastic; DNA Replication; Epidermal Growth Factor; Guanosine Diphosphate; Guanosine Triphosphate; Mice; Oncogene Proteins; Oncogenes; Platelet-Derived Growth Factor; Protein Binding; Protein-Tyrosine Kinases; Proto-Oncogene Proteins p21(ras)

1990
Tumour progression in experimental oral carcinogenesis is associated with changes in EGF and TGF-beta receptor expression and altered responses to these growth factors.
    Carcinogenesis, 1990, Volume: 11, Issue:6

    This study examined the characteristics of premalignant oral epithelial cell lines derived from non-invasive palatal and lingual mucosa of rats painted with the carcinogen 4-nitroquinoline N-oxide (4NQO) in vivo. In contrast to normal keratinocytes, premalignant epithelial cells had an extended life span, were independent of 3T3 fibroblast support, and expressed variable anchorage independence in gel culture and tumorigenicity in athymic mice. The expression of these functional phenotypes did not correlate with the duration of 4NQO treatment. Keratinocytes from 4NQO-treated tissues predominantly had fewer epidermal growth factor (EGF) receptors than normal controls. The expression of high-affinity EGF receptors paralleled the emergence of the anchorage-independent phenotype and was markedly elevated in tumorigenic cell lines. Cell lines with an extended life span expressed fewer transforming growth factor beta 1 (TGF-beta) receptors than their normal counterparts though the loss of these receptors appeared to be unrelated to either anchorage independence or tumorigenicity. Normal keratinocytes were stimulated and inhibited, in a dose-dependent manner, by EGF and TGF-beta respectively. By contrast, a cell line that was immortal, anchorage dependent and non-tumorigenic showed reduced sensitivity to stimulation by EGF and was inhibited only by high concentrations of TGF-beta. Cells that were immortal, anchorage independent and tumorigenic, however, were refractory to EGF and were inhibited only by high concentrations of TGF-beta. There was no correlation between the expression of EGF or TGF-beta cell surface receptors and the response to ligand binding. The results show that tumour progression in rat oral epithelial cells is associated with a progressive independence of growth factor control. The number and distribution of EGF and TGF-beta receptors may be useful markers in more closely defining the stages of epithelial tumour progression.

    Topics: 4-Nitroquinoline-1-oxide; Animals; Cell Division; Cell Line; Cell Transformation, Neoplastic; Cells, Cultured; Epidermal Growth Factor; ErbB Receptors; Keratinocytes; Kinetics; Male; Mice; Mice, Nude; Mouth Mucosa; Mouth Neoplasms; Palate; Rats; Rats, Inbred Strains; Receptors, Cell Surface; Receptors, Transforming Growth Factor beta; Tongue; Transforming Growth Factors

1990
High frequency of resistance of human squamous carcinoma cells to the anti-proliferative action of transforming growth factor beta.
    Cancer communications, 1990, Volume: 2, Issue:11

    Transforming Growth Factor beta is a potent autocrine inhibitor of the growth of untransformed keratinocytes. We found each of eight human squamous carcinoma cell lines to be refractory to the anti-proliferative action of Transforming Growth Factor beta. Although each of these carcinoma cell lines expressed the 53-65 kDa (type I) and the 280-300 (type III) Transforming Growth Factor beta-receptor proteins, the 73-85 kDa (type II) species was detectable in only one of these cell lines. Furthermore, although Transforming Growth Factor beta-sensitive non-neoplastic mouse keratinocytes expressed type II binding proteins, human keratinocytes did not. Our findings suggest that resistance to the growth-inhibitory actions of Transforming Growth Factor beta is a common feature of human squamous carcinoma cell lines but does not correlate with the expression of cell-surface receptors for this growth factor.

    Topics: Carcinoma, Squamous Cell; Cell Division; Cell Transformation, Neoplastic; DNA; Drug Resistance; Epidermal Growth Factor; Humans; Keratinocytes; Kinetics; Receptors, Cell Surface; Receptors, Transforming Growth Factor beta; Transforming Growth Factor beta; Tumor Cells, Cultured

1990
Stromal influences on transformation of human mammary epithelial cells overexpressing c-myc and SV40T.
    Journal of cellular physiology, 1990, Volume: 145, Issue:2

    The proto-oncogene c-myc and the oncogene SV40T, both of which have been implicated in the process of cellular immortalization in vitro, have been introduced via amphotropic retroviral expression vectors into the human mammary epithelial cell (HMEC) line 184A1N4 (A1N4). Two stable cell lines were established by growth in selective medium and were found to overexpress either c-myc (A1N4-myc) or SV40T antigen (A1N4-T). Neither the A1N4, A1N4-myc, or A1N4-T cells will grow in soft agar or form tumors in nude mice. However, A1N4-T or A1N4-myc cells, but not the parental A1N4 cells, form colonies in soft agar in response to either epidermal growth factor (EGF), transforming growth factor alpha (TGF alpha), or basic fibroblast growth factor (bFGF). Like EGF and TGF alpha, bFGF is moderately mitogenic for the anchorage-dependent growth (ADG) of all three cell lines. Further, co-cultivation of A1N4-T or A1N4-myc cells with primary diploid mammary fibroblasts can also induce the anchorage-independent growth (AIG) and stimulate the ADG of A1N4-T or A1N4-myc. In addition, conditioned medium obtained from these mammary fibroblasts also stimulated the AIG of the A1N4-T and A1N4-myc cells and was found to contain immunoreactive TGF alpha and bioactive FGF. The mammary fibroblasts express specific mRNA transcripts for bFGF and acidic FGF (aFGF). These results suggest that growth factors such as TFG alpha or FGF, which may be derived from the adjacent mammary stroma, might influence in a paracrine manner the phenotypic characteristics of a population of human mammary epithelial cells toward transformation.

    Topics: Agar; Antigens, Viral, Tumor; Breast; Cell Division; Cell Line, Transformed; Cell Membrane; Cell Transformation, Neoplastic; Epidermal Growth Factor; Epithelium; ErbB Receptors; Fibroblast Growth Factor 2; Fibroblast Growth Factors; Fibroblasts; Humans; Proto-Oncogene Mas; Proto-Oncogene Proteins c-myc; Receptors, Cell Surface; Receptors, Fibroblast Growth Factor; Simian virus 40; Transforming Growth Factor alpha

1990
Inhibition of PC-12 cell differentiation by the immediate early gene fra-1.
    Oncogene, 1990, Volume: 5, Issue:12

    The rat pheochromocytoma cell line (PC-12) offers a powerful in vitro model to study the mechanism of growth factor-induced differentiation and proliferation. Within minutes of addition, agents such as nerve growth factor (NGF), epidermal growth factor (EGF), basic fibroblast growth factor (bFGF) and dibutyryl cyclic AMP (db cAMP) rapidly activate cellular immediate early genes such as c-fos, c-jun, jun-B, and egr-1. fra-1, a member of the immediate early gene family, follows a distinctly later time course of induction than c-fos, c-jun, jun-B, and egr-1, suggesting that fra-1 may attenuate the action of genes induced earlier. We demonstrate that constitutive expression of fra-1 in PC-12 cells results in pronounced inhibition of NGF-induced differentiation. Transcriptional activation of c-fos, c-jun, jun-B, and egr-1 by NGF, EGF, and db cAMP was down-regulated to a varying extent whereas NGF-induced ornithine decarboxylase (ODC) was not affected. Expression of jun-D was not affected in PC-12 fra-1 cells. Transfection of fos and egr-1 promoter-chloramphenicol acetyl transferase (CAT) plasmid into these stable fra-1-expressing PC-12 cells revealed that repression of fos and egr-1 was exerted at the promoter level. Thus deregulated fra-1 expression may inhibit PC-12 cell differentiation by altering the patterns of immediate early gene expression.

    Topics: Adrenal Gland Neoplasms; Animals; Bucladesine; Cell Differentiation; Cell Transformation, Neoplastic; Cells, Cultured; DNA-Binding Proteins; Down-Regulation; Early Growth Response Protein 1; Epidermal Growth Factor; Gene Expression Regulation, Neoplastic; Immediate-Early Proteins; Nerve Growth Factors; Pheochromocytoma; Promoter Regions, Genetic; Proto-Oncogene Proteins; Proto-Oncogene Proteins c-fos; Proto-Oncogene Proteins c-jun; Rats; Signal Transduction; Transcription Factors; Transcription, Genetic; Transfection; Tumor Cells, Cultured

1990
Potentiation of oncogenic N-ras-induced neurite outgrowth and ornithine decarboxylase activity by phorbol dibutyrate and protein kinase inhibitor H-8.
    Journal of cellular physiology, 1990, Volume: 143, Issue:1

    A recombinant N-ras oncogene, under the transcriptional control of a corticosteroid-inducible mouse mammary tumor virus (MMTV) promoter, has been stably transfected into a PC12 rat pheochromocytoma subline. This cell line, designated UR61, undergoes N-ras-induced neurite outgrowth and cessation of division when treated with dexamethasone (Guerrero et al.: Biochemical and Biophysical Research Communications 150:1185-1192, 1988). We have employed the UR61 cell line as a model for ras oncogene-induced neuronal differentiation. In UR61 cells, dexamethasone-induced expression of the recombinant N-ras gene resulted in time-dependent expression of ornithine decarboxylase enzyme (ODC) activity. Prompted by recent reports of possible functional (Lacal et al.: Molecular and Cellular Biology 7:4146-4149, 1987; Wolfman and Macara: Nature 325: 359-361, 1987) and direct (Jeng et al.: Biochemical and Biophysical Research Communications 145:782-788, 1987) interactions between oncogene ras-coded p21 and protein kinase C (PK-C; Ca++/phospholipid-dependent protein kinase), we employed the protein kinase inhibitor H-8 (N-[2-(methylamino)ethyl]-5-isoquinoline sulfonamide dihydrochloride) and phorbol 12,13-dibutyrate (PDBu) to investigate this putative interaction in the UR61 cells, where ODC activity and neurite outgrowth were used as indicators of oncogenic N-ras action. Treatment of UR61 cells with PDBu depleted cells of PK-C and failed to promote neurite outgrowth but enhanced N-ras-induced neurite outgrowth and ODC activity. H-8, which suppressed ODC induction by forskolin and phorbol myristate acetate, enhanced both N-ras-induced ODC activity and neurite outgrowth. Inhibition of ODC activity by difluoromethylornithine (DFMO) did not suppress oncogenic ras-induced neurite outgrowth, suggesting that these two ras-triggered events are mechanistically independent. These findings suggest that certain actions of N-ras can occur in cells depleted of PK-C, and thus, the role of PK-C in ras-induced differentiation differs from its role in ras-induced mitogenesis and transformation.

    Topics: Animals; Cell Differentiation; Cell Transformation, Neoplastic; Colforsin; Dexamethasone; Epidermal Growth Factor; Isoquinolines; Nerve Growth Factors; Neurons; Oncogene Protein p21(ras); Ornithine Decarboxylase; Phorbol 12,13-Dibutyrate; Protein Kinase C; Protein Kinase Inhibitors; Rats; Tetradecanoylphorbol Acetate; Tretinoin; Tumor Cells, Cultured

1990
Characterization of a serum-free culture system comparing growth factor requirements of transformed and untransformed cells.
    Experimental cell research, 1990, Volume: 188, Issue:2

    We describe the first completely serum-free model culture system for comparing growth control in transformed and untransformed cells. Continuous maintenance of untransformed AKR-2B fibroblasts and chemically transformed AKR-MCA cells in the presence of serum-free medium containing epidermal growth factor (E), insulin (I), and transferrin (T) resulted in cell lines which proliferated with similar doubling times (14 h), comparable to parental lines maintained in 10% serum (16 h). The transformed MCA-SF cells and untransformed AKR-SF cells did not differ in their saturation densities in medium containing E + I + T. However, the monolayer proliferation of MCA-SF cells was significantly greater than that of the AKR-SF cells in the presence of E + T, I + T, or T alone. Both cell lines required T to proliferate in monolayer culture. [3H]-Thymidine incorporation experiments and autoradiographic analysis indicated that quiescent MCA-SF cells could reenter the cell cycle by addition of nutrients alone. The combination of E + I + T produced no additional stimulation of DNA synthesis. In contrast, individual polypeptide growth factors (E, I, IGF-I, PDGF, FGF a or b, or TGF-beta 1) were required to elicit a mitogenic response in the untransformed AKR-SF cells. Peak mitogenesis occurred from 18-20 h for all growth factors except TGF-beta 1 (32 h). Neither AKR-SF nor MCA-SF cells could grow with anchorage independence in serum-free medium, unless both TGF-beta 1 and FGF a or b were simultaneously present. The results indicate that this well-defined, serum-free model system can be utilized to detect growth factor-related alterations associated with the transformed state.

    Topics: Animals; Blood; Cell Division; Cell Line; Cell Line, Transformed; Cell Transformation, Neoplastic; Culture Media; DNA; Epidermal Growth Factor; Fibroblast Growth Factors; Growth Substances; Insulin; Insulin-Like Growth Factor I; Platelet-Derived Growth Factor; Transferrin; Transforming Growth Factors

1990
Epidermal growth factor receptor, but not c-erbB-2, activation prevents lactogenic hormone induction of the beta-casein gene in mouse mammary epithelial cells.
    Molecular and cellular biology, 1990, Volume: 10, Issue:8

    The HC11 cell line was isolated from mammary gland cells of pregnant mice. The cells displayed a normal phenotype and retained some characteristics of mammary epithelial cell differentiation. After treatment with the lactogenic hormones prolactin and glucocorticoids, the HC11 cells expressed the milk protein beta-casein. Various oncogenes were transfected and expressed in HC11 cells. The oncogenes were tested for their transformation ability and for their effects upon the differentiation of the HC11 cells. All of the oncogenes tested, including activated human Ha-ras, human transforming growth factor-alpha, activated rat neuT, and human c-erbB-2 activated by a point mutation in the transmembrane domain, caused transformation of the HC11 cells, as shown by tumor formation in nude mice. HC11 cells expressing the neuT and activated c-erbB-2 genes synthesized beta-casein in response to lactogenic hormones, whereas those expressing the Ha-ras or transforming growth factor-alpha oncogenes were no longer able to respond to the lactogenic hormones. This inhibition of beta-casein production occurs at the transcriptional level and in the transforming growth factor-alpha-transformed cells is due to an autocrine mechanism involving the activation of the epidermal growth factor receptor. This suggests that, although the c-erbB-2 and epidermal growth factor receptors are structurally quite similar, their activation has different effects upon mammary epithelial cell differentiation.

    Topics: Animals; Antibodies, Monoclonal; Caseins; Cell Line; Cell Transformation, Neoplastic; Clone Cells; Dexamethasone; Epidermal Growth Factor; Epithelium; ErbB Receptors; Female; Gene Expression Regulation; Genes; Insulin; Mammary Glands, Animal; Mice; Plasmids; Pregnancy; Prolactin; Protein-Tyrosine Kinases; Proto-Oncogene Proteins; Proto-Oncogenes; Rats; Transfection; Transforming Growth Factors

1990
Vanadium compounds promote the induction of morphological transformation of hamster embryo cells with no effect on gap junctional cell communication.
    Cell biology and toxicology, 1990, Volume: 6, Issue:3

    Vanadium compounds were found to promote the induction of morphological transformation of hamster embryo cells. Exposure of the cells to Na-O-vanadate, vanadin (V) oxide or vanadin (IV) oxide sulfate following pre-exposure to a low concentration of benzo[a]pyrene, potentiated the induction of transformed colonies similar to 12-O-tetradecanoylphorbol-13-acetate. Unlike this phorbol ester, vanadium compounds did not inhibit intercellular communication, or active protein kinase C. Nor did vanadate influence the reoccurrence of communication after removal of a communication blocking phorbol ester. On the other hand, vanadate showed strong synergism with the phorbol ester on induction of transformed morphology in the phorbol ester sensitive cell line BPNi. This suggests that vanadium and tumor promoting phorbol esters mediate their effect on the induction of morphological transformation of hamster embryo cells through different mechanisms.

    Topics: Animals; Cell Communication; Cell Transformation, Neoplastic; Cells, Cultured; Cricetinae; Embryo, Mammalian; Enzyme Activation; Epidermal Growth Factor; Intercellular Junctions; Mesocricetus; Protein Kinase C; Vanadium

1990
Characterization of epidermal growth factor receptor induction by retinoic acid in a chemically transformed rat liver cell line.
    Cell growth & differentiation : the molecular biology journal of the American Association for Cancer Research, 1990, Volume: 1, Issue:9

    Levels of epidermal growth factor (EGF) receptor expression vary widely among cell lines derived clonally from a chemically transformed population of rat liver epithelial cells. Retinoic acid (RA), a derivative of vitamin A that stimulates differentiation in a number of embryonal cell lines, increases the level of 125I-EGF binding in several clones of the transformed cell lines. One such cell line, GP6ac, which reverts to a less transformed phenotype when treated with RA, exhibited a 3-4-fold increase in surface EGF receptors with prolonged (2-5-day) RA exposure. The increase persisted as long as the cells were treated with RA. The increase in surface EGF receptors was due to induction of receptor biosynthesis, which occurred within 4 h at both the mRNA and protein levels and persisted until the RA was withdrawn. Paradoxically, the RA response was accompanied by an initial 40-50% decrease in 125I-EGF binding during the first 12 h of RA treatment. The decrease was due primarily to a reduction of receptor affinity. Since the phorbol ester 12-O-tetradecanoylphorbol-13-acetate also decreases 125I-EGF binding and increases EGF receptor biosynthesis in GP6ac cells, we tested the effect of RA in cells depleted of protein kinase C by prolonged treatment (18 h) with 10 microM 12-O-tetradecanoylphorbol-13-acetate. The absence of protein kinase C did not affect the induction of receptor mRNA and protein or the decrease in binding during the early period of RA exposure. This indicates that RA induction of EGF receptor synthesis in GP6ac cells involves signaling pathways distinct from those utilized by phorbol esters.(ABSTRACT TRUNCATED AT 250 WORDS)

    Topics: Animals; Blotting, Northern; Cell Line; Cell Transformation, Neoplastic; Cycloheximide; Epidermal Growth Factor; Epithelial Cells; ErbB Receptors; Gene Expression; In Vitro Techniques; Methylnitronitrosoguanidine; Precipitin Tests; Rats; RNA, Messenger; Tetradecanoylphorbol Acetate; Time Factors; Tretinoin

1990
Transformation of NIH3T3 fibroblasts by an expression vector for the human epidermal growth factor precursor.
    Oncogene, 1990, Volume: 5, Issue:8

    Epidermal growth factor (EGF) and transforming growth factor alpha (TGF alpha) bind to a common cell surface receptor that mediates their diverse biological activities. NIH3T3 fibroblasts transfected with either full-length EGF precursor (preproEGF) or proTGF alpha cDNA displayed distinct patterns of growth in culture. PreproEGF induced focal transformation, and transfectants grew in a chemically defined medium (CDM) at low cell density in the absence of added EGF. In contrast, TGF alpha failed to cause focal transformation, and transfectants grew in CDM in the absence of added growth factors only when seeded at high cell density. The 53 amino acid EGF portion of the preproEGF translation product was essential for its effects. These results indicate that constitutive expression of preproEGF is sufficient to establish autocrine growth of NIH3T3 expressing low levels of EGF receptors. At high cell density, where paracrine as well as autocrine effects of these growth factors would be evident, TGF alpha transfectants displayed at least as high or higher levels of EGF receptor (EGFR) tyrosine phosphorylation than preproEGF transfectants. Since quantitative levels of ligand expression did not account for differences in their transforming properties, preproEGF must be more efficient than proTGF alpha in binding and/or activating EGF receptors in an autocrine manner.

    Topics: Cell Transformation, Neoplastic; DNA; Epidermal Growth Factor; ErbB Receptors; Fibroblasts; Humans; Phosphorylation; Protein Precursors; Transfection; Transforming Growth Factors

1990
Mitogenic activity of phorbol esters and insulin-like growth factor 1 in chemically transformed mouse fibroblasts BP-A31: independent effects and differential sensitivity to inhibition by 3-isobutyl-1-methyl xanthine.
    Experimental cell research, 1989, Volume: 182, Issue:1

    Mitogenic effects of agents activating either the protein kinase C (PDGF; phorbol esters) or the insulin-like growth factor 1 (IGF1)-receptor pathway were studied in quiescent chemically transformed mouse fibroblasts (BP-A31), by evaluating the rate of [3H]thymidine incorporation. Each of these pathways alone was found to be sufficient to sustain progression through the entire cell division cycle. The mitogenic activity of phorbol 12-myristate 13-acetate (PMA) but not that of insulin was blocked by staurosporine (an inhibitor of protein kinase C), in support of the notion that protein kinase C activation was required for the PMA-induced cell cycle progression. The mitogenic effects of PMA were potentiated by cycloheximide pretreatment, and they were abolished by 3-isobutyl-1-methyl xanthine (IBMX; a cyclic nucleotide phosphodiesterase inhibitor). PDGF (known to activate the phospholipase C-protein kinase C pathway) also displayed mitogenic activity in the cycloheximide-pretreated BP-A31 cells, and its effects were prevented by IBMX. In contrast, the mitogenic effects of insulin (at concentrations where it activates the IGF1 receptor) or of IGF1 neither were notably influenced by cycloheximide pretreatment nor were inhibited by IBMX (in the presence of IBMX, the onset of S-phase was delayed by several hours). The expression of the c-fos gene was absent at quiescence; its induction by growth factors was not proportional to their mitogenic potency. Thus, c-fos expression was strongly induced by PMA but only weakly by insulin. IBMX was a powerful inducer of c-fos gene expression but caused a decrease in the level of c-myc mRNA.

    Topics: 1-Methyl-3-isobutylxanthine; Animals; Blotting, Northern; Cell Division; Cell Transformation, Neoplastic; Colforsin; Cyclic AMP; Cycloheximide; Epidermal Growth Factor; Gene Expression Regulation; Insulin; Insulin-Like Growth Factor I; Mice; Platelet-Derived Growth Factor; Proto-Oncogene Proteins; Proto-Oncogene Proteins c-fos; Somatomedins; Suramin; Tetradecanoylphorbol Acetate; Theophylline

1989
[Effects of conditioned media of rat pleural mesothelial cells, transformed in vivo or in vitro by asbestos, on the proliferation of normal mesothelial cells. Comparison with epidermal growth factor, EGF].
    Comptes rendus de l'Academie des sciences. Serie III, Sciences de la vie, 1989, Volume: 309, Issue:15

    The effect of conditioned media obtained from in vivo or in vitro asbestos-transformed rat pleural mesothelial cells on growth of normal mesothelial cells was studied. It was found that both conditioned media led to a reduction of cell proliferation as shown by a decrease in the following parameters: cell number, tritiated thymidine incorporation and percentages of S cells. Similar effects were obtained with EGF (Epidermal Growth Factor).

    Topics: Animals; Asbestos; Cell Count; Cell Division; Cell Transformation, Neoplastic; Culture Media; Epidermal Growth Factor; In Vitro Techniques; Pleura; Rats; Thymidine

1989
Inhibition of epidermal growth factor/transforming growth factor-alpha-stimulated cell growth by a synthetic peptide.
    Journal of cellular physiology, 1989, Volume: 141, Issue:2

    Estrogen-stimulated growth of the human mammary adenocarcinoma cell line MCF-7 is significantly inhibited by monoclonal antibodies to the epidermal growth factor (EGF) receptor that act as antagonists of EGF's mitogenic events by competing for high-affinity EGF receptor binding sites. These antibodies likewise inhibit the EGF or transforming growth factor-alpha (TGF-alpha)-stimulated growth of these MCF-7 cells. An analogous pattern of specific EGF or TGF-alpha growth inhibitory activity was obtained using a synthetic peptide analog encompassing the third disulfide loop region of TGF-alpha, but containing additional modifications designed for increased membrane affinity [( Ac-D-hArg(Et)2(31),Gly32,33]HuTGF-alpha(31-43)NH2). The growth factor antagonism by this synthetic peptide was specific in that it inhibited EGF, TGF-alpha, or estrogen-stimulated growth of MCF-7 cells but did not inhibit insulin-like growth factor-1 (IGF-1)-stimulated cell growth. Altogether, these results suggest that a significant portion of the estrogen-stimulated growth of these MCF-7 cells is mediated in an autocrine/paracrine manner by release of EGF or TGF-alpha-like growth factors. The TGF-alpha peptide likewise inhibited EGF- but not fibroblast growth factor (FGF)- or platelet-derived growth factor (PDGF)-stimulated growth of NIH-3T3 cells in completely defined media; but had no effect on growth or DNA synthesis of G0-arrested cells, nor did it effect growth of NR-6 cells, which are nonresponsive to EGF. Although this synthetic peptide did not directly compete with EGF for cell surface receptor binding, it exhibited binding to a cell surface component (followed by internalization), which likewise was not competed by EGF. The peptide did not directly inhibit EGF-stimulated phosphorylation of the EGF receptor, nor did it inhibit phosphorylation of an exogenous substrate, angiotensin II, by activated EGF receptor. The TGF-alpha peptide did, however, affect the structure of laminin as manifested by laminin self-aggregation; this affect on laminin may, in turn, have a modulatory effect on EGF-mediated cell growth.

    Topics: Adenocarcinoma; Antibodies; Breast Neoplasms; Cell Line; Cell Transformation, Neoplastic; Epidermal Growth Factor; ErbB Receptors; Estradiol; Estrogens; Female; Humans; Laminin; Peptides; Receptors, Immunologic; Receptors, Laminin; Transforming Growth Factors; Tumor Cells, Cultured

1989
Native and a synthetic analogue of the malignancy-associated parathyroid hormone-like protein have in vitro transforming growth factor-like properties.
    The Journal of clinical investigation, 1989, Volume: 83, Issue:3

    A human parathyroid-like protein (PLP) has recently been isolated and cloned from human tumors associated with the paraneoplastic syndrome, humoral hypercalcemia of malignancy. PLP shares NH2-terminal amino acid sequence similarity with PTH but has a unique primary structure thereafter. Studies reported to date have indicated that both native and synthetic amino-terminal PLP polypeptides display actions in vivo and in vitro that are similar to those of PTH. We report here that purified native PLP and synthetic 36Tyr(1-36)amide human PLP induce epidermal growth factor-dependent transformation of NRK 49F cells in soft agar. Further, the synthetic peptide induces a significant increase in the biosynthesis of fibronectin by human dermal fibroblasts. (1-34)PTH does not display either of these biological activities. These data indicate that there are qualitative differences between PTH and the recently identified PLP. The latter hormone appears to possess transforming growth factor-like properties that may be relevant to its physiological actions.

    Topics: Cell Transformation, Neoplastic; Cyclic AMP; Epidermal Growth Factor; Fibroblasts; Fibronectins; Humans; Neoplasm Proteins; Parathyroid Hormone; Parathyroid Hormone-Related Protein; Transforming Growth Factors

1989
AP1/jun function is differentially induced in promotion-sensitive and resistant JB6 cells.
    Science (New York, N.Y.), 1989, May-05, Volume: 244, Issue:4904

    Tumor promoters may bring about events that lead to neoplastic transformation by inducing specific promotion-relevant effector genes. Functional activation of the transacting transcription factor AP-1 by the phorbol ester 12-O-tetradecanoylphorbol-13-acetate (TPA) may play an essential role in this process. Clonal genetic variants of mouse epidermal JB6 cells that are genetically susceptible (P+) or resistant (P-) to promotion of transformation by TPA were transfected with 3XTRE-CAT, a construct that has AP-1 cis-enhancer sequences attached to a reporter gene encoding chloramphenicol acetyltransferase (CAT). Transfected JB6 P+, but not P- variants, showed TPA-inducible CAT synthesis. Epidermal growth factor, another transformation promoter in JB6 cells, also caused P+ specific induction of CAT gene expression. These results demonstrate an association between induced AP-1 function and sensitivity to promotion of neoplastic transformation.

    Topics: Animals; Cell Line; Cell Transformation, Neoplastic; Chloramphenicol O-Acetyltransferase; Cloning, Molecular; DNA-Binding Proteins; Epidermal Growth Factor; Epidermis; Gene Expression Regulation; Genetic Variation; Kinetics; Mice; Nucleic Acid Hybridization; Plasmids; Promoter Regions, Genetic; Proto-Oncogene Proteins; Proto-Oncogene Proteins c-jun; Simplexvirus; Tetradecanoylphorbol Acetate; Transcription Factors; Transfection

1989
Mechanisms of pancreatic beta-cell growth and regeneration: studies on rat insulinoma cells.
    Experimental and clinical endocrinology, 1989, Volume: 93, Issue:2-3

    Information about the mechanism of beta-cell growth and regeneration may be obtained by studies of insulinoma cells. In the present study the growth and function of the rat insulinoma cell lines RINm5F and 5AH were evaluated by addition of serum, hormones, and growth factors. It was found that transferrin is the only obligatory factor whereas growth hormone, epidermal growth factor, fibroblast growth factor, and TRH had modulating effects. A heat-labile heparin binding serum factor which stimulated thymidine incorporation but not cell proliferation was demonstrated in human serum. Measurements of insulin mRNA content showed that the insulinoma cells only contained about 2% of that of normal rat beta-cells. These results are discussed in relation to the role of growth factors, oncogenes, and differentiation in the growth and regeneration of beta-cells.

    Topics: Adenoma, Islet Cell; Animals; Cell Division; Cell Line, Transformed; Cell Transformation, Neoplastic; Epidermal Growth Factor; Fibroblast Growth Factors; Growth Hormone; Insulin; Insulinoma; Islets of Langerhans; Pancreas; Pancreatic Neoplasms; Rats; Regeneration; RNA, Messenger; Thymidine; Thyrotropin-Releasing Hormone; Transferrin; Tumor Cells, Cultured

1989
Basic fibroblast-like growth factor is present in the conditioned medium of simian sarcoma virus transformed NRK cells.
    Biochemical and biophysical research communications, 1989, Nov-15, Volume: 164, Issue:3

    The conditioned medium of Simian sarcoma virus (SSV)-transformed NRK cells contains at least two activities that down regulate the epidermal growth factor receptor. To identify these activities, we analyzed the medium for the presence of factors both related to and distinct from the v-sis oncogene product. Fractionation of the conditioned medium from SSV-transformed NRK cells by chromatography on heparin-Sepharose yielded two active fractions capable of inhibiting EGF binding. The first component, which eluted at 0.8 M NaCl, is able to induce autophosphorylation of the platelet-derived growth factor (PDGF) receptor, is a mitogen for Swiss 3T3 cells and corresponds to the PDGF B chain product of the v-sis oncogene. The second component requires 2 M NaCl for elution, is mitogenic for Swiss 3T3 cells and inhibits high affinity EGF binding through a protein kinase C-independent pathway, all properties of basic FGF. These results suggest that the conditioned medium of v-sis-transformed cells contains at least two factors that can act in an autocrine capacity, one derived from v-sis and one corresponding to basic FGF.

    Topics: Animals; Cell Line; Cell Transformation, Neoplastic; Cells, Cultured; Culture Media; DNA Replication; Epidermal Growth Factor; ErbB Receptors; Fibroblast Growth Factors; Kidney; Kinetics; Mice; Phorbol 12,13-Dibutyrate; Phosphorylation; Platelet-Derived Growth Factor; Proto-Oncogenes; Rats; Receptors, Cell Surface; Receptors, Platelet-Derived Growth Factor; Retroviruses, Simian; Sarcoma Virus, Woolly Monkey

1989
Bovine papillomavirus type I induces resistance to Ca+(+)-induced terminal differentiation in murine keratinocytes.
    Cancer communications, 1989, Volume: 1, Issue:2

    Bovine papillomavirus type 1 (BPV-1) was expressed in established mouse BALB/MK epidermal keratinocytes. In each of the transfected cell lines, DNA synthesis was stimulated by epidermal growth factor (EGF) and inhibited by type beta transforming growth factor to an extent similar to that in parental cells. In contrast, the BPV-1-transfectants were resistant to the induction of terminal differentiation by extracellular Ca(+)+. First, BPV transfectants continued to respond to EGF in the presence of Ca(+)+, whereas the growth of BALB/MK cells was arrested. The characteristics of EGF-binding were identical in the two cell lines. Second, the Ca+(+)-switch failed to induce the activation of epidermal transglutaminase in BPV-1-transfectants. Thus, BPV-1 caused mouse keratinocytes to become resistant to Ca+(+)-induced differentiation without otherwise affecting the control of cell growth.

    Topics: Animals; Bovine papillomavirus 1; Calcium; Cell Differentiation; Cell Line; Cell Transformation, Neoplastic; Cells, Cultured; DNA Replication; Epidermal Growth Factor; ErbB Receptors; Keratinocytes; Mice; Mice, Inbred BALB C; Papillomaviridae; Transcription, Genetic; Transfection; Transforming Growth Factors

1989
A chimeric EGF-R-neu proto-oncogene allows EGF to regulate neu tyrosine kinase and cell transformation.
    The EMBO journal, 1989, Volume: 8, Issue:1

    The neu oncogene, characterized by Weinberg and colleagues, is a transforming gene found in ethylnitrosourea-induced rat neuro/glioblastomas; its human proto-oncogene homologue has been termed erbB2 or HER2 because of its close homology with the epidermal growth factor receptor (EGF-R) gene (c-erbB1). Expression of the rat neu oncogene is sufficient for transformation of mouse NIH 3T3 fibroblasts in culture and for the development of mammary carcinomas in transgenic mice, but the neu proto-oncogene has not been associated with cell transformation. We constructed a vector for expression of a chimeric cDNA and hybrid protein consisting of the EGF-R extracellular, transmembrane and protein kinase C-substrate domains linked to the intracellular tyrosine kinase and carboxyl terminal domain of the rat neu cDNA. Upon transfection with the construct, NIH 3T3 cells gave rise to EGF-R antigen-positive cell clones with varying amounts of specific EGF binding. Immunofluorescence and immunoprecipitation using neu- and EGF-receptor specific antibodies demonstrated a correctly oriented and positioned chimeric EGF-R-neu protein of the expected apparent mol. wt on the surface of these cells. EGF or TGF alpha induced tyrosine phosphorylation of the chimeric receptor protein, stimulated DNA synthesis of EGF-R-neu expressing cells and led to a transformed cell morphology and growth in soft agar. In contrast, the neu proto-oncogene did not show kinase activity or transforming properties when expressed at similar levels in NIH 3T3 cells.(ABSTRACT TRUNCATED AT 250 WORDS)

    Topics: Amino Acid Sequence; Animals; Base Sequence; Cell Transformation, Neoplastic; Enzyme Induction; Epidermal Growth Factor; ErbB Receptors; Genes, Synthetic; Genetic Vectors; Mice; Molecular Sequence Data; Phosphorylation; Protein-Tyrosine Kinases; Proto-Oncogene Mas; Proto-Oncogene Proteins; Proto-Oncogenes; Receptor, ErbB-2; Recombinant Fusion Proteins; Recombinant Proteins; Transforming Growth Factors; Tumor Stem Cell Assay

1989
HER2 cytoplasmic domain generates normal mitogenic and transforming signals in a chimeric receptor.
    The EMBO journal, 1989, Volume: 8, Issue:1

    We have investigated the biological function of an unidentified human growth factor, the ligand of the putative HER2 receptor, by characterizing the signalling properties of its receptor. HER2 (or c-erbB-2), the human homolog of the rat neu proto-oncogene, encodes a transmembrane glycoprotein of the tyrosine kinase family that appears to play an important role in human breast carcinoma. Since a potential ligand for HER2 has not yet been identified, it has been difficult to analyze the biochemical properties and biological function of this cell surface protein. For this reason, we replaced the HER2 extracellular domain with the closely related ligand binding domain sequences of the epidermal growth factor (EGF) receptor, and examined the ligand-induced biological signalling potential of this chimeric HER1-2 protein. This HER1-2 receptor is targetted to the cell surface of transfected NIH 3T3 cells, forms high and low affinity binding sites, and generates normal mitogenic and cell transforming signals upon interaction with EGF or TGF alpha. The constitutive activation of wild-type HER2 in transfected NIH 3T3 cells suggests the possibility that these cells synthesize the as yet unidentified HER2 ligand and activate HER2 by an autocrine mechanism.

    Topics: Animals; Binding Sites; Cell Line; Cell Transformation, Neoplastic; Epidermal Growth Factor; ErbB Receptors; Genes, Synthetic; Humans; Mice; Mitosis; Phosphorylation; Protein-Tyrosine Kinases; Proto-Oncogene Mas; Proto-Oncogene Proteins; Receptor, ErbB-2; Recombinant Fusion Proteins; Recombinant Proteins; Transforming Growth Factors

1989
Production of hepatocellular carcinoma by oval cells: cell cycle expression of c-myc and p53 at different stages of oval cell transformation.
    Cancer research, 1989, Mar-15, Volume: 49, Issue:6

    In rats maintained on a carcinogenic diet (choline deficient containing 0.1% ethionine), the levels of c-myc and p53 mRNAs increased by 4 wk after animals were placed on the diet. Cell isolation studies showed that the change in c-myc takes place in oval cells, while p53 increases predominantly in oval cells but also in hepatocytes. To determine whether this increase is a consequence of cell proliferation or is associated with transformation, we have developed an in vitro model of hepatocarcinogenesis using epithelial cells isolated from the livers of rats fed the carcinogenic diet. When maintained in vitro with infrequent subculture, this cell line (LE/6) undergoes spontaneous transformation. Inoculation s.c. of the transformed cells into nude mice yields tumors histologically identified as hepatocellular carcinoma. We have used these cell lines to compare the cell cycle expression of c-myc and p53 mRNAs in untransformed, partially transformed, and tumorigenic LE/6 cells. We find that the expression of both genes is under cell cycle control in untransformed and partially transformed cells. However, complete transformation of this cell line is associated with constitutive expression of myc but not p53 transcripts. On the basis of this work we suggest that constitutive expression of c-myc may be a late event in hepatocarcinogenesis.

    Topics: Animals; Blotting, Southern; Cell Cycle; Cell Transformation, Neoplastic; Epidermal Growth Factor; Gene Expression Regulation; Liver; Liver Neoplasms, Experimental; Male; Mice; Neoplasm Proteins; Neoplasm Transplantation; Phosphoproteins; Proto-Oncogenes; Rats; Rats, Inbred Strains; Tumor Suppressor Protein p53

1989
Glucocorticoid dexamethasone reversibly complements EJ-RAS oncogene to transform mouse embryo BALB-3T3 cells.
    Journal of cellular biochemistry, 1989, Volume: 41, Issue:4

    EJ-A is a Balb-3T3 transfectant cell line that bears a small number of EJ-ras oncogene copies/cell, has low EJ-ras expression, and resembles the parental cell line in displaying a non-transformed phenotype. The glucocorticoid hormone dexamethasone reversibly induces transformation traits in EJ-A cells, namely: 1) morphological transformation; 2) increased growth rate and saturation density; 3) reduced G1 length; and 4) independence of the FGF requirement to initiate DNA synthesis. Western blot analysis revealed that dexamethasone does not increase the p21ras protein intracellular level. beta-IFN, added to the culture medium, does not suppress the dexamethasone-induced growth stimulation and morphological transformation. Therefore, glucocorticoid hormones can complement low EJ-ras expression to transform Balb-3T3 cells, by a mechanism that is likely to be independent of p21ras increase and beta-IFN decrease.

    Topics: Animals; Cell Line, Transformed; Cell Transformation, Neoplastic; Cocarcinogenesis; Dexamethasone; DNA; Dose-Response Relationship, Drug; Epidermal Growth Factor; Fibroblast Growth Factors; Genes, ras; Insulin; Interferon Type I; Mice; Mice, Inbred BALB C; Oncogene Protein p21(ras); Transfection; Transformation, Genetic

1989
Transforming growth factor alpha production and epidermal growth factor receptor expression in normal and oncogene transformed human mammary epithelial cells.
    Molecular endocrinology (Baltimore, Md.), 1989, Volume: 3, Issue:1

    We have characterized the expression of transforming growth factor alpha (TGF alpha) and its receptor, the epidermal growth factor receptor (EGF-R), in normal and malignantly transformed human mammary epithelial cells. Human mammary epithelial cells were derived from a reduction mammoplasty (184), immortalized by benzo-a-pyrene (184A 1N4), and further transformed by the oncogenes simian virus 40 T (SV40 T), v-Ha-ras, and v-mos alone or in combination using retroviral vectors. 184 and 184A 1N4 cells require EGF for anchorage-dependent clonal growth. In mass culture, they secrete TGF alpha at high concentrations and exhibit an attenuated requirement for exogenous EGF/TGF alpha. SV40 T transformed cells have 4-fold increased EGF-R, have acquired the ability to clone in soft agar with EGF/TGF alpha supplementation, but are not tumorigenic. Cells transformed by v-mos or v-Ha-ras are weakly tumorigenic and capable of both anchorage dependent and independent growth in the absence of EGF/TGF alpha. Cells transformed by both SV40 T and v-Ha-ras are highly tumorigenic, are refractory to EGF/TGF alpha, and clone with high efficiency in soft agar. The expression of v-Ha-ras is associated with a loss of the high (but not low) affinity binding component of the EGF-R. Malignant transformation and loss of TGF alpha/EGF responsiveness did not correlate with an increase in TGF alpha production. Thus, TGF alpha production does not appear to be a tumor specific marker for human mammary epithelial cells. Differential growth responses to EGF/TGF alpha, rather than enhanced production of TGF alpha, may determine the transition from normal to malignant human breast epithelium.

    Topics: Animals; Blotting, Southern; Breast Neoplasms; Cell Line, Transformed; Cell Transformation, Neoplastic; Epidermal Growth Factor; Epithelium; ErbB Receptors; Gene Expression Regulation; Humans; Mammary Glands, Animal; Nucleic Acid Hybridization; Oncogenes; RNA, Messenger; Transforming Growth Factors

1989
Analysis of mammalian fibroblast transformation by normal and mutated human EGF receptors.
    Oncogene, 1989, Volume: 4, Issue:3

    Activation of the EGF receptor (c-erbB) tyrosine kinase has been implicated in tumorigenesis, either by overexpression of the normal receptor in the presence of EGF, or through expression of a truncated receptor lacking the EGF binding domain as in the viral oncogene v-erbB. Here, normal and truncated human EGF receptors expressed in Rat1 fibroblasts were analysed for receptor tyrosine kinase activity and several transformation parameters in comparison with polyoma middle T and EJ-ras. Expression of a truncated EGF receptor lacking the extracellular ligand binding domain induced transformation of immortalized rodent fibroblasts and appears to activate the intrinsic tyrosine kinase. The transformed phenotype becomes enhanced by further truncation of the C-terminal domain containing the tyrosine autophosphorylation sites P1 and P2. Over expression of EGF receptors with an intact extracellular region in transfected Rat1 cells shows EGF dependent transformation, which is reduced by C-terminal truncation. Transformation is dependent on the cellular receptor concentration and can be selected as a stable phenotype. We conclude that expression of receptors with a truncated EGF-binding domain alone is sufficient to transform mammalian fibroblasts, in contrast to chick fibroblasts transformed by v-erbB where additional deletion of C-terminal receptor sequences appears to be an absolute requirement.

    Topics: Animals; Cell Division; Cell Line; Cell Transformation, Neoplastic; DNA Mutational Analysis; Epidermal Growth Factor; ErbB Receptors; Fibroblasts; Gene Expression Regulation; Mice; Mutation; Protein-Tyrosine Kinases; Rats

1989
Acidic cellular environments: activation of latent TGF-beta and sensitization of cellular responses to TGF-beta and EGF.
    International journal of cancer, 1989, May-15, Volume: 43, Issue:5

    Transient (about 2 hr) acidification to approx. pH 5.0 of agar-gelled overlayers containing untransformed NRK-49F or KiMSV-transformed NRK-49F cells in the presence of fetal calf serum or crude 49F-cell conditioned medium, as sources of latent TGF-beta, elicited EGF-dependent colony formation of 49F cells and inhibited spontaneous growth of transformed cells. Pure, active TGF-beta (porcine, type I) had the same effects on these respective cell types, suggesting that the above results were due to activation of latent TGF-beta in the transiently acidic cellular environment. Similar acidifications in the absence of a source of latent TGF-beta enhanced the positive growth response of 49F and AKR-2B cells to EGF and active TGF-beta and also the negative growth response of KiMSV-transformed 49F cells to active TGF-beta. These results are compatible with the idea that acidic cellular environments, particularly in tumor tissues, are conducive to activation of latent TGF-beta, perhaps in conjunction with other activating mechanisms, and to an enhanced response to some growth factors. However, the heterogeneity of cell populations within tumoral masses presents an obstacle to a clear understanding of the consequences of such activation.

    Topics: Agar; Animals; Blood; Cell Division; Cell Line; Cell Transformation, Neoplastic; Culture Media; Epidermal Growth Factor; Hydrogen-Ion Concentration; Lactates; Lactic Acid; Mice; Transforming Growth Factors

1989
Transformation of an established mouse mammary epithelial cell line following transfection with a human transforming growth factor alpha cDNA.
    Molecular carcinogenesis, 1989, Volume: 2, Issue:1

    To determine whether the enhanced expression of transforming growth factor alpha (TGF alpha) is sufficient to induce the neoplastic transformation of an immortalized population of mammary epithelial cells, we cotransfected NOG-8 cells, a cloned mouse mammary epithelial cell line, with a simian virus 40-human TGF alpha cDNA expression vector plasmid and a pSV2neo plasmid. After cotransfection, nine G418-resistant NOG-8 colonies were cloned and expanded. All clones were subsequently analyzed for TGF alpha mRNA expression by northern blot analysis, TGF alpha secretion, anchorage-dependent growth in serum-free medium, anchorage-independent growth in soft agar, and tumorigenicity in nude mice. Three TGF alpha-transfected NOG-8 clones expressed high levels of a specific TGF alpha mRNA, secreted elevated levels of TGF alpha into the culture medium (177-595 ng/10(8) cells/48 h), exhibited an enhanced growth rate, grew aggressively as colonies in soft agar, and formed undifferentiated, invasive carcinomas in nude mice. A neutralizing mouse monoclonal antibody generated against the low molecular weight human TGF alpha peptide was able to inhibit colony formation in soft agar by TGF alpha-transfected NOG-8 clones that produced high levels by TGF alpha. This inhibition suggested that TGF alpha acted through an external autocrine loop. NOG-8 cells and NOG-8 cells transfected with a pSV2neo plasmid alone secreted very low levels of TGF alpha, failed to grow as colonies in soft agar and did not form tumors in nude mice. These results demonstrate that overexpression of a human TGF alpha cDNA in immortalized, nontransformed mouse mammary epithelial cells can induce a transformed phenotype in vitro and can facilitate tumor formation in vivo.

    Topics: Animals; Antibodies, Monoclonal; Cell Line; Cell Transformation, Neoplastic; DNA; Epidermal Growth Factor; Epithelium; ErbB Receptors; Gene Amplification; Mammary Glands, Animal; Mammary Neoplasms, Experimental; Mice; Mice, Nude; Transfection

1989
Regulation of transforming growth factor alpha messenger RNA expression in a chemically transformed rat hepatic epithelial cell line by phorbol ester and hormones.
    Cancer research, 1989, Jul-01, Volume: 49, Issue:13

    Transforming growth factor alpha (TGF-alpha) is produced by many transformed cells, but little is known about the regulation of its expression. We examined TGF-alpha mRNA levels in a set of cloned neoplastic cell lines derived by chemical transformation of a normal rat liver epithelial cell. The untransformed parental cell line, WB-344, did not express a detectable level of TGF-alpha mRNA, whereas GP6ac, a transformed line capable of autonomous growth in soft agar, expressed TGF-alpha. When GP6ac cells were treated with agents thought to regulate protein kinase C activity, e.g., the tumor promoter, 12-O-tetradecanoylphorbol-13-acetate (TPA), TGF-alpha mRNA levels increased by 8- to 11-fold. The induction of TGF-alpha mRNA was detectable at 2 h, was maximal at 8-12 h, and declined by 24 h. Angiotensin, bradykinin, epinephrine, and epidermal growth factor also increased TGF-alpha mRNA by 2- to 5-fold. In contrast, parental WB cells neither expressed TGF-alpha mRNA, nor responded to TPA. TPA also increased epidermal growth factor receptor mRNA in GP6ac cells but the effect was less prolonged; maximal levels were seen at 4 h after TPA exposure and returned to control levels by 12 h. TPA increased TGF-alpha mRNA in GP6ac cells, in part, by increasing transcription of the TGF-alpha gene as measured by run-on transcription rates in isolated nuclei. In addition, the induction of TGF-alpha by TPA was blocked by concurrent incubation with agents that inhibit protein synthesis. However, if TPA was present for at least 2 h, subsequent addition of cycloheximide enhanced the effect of TPA. This indicates that the induction of TGF-alpha in GP6ac cells is comprised of at least two phases demarcated by the requirement for protein synthesis. The time course of induction and the sensitivity to inhibition of protein synthesis distinguish the effect of TPA on TGF-alpha mRNA from that of other genes regulated by TPA, e.g., c-myc and c-fos. These data also suggest that chemical transformation of rat liver epithelial cells leads to expression of TGF-alpha mRNA, and that once expressed, TGF-alpha mRNA can be modulated in a protein kinase C-dependent manner.

    Topics: Animals; Cell Transformation, Neoplastic; Cycloheximide; Epidermal Growth Factor; ErbB Receptors; Gene Expression Regulation; Hormones; In Vitro Techniques; Liver; Protein Kinase C; Rats; RNA, Messenger; Tetradecanoylphorbol Acetate; Time Factors; Transcription, Genetic; Transforming Growth Factors

1989
Growth stimulation, altered regulation of epidermal growth factor receptors, and autocrine transformation of spontaneously transformed normal rat kidney cells by transforming growth factor beta.
    Cancer research, 1989, Jul-15, Volume: 49, Issue:14

    The tumorigenic NRK-PT14 cell line requires exogenous epidermal growth factor (EGF), but has lost the requirement for transforming growth factor beta (TGF-beta) for anchorage-independent growth, compared to normal rat kidney (NRK) cells. Development of an optimized serum-free medium for the growth of these cells revealed that NRK-PT14 cells also exhibit a qualitatively altered sensitivity to exogenous type 1 TGF-beta, compared to NRK cells. EGF-induced serum-free monolayer growth of NRK-PT14 cells was stimulated 2-fold by TGF-beta under conditions where growth of NRK cells was inhibited by 67%. TGF-beta only stimulated the growth of NRK-PT14 cells when EGF was present and when EGF was added before TGF-beta. In addition, the stimulation of EGF-induced NRK-PT14 cell growth by TGF-beta was associated with a specific, reversible loss of the high-affinity subpopulation of EGF receptors from the surface of these cells. Treatment of NRK cells with TGF-beta resulted in an increase in this EGF receptor population. Finally, EGF-induced anchorage-independent growth of NRK-PT14 cells was shown to be dependent on secreted TGF-beta, demonstrating an autocrine role for TGF-beta in the transformed phenotype of these cells. Autocrine transformation of NRK-PT14 cells by TGF-beta may result directly from the acquisition of an altered (positive) sensitivity to this growth factor.

    Topics: Animals; Cell Division; Cell Line; Cell Transformation, Neoplastic; Culture Media; Epidermal Growth Factor; ErbB Receptors; Kidney; Kinetics; Rats; Transforming Growth Factors

1989
The effect of 1.25 dihydroxyvitamin D3 on binding and internalization of epidermal growth factor in cultures cells. Studies on BT-20 cells using quantitative electron microscope autoradiography.
    Experimental cell research, 1989, Volume: 184, Issue:2

    The biological effects of 1.25 (OH)2D3 on epidermal growth receptor (EGF-R) and on EGF internalization were examined in human mammary carcinoma BT-20 cells. In this cell line, with known amplification of the epidermal growth factor receptor gene. EGF was not stimulatory for growth. Biological assay and quantitative EM autoradiography combined with iodinated ligand binding to specific receptors demonstrated that the number of binding sites unit of length of plasma membrane was 2.48-fold higher in treated than in control cells. I-EGF was progressively internalized in a time-and temperature-dependent manner after selective association with the membrane-coated pits. No modification of the time course of I-EGF internalization was noted in the control and in the treated cells, but a different distribution of the labeling in the subcellular compartment was observed in treated cells. In 1.25(OH)2D3-treated batches, the grain density remained low in the receptosomes throughout the experiment, whereas it was high and occurred early in the lysosomes. On the other hand, in control cells, the grain density of the receptosomes was high, whereas it occurred late and was relatively low in the lysosomes. These data suggest that 1.25(OH)2D3 is a regulator of EGF-R level in BT-20 cell line, but it cannot affirmed whether this effect is direct or mediated by other parameters.

    Topics: Autoradiography; Breast; Breast Neoplasms; Calcitriol; Carcinoma; Cell Division; Cell Line; Cell Transformation, Neoplastic; Epidermal Growth Factor; ErbB Receptors; Female; Humans; Iodine Radioisotopes; Microscopy, Electron; Organelles; Tumor Cells, Cultured

1989
Nuclear transport in 3T3 fibroblasts: effects of growth factors, transformation, and cell shape.
    The Journal of cell biology, 1988, Volume: 106, Issue:1

    Nucleocytoplasmic transport of fluorescent-labeled macromolecules was investigated in transformed and nontransformed 3T3 fibroblasts. Insulin and epidermal growth factor enhanced transport three-fold after 1-2-h incubation with nontransformed adhering fibroblasts; no enhancement of transport was observed for spherical unattached fibroblasts. The concentration of growth factor for maximal enhancement was 3-10 nM. Nuclear transport for Kirsten murine sarcoma virus-transformed BALB/c 3T3 fibroblasts, however, was maximally enhanced before addition of growth factors; addition of insulin or epidermal growth factor causes no additional transport enhancement. Transformation also minimizes cell shape effects on macromolecular nuclear transport. These results provide evidence that protein growth factors and oncogenic transformation may use a similar mechanism for activation of nuclear transport.

    Topics: Animals; Biological Transport; Cell Line; Cell Nucleus; Cell Transformation, Neoplastic; Cell Transformation, Viral; Cytoplasm; Dextrans; Diffusion; Epidermal Growth Factor; Growth Substances; Insulin; Mice; Nuclear Envelope; Receptors, Cell Surface; Spectrometry, Fluorescence

1988
Transforming growth factor alpha in chemically transformed hamster oral keratinocytes.
    Cancer research, 1988, Jun-01, Volume: 48, Issue:11

    The cheek pouch of the Syrian hamster is an excellent tissue for the experimental induction of oral cancer by carcinogenic chemicals. Lysate prepared from a cell line (HCPC-1) derived from one of these hamster oral tumors greatly increased the growth of these oral tumor cells in vitro. We now show that the mitogenic substance, transforming growth factor alpha (TGF-alpha), is present in all of the chemically transformed hamster oral tumors examined (in vitro and in vivo). In no adult normal tissue of the Syrian hamster can we detect expression of TGF-alpha. TGF-alpha could be partly or wholly responsible for the mitogenic activity detected in the lysate of the chemically transformed hamster oral keratinocytes. Both normal and chemically transformed hamster oral keratinocytes express the receptor to epidermal growth factor. The consistent detection of TGF-alpha and epidermal growth factor receptor mRNAs in these hamster oral tumor cells suggests that an autocrine growth mechanism might be operative. This hamster cheek pouch oral cancer model can be used for the molecular analysis of how TGF-alpha and epidermal growth factor receptor might be involved in the malignant transformation of epithelial tissues.

    Topics: 9,10-Dimethyl-1,2-benzanthracene; Animals; Carcinoma, Squamous Cell; Cell Line; Cell Transformation, Neoplastic; Cloning, Molecular; Cricetinae; DNA Replication; Epidermal Cells; Epidermal Growth Factor; Epidermis; ErbB Receptors; Growth Substances; Keratins; Mouth Neoplasms; Peptides; Transforming Growth Factors

1988
Loss of growth factor dependence and conversion of transforming growth factor-beta 1 inhibition to stimulation in metastatic H-ras-transformed murine fibroblasts.
    Cancer research, 1988, Dec-15, Volume: 48, Issue:24 Pt 1

    Cell lines with varying tumorigenic and metastatic potentials have been obtained by transformation of 10T 1/2 fibroblasts using radiation or transfection with T-24 H-ras. We have observed an inverse relationship between metastatic potential and dependence on serum for growth. The effects of basic fibroblast growth factor, platelet-derived growth factor, epidermal growth factor, and transforming growth factor-beta 1 (TGF-beta 1) on these lines were then examined to determine if the changes in the serum dependence of metastatic cells may be due to altered responsiveness to specific growth factors (GFs). Cells were grown in monolayer culture and DNA synthesis was measured by [CH3-3H]thymidine incorporation experiments. Both metastatic and nonmetastatic cells were shown to be equivalent in their diminished responsiveness to basic fibroblast growth factor, platelet-derived growth factor, and epidermal growth factor as compared to their nontransformed, parental 10T 1/2 cells. However, a unique response of metastatic cells to TGF-beta 1 was identified. While TGF-beta 1 inhibited DNA synthesis in 10T 1/2 cells and a nonmetastatic tumor, cells with intermediate to high metastatic ability were stimulated up to 5.8-fold by TGF-beta 1. Interestingly, epidermal growth factor abrogated the TGF-beta 1 inhibition of the parental 10T 1/2 cells, but had no effect on the TGF-beta 1 response of any metastatic line. Therefore, metastatic but not nonmetastatic cells, demonstrated a dramatically altered sensitivity to TGF-beta 1, a response which may be important in determining metastatic potential.

    Topics: Animals; Cell Line; Cell Transformation, Neoplastic; DNA Replication; Epidermal Growth Factor; Fibroblast Growth Factors; Fibroblasts; Fibrosarcoma; Genes, ras; Growth Substances; Mice; Mice, Inbred BALB C; Neoplasm Metastasis; Platelet-Derived Growth Factor; Transforming Growth Factors

1988
Modified responsiveness of v-Ha-ras-transfected rat fibroblasts to growth factors and a tumor promoter.
    Molecular carcinogenesis, 1988, Volume: 1, Issue:2

    The correlation of the phenotypic changes of v-Ha-ras transfected cells with the expression of p21ras and the modified responses to growth factors and a tumor promoter were examined. Transfection of the v-Ha-ras gene together with the neomycin-resistance gene into 208F rat fibroblasts yielded transformed clones characterized by morphological changes, anchorage-independent growth, and tumorigenicity in nude mice. The degrees of these biological alterations were parallel with the expression of mRNA and protein of the ras gene. In ras-transformed cells, anchorage-independent growth was stimulated by epidermal growth factor (EGF), insulin, bombesin, and fibroblast growth factor, whereas in the parental 208F cells, anchorage-independent growth was observed only in the presence of EGF, and there were many fewer EGF-induced colonies than those in the ras-transformed clones. A tumor promoter, 12-O-tetradecanoylphorbol-13-acetate (TPA) also augmented anchorage-independent growth of ras-transformed cells and induced morphological changes in monolayer cultures without altering the expression of the ras gene or phosphorylation of the p21ras protein. Retinoic acid inhibited the TPA-induced anchorage-independent growth. These results showed a good correlation of the expression of p21ras with the phenotypic changes and the increased sensitivity of the p21ras-expressing cells to the stimulation of growth factors and tumor promoter.

    Topics: Animals; Blotting, Northern; Bombesin; Carcinogenicity Tests; Carcinogens; Cell Adhesion; Cell Transformation, Neoplastic; Cells, Cultured; Epidermal Growth Factor; Fibroblasts; Mice; Mice, Nude; Oncogenes; Phenotype; Proto-Oncogene Proteins; Proto-Oncogene Proteins p21(ras); Rats; Tetradecanoylphorbol Acetate; Transfection

1988
Comparison of mouse pro-1 and pro-2 transfectants for responses to tumor promoters and antipromoters.
    Cancer research, 1988, Nov-01, Volume: 48, Issue:21

    A previous report demonstrated that mouse JB6 cells transformed to promotion sensitive (P+) phenotype by transfection with an activated promotion sensitivity (pro) gene showed both evidence for the presence of the transfected gene and sensitivity to phorbol ester induced transformation similar to that observed in parental P+ cells. In addition, pro-1 and pro-2 transfectants were similar to each other in phorbol ester response. The current report extends these findings to ask whether pro-1 or pro-2 transfectants are also sensitive to promotion of transformation by other classes of tumor promoters such as epidermal growth factor (EGF), lanthanides, and phthalate esters and to inhibition of phorbol ester promoted transformation by several classes of antipromoters. The results showed that both pro-1 and pro-2 transfectants resembled parental P+ cells in sensitivity to promotion of anchorage independent transformation by lanthanides and by diethylhexylphthalate. In addition both pro-1 and pro-2 transfectants showed inhibition of phorbol ester induced transformation by antipromoters ganglioside GT1b, ethylene glycol bis(beta-aminoethyl ether)-N,N,N',N'-tetraacetic acid, and forskolin. Thus the pathways implicated by these inducers and inhibitors of transformation appear similar to those implicated for parental P+ cells and similar when controlled by pro-1 or pro-2. The single differential response was that of EGF-induced transformation. pro-2 transfectants but not pro-1 transfectants were sensitive to EGF-induced neoplastic transformation. The nonresponsiveness could not be attributed to lack of EGF receptors since 125I-EGF binding to pro-1 transfectants was similar to that for pro-2 transfectants and parental P+ cells. Thus pro genes transfer responsiveness to a C-kinase mediated promotion of transformation pathway and to putatively non-C kinase pathways triggered by lanthanides or phthalate esters, but not necessarily to an EGF receptor kinase mediated pathway.

    Topics: Animals; Cell Line; Cell Transformation, Neoplastic; Diethylhexyl Phthalate; Egtazic Acid; Epidermal Growth Factor; Mice; Mice, Inbred BALB C; Protein Kinase C; Tetradecanoylphorbol Acetate; Transfection

1988
Ligand activation of overexpressed epidermal growth factor receptors transforms NIH 3T3 mouse fibroblasts.
    Proceedings of the National Academy of Sciences of the United States of America, 1988, Volume: 85, Issue:5

    The cell surface receptor for the mitogenic peptide epidermal growth factor (EGF) is involved in control of normal cell growth and may play a role in the genesis of human neoplasia such as squamous carcinoma and glioblastoma. Soft-agar growth and focus-formation experiments with NIH 3T3 mouse fibroblasts transfected with an expression plasmid demonstrated the ligand-dependent transforming potential of the human EGF receptor without structural alterations. Activation of overexpressed normal receptor alone appears to be sufficient for transformation of NIH 3T3 cells in vitro.

    Topics: Animals; Cell Line; Cell Transformation, Neoplastic; Epidermal Growth Factor; ErbB Receptors; Gene Amplification; Gene Expression Regulation; Ligands; Mice; Molecular Weight; Peptides; Transforming Growth Factors

1988
Epidermal growth factor enhances N-ethyl-N-nitrosourea-induced morphological transformation of Syrian hamster embryo cells.
    Carcinogenesis, 1988, Volume: 9, Issue:4

    To improve the usefulness of the Syrian hamster embryo (SHE) cell transformation system as a short-term test, it was investigated whether the variation in results due to serum variability could be reduced by the addition of epidermal growth factor (EGF). It was found that EGF-significantly (3-7-fold) enhanced the frequency of morphological transformation induced by N-ethyl-N-nitrosourea or benzo[a]pyrene if added to growth medium supplemented with a batch of serum which had a low ability to support transformation. Furthermore, addition of EGF to the assay medium enabled the demonstration of dose dependence of transformation with relatively small group sizes (up to 2000 colonies). Finally, it was observed that the transformed phenotype was easier to recognize in the presence of EGF. These data suggest that routine addition of EGF to the assay medium might reduce variability and enhance sensitivity of the SHE transformation assay.

    Topics: Animals; Benzo(a)pyrene; Cell Transformation, Neoplastic; Cells, Cultured; Cricetinae; Embryo, Mammalian; Epidermal Growth Factor; Ethylnitrosourea; Mesocricetus

1988
Transforming growth factor beta as a potent promoter in two-stage BALB/c 3T3 cell transformation.
    Cancer research, 1988, May-15, Volume: 48, Issue:10

    We have tested transforming growth factor beta (TGF beta) in the two-stage BALB/c 3T3 cell transformation assay for possible tumor-promoting activity, since it has several effects similar to those of tumor-promoting phorbol esters. After initiation of BALB/c 3T3 cells with 3-methylchol-anthrene, treatment with TGF beta at 1 ng/ml alone or in combination with epidermal growth factor (EGF) for 4 weeks enhanced the number of transformed foci by 5- to 6-fold in comparison with uninitiated cells. Initiation treatment alone induced no or very few transformed foci in several assays. Treatment with phorbol-12,13-didecanoate (PDD) at 100 ng/ml for 4 weeks enhanced the number of transformed foci in initiated BALB/c 3T3 cells by 4- to 5-fold in comparison with uninitiated cells. Thus, TGF beta at 1 ng/ml is as potent as PDD at 100 ng/ml for tumor-promoting activity in the two-stage BALB/c 3T3 cell transformation assay. The enhancing effect of TGF beta was dose-related in the dose range tested (0.03-1 ng/ml) and was not reversible. Some of the foci induced by combined MCA-TGF beta-EGF treatment were cloned, and eight out of nine clones tested produced tumors in nude mice. TGF beta (1 ng/ml) plus EGF (2 ng/ml) increased the saturation density to a similar extent as PDD (100 ng/ml) but did not affect the growth of BALB/c 3T3 cells. We observed no change in junctional intercellular communication, as measured by the dye transfer method, when cells were treated with TGF beta during the two-stage BALB/c 3T3 cell transformation assay. Nevertheless, there was selective communication between transformed and surrounding nontransformed cells; MCA-TGF beta transformed cells intercommunicated among themselves but not with surrounding nontransformed cells. Our results indicate that TGF beta has potent tumor-promoting activity in vitro, but that this activity is not mediated by a complete blockage of intercellular communication, as is suggested for phorbol ester tumor promoters.

    Topics: Animals; Cell Communication; Cell Line; Cell Transformation, Neoplastic; Dose-Response Relationship, Drug; Epidermal Growth Factor; Methylcholanthrene; Mice; Peptides; Phorbol 12,13-Dibutyrate; Phorbol Esters; Transforming Growth Factors

1988
Transforming growth factor beta 1 positively regulates its own expression in normal and transformed cells.
    The Journal of biological chemistry, 1988, Jun-05, Volume: 263, Issue:16

    Transforming growth factor beta 1 (TGF-beta 1) regulates the growth, differentiation, or function of nearly all cell types. We now report that TGF-beta 1 increases steady-state levels of its own message in six different normal and transformed cells in culture. Accumulation of TGF-beta 1 mRNA can be detected by Northern blot analysis within 3 h of addition of the peptide to cells, and enhanced message levels persist as long as TGF-beta 1 is present in the culture medium. This autoinduction is half-maximal at approximately 10 PM TGF-beta 1, and maximal stimulation corresponds to a 2-3-fold increase in transcript levels. In normal rat kidney cells, the rise in TGF-beta 1 mRNA is actinomycin D-sensitive and is accompanied by a parallel (approximately 3-fold) increase in secretion of TGF-beta 1 protein in the culture medium of treated cells, as detected by immunoprecipitation of biosynthetically labeled 35S-labeled TGF-beta 1 using specific anti-TGF-beta 1 antibodies. Treatment of normal rat kidney cells with either epidermal growth factor or platelet-derived growth factor also results in an increase in TGF-beta 1 mRNA (2-3-fold), although epidermal growth factor and TGF-beta 1 appear to act via distinct mechanisms since their combined effects are greater than additive.

    Topics: Cell Line; Cell Transformation, Neoplastic; Collagen; Cycloheximide; Dactinomycin; Epidermal Growth Factor; Gene Expression Regulation; Glyceraldehyde-3-Phosphate Dehydrogenases; Homeostasis; Humans; Peptides; Platelet-Derived Growth Factor; RNA, Messenger; Transforming Growth Factors

1988
Truncation of the human EGF receptor leads to differential transforming potentials in primary avian fibroblasts and erythroblasts.
    The EMBO journal, 1988, Volume: 7, Issue:10

    The transforming capacity of the normal and mutant human EGF receptor (EGFR) was investigated in primary chicken cells. In fibroblasts, both N- and C-terminal truncations resulted in a weak, additive oncogenic activity. However, not even double truncations caused a v-erbB-like phenotype. Upon EGF-binding, on the other hand, both normal and C-terminally truncated EGFRs resembled v-erbB in their fibroblast transforming potential. In erythroblasts, N-terminal truncation was sufficient to induce constitutive self-renewal, which was enhanced by deletion of 32 C-terminal amino acids but abolished by a larger truncation of 202 amino acids. In contrast to the normal EGFR, the receptor lacking 32 C-terminal amino acids resembled v-erbB in conferring erythropoietin independence for spontaneous differentiation to the transformed erythroblasts. Our results indicate that the C-terminal domain of the EGFR is non-essential in fibroblast transformation, but seems to be crucial for both self renewal induction and specificity of receptor function in erythroblasts.

    Topics: Animals; Cell Differentiation; Cell Division; Cell Transformation, Neoplastic; Cells, Cultured; Chickens; DNA Mutational Analysis; Epidermal Growth Factor; ErbB Receptors; Erythroblasts; Erythropoietin; Fibroblasts; Gene Expression Regulation; Humans; Leukemia, Experimental; Mitosis; Oncogene Proteins, Viral; Protein Binding; Structure-Activity Relationship

1988
Platelet-derived growth factor or basic fibroblast growth factor induce anchorage-independent growth of human fibroblasts.
    Journal of cellular physiology, 1988, Volume: 137, Issue:3

    Anchorage-independent growth, i.e., growth in semi-solid medium is considered a marker of cellular transformation of fibroblast cells. Diploid human fibroblasts ordinarily do not exhibit such growth but can grow transiently when medium contains high concentrations of fetal bovine serum. This suggests that some growth factor(s) in serum is responsible for anchorage-independent growth. Much work has been done to characterize the peptide growth factor requirements of various rodent fibroblast cells for anchorage-independent growth; however, the requirements of human fibroblasts are not known. To determine the peptide growth factor requirements of human fibroblasts for anchorage-independent growth, we used medium containing serum that had had its peptide growth factors inactivated. We found that either platelet-derived growth factor (PDGF) or the basic form of fibroblast growth factor (bFGF) induced anchorage-independent growth. Epidermal growth factor (EGF) did not enhance the growth induced by PDGF, or did so only slightly. Transforming growth factor beta (TGF-beta) decreased the growth induced by PDGF. EGF combined with TGF-beta induced colony formation in semi-solid medium at concentrations at which neither growth factor by itself was effective, but the combination was much less effective in stimulating anchorage-independent growth than PDGF or bFGF. This work showed that PDGF, or bFGF, or EGF combined with TGF-beta can stimulate anchorage-independent growth of nontransformed human fibroblasts. The results support the idea that cellular transformation may reduce or eliminate the need for exogenous PDGF or bFGF.

    Topics: Blood; Cell Division; Cell Transformation, Neoplastic; Cells, Cultured; Culture Media; Epidermal Growth Factor; Fibroblast Growth Factors; Fibroblasts; Humans; Platelet-Derived Growth Factor; Transforming Growth Factors

1988
[Growth and functions of mouse submandibular gland epithelial cells in culture and in their chemically-transformed counterparts].
    [Osaka Daigaku shigaku zasshi] The journal of Osaka University Dental Society, 1988, Volume: 33, Issue:1

    Topics: Animals; Cell Transformation, Neoplastic; Cells, Cultured; Epidermal Growth Factor; Epithelial Cells; Epithelium; Male; Mice; Submandibular Gland

1988
Autocrine regulation of cell proliferation by estradiol and hydroxytamoxifen of transformed mouse Leydig cells in serum-free culture.
    Endocrinology, 1988, Volume: 122, Issue:1

    We have previously reported that the cloned cell line (B-1-A-2) derived from an estrogen-responsive mouse Leydig cell tumor shows an estrogen-dependent enhancement of cell proliferation in medium supplemented with charcoal-dextran-stripped fetal bovine serum. To avoid the involvement of unknown factors present in the serum in the pathway for estrogen-dependent cell growth, the present study was designed to establish a serum-free culture system to which growth factors could be added. To this end, we subcloned B-1 cells from the parental tumor cell line. The proliferation of B-1 cells was markedly stimulated by the addition of 10(-11)-10(-8) M estradiol into the serum-free medium [Eagle's Minimum Essential Medium-Ham's F-12 (1:1, vol/vol) containing 0.2% (wt/vol) BSA]. Epidermal growth factor (0.1-50 ng/ml) or insulin (0.1-50 micrograms/ml) alone or in combination with 10(-8) M estradiol did not affect the proliferation rate of B-1 cells. In contrast, a greater than 10-fold molar excess of 4-hydroxytamoxifen blocked estradiol-induced cell proliferation, while 4-hydroxytamoxifen alone failed to show a stimulatory effect on cell multiplication. Additionally, the conditioned medium collected from estradiol-stimulated cells was found to contain a growth-promoting factor(s) whose activity was not antagonized by 4-hydroxytamoxifen. Nonstimulated cells secreted a significant but low level of the growth-promoting factor. Finally, B-1 cells were found to be estrogen dependent for cell proliferation in BALB/c mice. Their growth was markedly inhibited by the administration of tamoxifen to the host mice. These results indicate that the serum-free culture system presented here is suitable for studying the autocrine mechanisms of cell growth regulated by estrogens as well as triphenylethylene compounds.

    Topics: Animals; Cell Division; Cell Line; Cell Transformation, Neoplastic; Cells, Cultured; Culture Media; Epidermal Growth Factor; Estradiol; Estrogen Antagonists; Insulin; Kinetics; Leydig Cell Tumor; Male; Mice; Mice, Inbred BALB C; Receptors, Estrogen; Tamoxifen; Testicular Neoplasms

1988
The role of polypeptide growth factors in phenotypic transformation of normal rat kidney cells.
    The Journal of biological chemistry, 1988, Jan-05, Volume: 263, Issue:1

    A serum-free assay has been established for studying the role of polypeptide growth factors in inducing loss of density-dependent inhibition of growth of normal rat kidney (NRK) cells. The process has been characterized by measuring the time course of [3H]thymidine incorporation into confluent, quiescent NRK cultures stimulated by defined polypeptide growth factors, in combination with cell counting studies, increases in DNA content, and cell cycle analysis by means of a fluorescence-activated cell sorter. It is shown that none of the growth factors tested (epidermal growth factor, platelet-derived growth factor, transforming growth factor-beta, and retinoic acid) is able to induce loss of density-dependent inhibition of growth by itself, but strong synergism was observed when combinations of growth factors were tested. None of the above factors was found to be essential, however, since any combination of three of the above four growth factors strongly induced the process. Strong parallels were observed between the growth factor requirements for inducing loss of density-dependent inhibition of growth under serum-free conditions and the requirements for induction of anchorage-independent proliferation under growth factor-defined assay conditions. This indicates that most likely the same cellular processes underlie these two aspects of phenotypic transformation, although data indicate that anchorage-independent proliferation may be a more restricted property of phenotypic transformation than loss of density dependence of proliferation. It is concluded that phenotypic transformation of NRK cells does not require specific polypeptide growth factors, but reflects the ability of these cells to respond to multiple growth factors.

    Topics: Animals; Cell Division; Cell Line; Cell Transformation, Neoplastic; Epidermal Growth Factor; Growth Substances; Insulin; Kidney; Kinetics; Peptides; Platelet-Derived Growth Factor; Rats; Transforming Growth Factors; Tretinoin

1988
Comparative analysis of the involvement of p53, c-myc and c-fos in epidermal growth factor-mediated signal transduction.
    Experimental cell research, 1987, Volume: 169, Issue:2

    p53, a transformation-related protein located in the nucleus, shares several properties with the product of the nuclear proto-oncogene c-myc. The latter is transiently induced after different membrane-originating stimuli. A similar observation has been made with c-fos, a gene that also belongs to the 'nuclear' class of oncogenes. Here we show that p53, unlike the products of the c-myc and c-fos genes, is not induced by the signal generated by the interaction between epidermal growth factor (EGF) and its receptor. Hence, p53 does not appear to be involved in EGF signal transduction. In order to draw this conclusion we have used an EGF receptor gene-amplified human breast tumor cell line that is growth-inhibited by EGF, and exponentially growing normal human fibroblasts.

    Topics: Cell Line; Cell Transformation, Neoplastic; Epidermal Growth Factor; Humans; Neoplasm Proteins; Nucleoproteins; Phosphoproteins; Poly A; Proto-Oncogene Mas; Proto-Oncogenes; RNA; RNA, Messenger; Transduction, Genetic; Tumor Suppressor Protein p53

1987
Growth in culture and tumorigenicity after transfection with the ras oncogene of liver epithelial cells from carcinogen-treated rats.
    Cancer research, 1987, Aug-01, Volume: 47, Issue:15

    Two epithelial cell lines designated LE/2 and LE/6 were established from cells isolated by centrifugal elutriation from the livers of carcinogen-treated rats. Both cell lines exhibit some characteristics of fetal liver cells, such as the expression of the 2.3-kilobase alpha-fetoprotein mRNA, aldolase A, and lactate dehydrogenases 4 and 5. Primary cultures contain gamma-glutamyl transferase-positive cells which do not proliferate in vitro. After the first passage, the LE/2 and LE/6 cell lines are uniformly gamma-glutamyl transferase negative. Neither cell line is transformed as assayed by morphology, anchorage-independent growth, or tumor formation in nude mice. By the 50th passage, LE/6 cells form numerous colonies in soft agar in the presence of epidermal growth factor, while no colonies grow in medium lacking this growth factor. Clonal cell populations derived from five epidermal growth factor-induced soft agar colonies were not tumorigenic in nude mice. This indicates that, although epidermal growth factor-responsive late passage cells had acquired some of the phenotypic properties commonly associated with tumor cells, these cells were not fully transformed. Transformation of LE/6 cells was accomplished by transfection of the rasH oncogene (EJ). Subcutaneous inoculation of rasH (EJ)-transfected LE/6 cells produced tumors at the site of injection with histological features of moderate to well-differentiated trabecular hepatocellular carcinomas. Tumor cell lines derived from the nude mouse tumors are gamma-glutamyl transferase positive and express alpha-fetoprotein mRNA. One clonal cell line expresses both alpha-fetoprotein and albumin mRNA. These results show that nonparenchymal liver epithelial cells transfected with an activated oncogene can give rise to differentiated hepatocellular tumors similar to those induced in livers of rats fed a carcinogenic diet.

    Topics: alpha-Fetoproteins; Animals; Cell Transformation, Neoplastic; Cells, Cultured; Choline Deficiency; Epidermal Growth Factor; Epithelium; gamma-Glutamyltransferase; Isoenzymes; Liver; Liver Neoplasms, Experimental; Male; Mice; Mice, Nude; Neoplasm Proteins; Oncogenes; Proto-Oncogene Proteins; Proto-Oncogene Proteins p21(ras); Rats; Rats, Inbred Strains; Recombinant Proteins; RNA, Messenger; RNA, Neoplasm; Transfection

1987
Effect of 5-azacytidine on malignant transformation of a mutant derived from the mouse BALB/c 3T3 cell line resistant to transformation by chemical carcinogens.
    Cancer research, 1987, Sep-15, Volume: 47, Issue:18

    BALB/c 3T3 and its derivative MO-5, isolated as a monensin-resistant clone, showed a very low rate of spontaneous malignant transformation. Treatment of BALB/c 3T3 cells with benzo(alpha)pyrene, 4-nitroquinoline-N-oxide, N-methyl-N'-nitro-N-nitrosoguanidine, N-methyl-N-nitrosourea, or UV light irradiation significantly enhanced the rate of transformation, whereas the treatment of MO-5 cells with these carcinogens had only a slight if any effect. Exposure of MO-5 as well as BALB/c 3T3 to 5 or 10 microM 5-azacytidine for 3 to 7 days significantly increased the number of transformation foci. The Luria-Delbrück fluctuation test showed that spontaneous mutation frequency (mutants/cell/generation) was 1.2 X 10(-6) for BALB/c 3T3 and 7.1 X 10(-7) for MO-5, respectively, when appearance of cadmium-resistant clones was tested. N-Methyl-N'-nitro-N-nitrosoguanidine enhanced induced mutation frequency of ouabain-resistant and cadmium-resistant mutants of BALB/3T3 but it only slightly enhanced that of MO-5. Methylation status of DNA of MO-5 was compared with that of BALB/c 3T3 by comparing the cleavage patterns generated by the isoschizomeric restriction enzymes HpaII and MspI. DNA of MO-5 was found to be more methylated than that of BALB/c 3T3 in the vicinity of c-myc as well as the metallothionein-I gene. Aberrant DNA methylation in MO-5 and the cellular sensitivity to transformation by chemical carcinogens or 5-azacytidine are discussed.

    Topics: 4-Nitroquinoline-1-oxide; Animals; Azacitidine; Benzo(a)pyrene; Cell Line; Cell Transformation, Neoplastic; DNA; Epidermal Growth Factor; Metallothionein; Methylation; Methylnitronitrosoguanidine; Mice; Mice, Inbred BALB C; Mutation; Proto-Oncogenes; Tetradecanoylphorbol Acetate

1987
Evidence that the viral Ki-ras protein, but not the pp60v-src protein of ASV, stimulates proliferation through the PDGF receptor.
    Biochemical and biophysical research communications, 1987, Oct-14, Volume: 148, Issue:1

    Protamine sulfate (PS), a specific blocker of PDGF action, inhibited the proliferative response of tsKSV-NRK cells to a reactivated, temperature-sensitive viral Ki-RAS protein, but it did not affect the proliferative response of tsASV-NRK cells to a reactivated pp60v-src protein kinase. The inhibition by PS of the proliferation response of tsKSV-NRK cells to reactivated Ki-RAS protein was overcome by serum growth factors, notably EGF, and concentrated serum-free conditioned medium from cultured NRK cells infected with wild-type KSV, but not by a combination of PDGF and insulin. These observations suggest that the viral Ki-RAS protein, but not pp60v-src, stimulates proliferation exclusively by inducing the host cells to produce PDGF or PDGF-like mitogenic factors.

    Topics: Animals; Cell Division; Cell Transformation, Neoplastic; Epidermal Growth Factor; Kinetics; Kirsten murine sarcoma virus; Oncogene Protein pp60(v-src); Platelet-Derived Growth Factor; Protamines; Protein Kinases; Proto-Oncogene Proteins; Proto-Oncogene Proteins p21(ras); Receptors, Cell Surface; Receptors, Platelet-Derived Growth Factor; Retroviridae Proteins

1987
Construction of a novel oncogene based on synthetic sequences encoding epidermal growth factor.
    Science (New York, N.Y.), 1987, Jan-16, Volume: 235, Issue:4786

    The autocrine model postulates that constitutive release of a mitogenic growth factor can lead to uncontrolled proliferation and cell transformation. A synthetic polynucleotide encoding epidermal growth factor conferred a tumorigenic phenotype on cells. These cells were transformed through the action of an autocrine circuit having an extracellular component.

    Topics: Animals; Antibodies, Monoclonal; Cell Division; Cell Transformation, Neoplastic; DNA, Recombinant; Epidermal Growth Factor; Gene Expression Regulation; Mice; Neoplasms, Experimental; Oncogenes; Rats; Transfection

1987
Growth state-dependent regulation of protein kinase C in normal and transformed murine cells.
    Cancer research, 1987, Feb-15, Volume: 47, Issue:4

    We determined whether growth state can influence the action of protein kinase C by measuring protein kinase C activity in growing and stationary cultures of normal and transformed cells. Two approaches were used to measure protein kinase C: assay of intact cells for inhibition of epidermal growth factor (EGF) binding in response to phorbol dibutyrate (PDBu); and assay of detergent extracts for total calcium, phospholipid-dependent kinase activity. In extracts of growing and stationary Swiss 3T3 cells, the total amount of protein kinase C activity was similar, indicating that growth state does not alter the level of enzyme in the cell. The short-term response of Swiss 3T3 cells to an activator of protein kinase C also appeared to be independent of growth state, since the 50% effective dose for PDBu inhibition of EGF binding to its receptor was approximately 7 nM for both growth conditions. In contrast, the response of cells to long-term treatment with PDBu was significantly different depending upon the initial growth state of the cells. In both growth states, PDBu caused loss of protein kinase C activity, which reflected a loss in protein mass as determined by immunoblotting with antiserum to protein kinase C. However, the maximum decrease approached 100% in stationary cultures versus approximately 75% in growing cells. Protein kinase C levels in several transformed cell lines were subject to down modulation in a similar growth state-dependent manner. Further, the inhibition of EGF binding by tumor promoters following long-term treatment of Swiss 3T3 cells with PDBu also varied with growth state. In down modulated growing cells, PDBu caused almost complete inhibition of EGF binding, whereas in down modulated stationary cells, minimal inhibition of EGF binding by PDBu was observed. These results suggest that prolonged treatment with tumor promoters alters the sensitivity of cells to activators of protein kinase C in a growth state-dependent manner.

    Topics: Animals; Cell Line; Cell Transformation, Neoplastic; Dimethyl Sulfoxide; Dose-Response Relationship, Drug; Epidermal Growth Factor; Fibroblasts; Mice; Phorbol 12,13-Dibutyrate; Phorbol Esters; Protein Kinase C

1987
Human colon cancer cell lines secrete alpha TGF-like activity.
    British journal of cancer, 1987, Volume: 55, Issue:1

    Topics: Cell Line; Cell Transformation, Neoplastic; Clone Cells; Colonic Neoplasms; Epidermal Growth Factor; ErbB Receptors; Growth Substances; Humans; Neoplasm Proteins; Peptides; Transforming Growth Factors

1987
Loss of growth responsiveness to epidermal growth factor and enhanced production of alpha-transforming growth factors in ras-transformed mouse mammary epithelial cells.
    Journal of cellular physiology, 1987, Volume: 130, Issue:3

    A mouse mammary epithelial cell line, NMuMG, exhibits a low capacity to grow in semisolid medium as colonies and it is not tumorigenic in nude mice. In contrast, NMuMG cells which have been transformed by an activated c-Harvey ras proto-oncogene, NMuMG/rasH, or by the polyoma middle T-transforming gene, NMuMG/pyt, are able to grow in soft agar and, when injected into nude mice, produce undifferentiated carcinomas. Human epidermal growth factor (EGF) or human alpha-transforming growth factor (alpha TGF) can stimulate, in a dose-dependent fashion, the anchorage-independent growth of NMuMG and NMuMG/pyt cells in soft agar but fail to enhance the anchorage-independent growth of the NMuMGrasH cells. Likewise, human EGF or human alpha TGF is also able to stimulate the anchorage-dependent growth of normal NMuMG cells and NMuMG/pyt cells in a serum-free medium supplemented with insulin, transferrin, fetuin, and laminin, or in medium containing low concentrations of serum, whereas these same growth factors under comparable culture conditions have little or no effect upon the anchorage-dependent growth of the ras-transformed NMuMG-rasH cells. The biological refractoriness of the NMuMG/rasH cells to human EGF or human alpha TGF is reflected by a reduction in the total number of cell surface receptors for EGF and by an absence of a high-affinity population of binding sites for mouse [125l]EGF on these cells as compared to the NMuMG or NMuMG/pyt cells. In addition, concentrated conditioned medium (CM) obtained from NMuMG/rasH and NMuMG/pyt cells contains a relatively higher amount of biologically active TGFs than CM obtained from comparably treated NMuMG cells as measured by the ability to induce the anchorage-independent growth of normal rat kidney cells in soft agar. The higher levels of biologically active TGFs found in the CM from the transformed cells relative to the NMuMG cells is paralleled by a corresponding increase in the CM from these cells in the amount of immunoreactive alpha TGF, by an increase in the amount of EGF receptor-competing activity, and by an increase in the levels of alpha TGF mRNA in the NMuMG/rasH cells. These results demonstrate that mammary epithelial cells which have been transformed by an activated ras proto-oncogene, but not by the polyoma middle T-transforming gene, become unresponsive to exogenous EGF or alpha TGF.(ABSTRACT TRUNCATED AT 400 WORDS)

    Topics: Animals; Cell Division; Cell Line; Cell Transformation, Neoplastic; Epidermal Growth Factor; Epithelial Cells; ErbB Receptors; Female; Growth Substances; Kinetics; Mammary Glands, Animal; Mice; Oncogenes; Peptide Biosynthesis; Peptides; Proto-Oncogene Mas; Recombinant Proteins; Transforming Growth Factors

1987
A chimeric, ligand-binding v-erbB/EGF receptor retains transforming potential.
    Science (New York, N.Y.), 1987, Apr-10, Volume: 236, Issue:4798

    Comparison of amino acid sequences from human epidermal growth factor (EGF) receptor and avian erythroblastosis virus erbB oncogene product suggests that v-erbB represents a truncated avian EGF receptor gene product. Although both proteins are transmembrane tyrosine kinases, the v-erbB protein lacks most of the extracellular ligand-binding domain and a 32-amino acid cytoplasmic sequence present in the human EGF receptor. To test the validity of the proposed origin of v-erbB and to investigate the functional significance of the deleted extracellular sequences, a chimeric gene encoding the extracellular and the transmembrane domain of the human EGF receptor joined to sequences coding for the cytoplasmic domain of the avian erbB oncogene product was constructed. When expressed in Rat1 fibroblasts, this reconstituted gene product (HER-erbB) was transported to the cell surface and bound EGF. Its autophosphorylation activity was stimulated by interaction with the ligand. Expression of the HER-erbB chimera led to anchorage-independent cell growth in soft agar and EGF-induced focus formation in Rat1 monolayers. Thus, it appears that v-erbB protein sequences in the chimeric receptor retain their transforming activity under the influence of the human extracellular EGF-binding domain.

    Topics: Animals; Cell Cycle; Cell Line; Cell Transformation, Neoplastic; DNA, Recombinant; Epidermal Growth Factor; ErbB Receptors; Humans; Oncogenes; Phosphorylation; Protein-Tyrosine Kinases; Rats

1987
Bombesin induces c-fos and c-myc expression in quiescent Swiss 3T3 cells. Comparative study with other mitogens.
    Experimental cell research, 1987, Volume: 170, Issue:1

    We have studied the effect of the potent mitogen bombesin on the expression of c-fos and c-myc genes in quiescent mouse fibroblasts. We have demonstrated that bombesin rapidly induces a transient expression of c-fos mRNA followed by a more protracted elevation in c-myc mRNA levels. The intensity of the induction of expression of both proto-oncogenes depended on the dose of bombesin used. Prolonged treatment of the cells with TPA, which causes a selective decrease in protein kinase C activity, partially inhibited the induction of c-fos and c-myc gene expression by bombesin, similar to what has been observed with PDGF. However, a dramatic inhibition of the mitogenic response to bombesin--but not to PDGF--was found in TPA-treated cells. In contrast, TPA-treated cells showed an increased response to EGF with regard to proto-oncogene expression. The role of protein kinase C and Ca2+-dependent pathways in proto-oncogene induction by bombesin is discussed.

    Topics: Animals; Bombesin; Cell Transformation, Neoplastic; Cells, Cultured; DNA Replication; Epidermal Growth Factor; Mice; Mitogens; Platelet-Derived Growth Factor; Proto-Oncogenes; RNA, Messenger; Tetradecanoylphorbol Acetate; Transcription, Genetic

1987
Lifespan extension of basal cell nevus syndrome fibroblasts by transfection with mouse pro or v-myc genes.
    International journal of cancer, 1987, May-15, Volume: 39, Issue:5

    Dermal fibroblasts from patients with the autosomal dominant cancer-prone disease Basal Cell Nevus Syndrome (BCNS) exhibit a serum dependence, anchorage dependence and in vitro lifespan (about 20 population doublings or less) similar to those of fibroblasts from normal age-, race- and sex-matched controls. Transfection with v-myc or with an activated mouse pro-I gene (which specifies sensitivity to promotion of neoplastic transformation in JB6 mouse epidermal cells) specifically conferred partial immortality on the BCNS fibroblasts by substantially extending their population doubling levels by more than 19 population doublings. This suggests that either v-myc or pro-I gene can cooperate with BCNS gene(s) to produce an extension of lifespan or partial immortality. However, the transfected BCNS fibroblasts that escaped senescence were anchorage-dependent even after exposure to the tumor promoters 12-O-tetradecanoyl-phorbol-13-acetate (TPA), epidermal growth factor (EGF) or platelet-derived growth factor (PDGF). These observations indicate that BCNS fibroblasts differ from their normal counterparts in susceptibility to extended growth and may therefore be pre-neoplastic. It is clear that they require more than an activated pro or myc gene for progression to the tumor cell phenotype.

    Topics: Adult; Basal Cell Nevus Syndrome; Carcinoma, Basal Cell; Cell Division; Cell Survival; Cell Transformation, Neoplastic; Cells, Cultured; DNA; Epidermal Growth Factor; Female; Genetic Engineering; Humans; Male; Phenotype; Tetradecanoylphorbol Acetate

1987
Suppression of the EGF-dependent induction of c-myc proto-oncogene expression by transforming growth factor beta in a human breast carcinoma cell line.
    Biochemical and biophysical research communications, 1987, May-14, Volume: 144, Issue:3

    Alterations in c-myc proto-oncogene expression after treatment of human mammary carcinoma MDA-468 cells with epidermal growth factor (EGF) and/or transforming growth factor beta (TGF beta) have been investigated. A stimulation of c-myc messenger RNA was detected within 60 min after treatment with EGF. This induction persisted for at least 24 hr, albeit to a lower extent. The early and late increase in c-myc mRNA levels induced by EGF were inhibited by the presence of TGF beta. TGF beta alone induced little change in c-myc mRNA levels. The effect of TGF beta represents a novel action of this hormone at the level of gene expression.

    Topics: Breast Neoplasms; Cell Line; Cell Transformation, Neoplastic; Epidermal Growth Factor; Female; Growth Substances; Humans; Kinetics; Peptides; Proto-Oncogene Mas; Proto-Oncogenes; RNA, Messenger; Transcription, Genetic; Transforming Growth Factors

1987
The Ha-ras-induced transformed phenotype of rat-1 cells can be suppressed in hybrids with rat embryonic fibroblasts.
    Journal of cellular biochemistry, 1987, Volume: 34, Issue:1

    Somatic cell hybrids were isolated from fusions of diploid embryonic rat fibroblasts with transformed Rat-1 cells which contained 4 to 5 copies of the transforming human Ha-ras 1 gene. In contrast to their transformed parental cells four hybrid clones showed normal morphology, long latency periods of tumorigenicity in newborn rats, anchorage requirement of proliferation, and an eightfold-reduced amount of secreted transforming growth factor activity. Thus these hybrids are called suppressed with regard to expression of the Ha-ras-induced transformed phenotype. Tumorigenic derivatives of the suppressed hybrids that had segregated chromosomes were isolated. Since two of the tumorigenic hybrid clones showed the similar low level of secreted transforming growth factors as the suppressed hybrids, decreased production of transforming growth factor activity is unlikely to be a sufficient criterion for suppression of malignancy. Whereas one of the suppressed hybrids expressed the transforming gene product p21 at a level similar to that of the transformed parental cells, other suppressed hybrids expressed less p21. This suggests that the suppressed phenotype can be regulated at the posttranslational level of p21 but that additional controls of expression of p21 are likely to exist. DNA of the suppressed hybrids transformed Rat-1 cells to proliferation in the presence of semisolid agar. Thus the activated human Ha-ras gene in the suppressed hybrids retained its biological activity even though it did not transform these cells to tumorigenicity.

    Topics: Animals; Cell Transformation, Neoplastic; DNA; Epidermal Growth Factor; Fibroblasts; Hybrid Cells; Oncogenes; Phenotype; Rats; Transfection

1987
Expression of the c-myc proto-oncogene in growth stimulated porcine thyroid follicle cells.
    Acta endocrinologica. Supplementum, 1987, Volume: 281

    In the present investigation the expression of c-myc in porcine thyroid follicle cells following the incubation with TSH or EGF was studied. EGF did not only have a mitotic effect on thyroid follicles but also increased c-myc mRNA. The increase in c-myc RNA is an early event in EGF stimulated thyroid cells. TSH had no effect on c-myc RNA levels of porcine thyroid cells. EGF-binding and sensitization to the mitotic activity of EGF, however, is increased by TSH. While TSH stimulates cAMP production and functional activity but not growth or c-myc expression of porcine thyroid cells, EGF produces cell division accompanied with c-myc expression.

    Topics: Animals; Cell Division; Cell Transformation, Neoplastic; Cells, Cultured; Epidermal Growth Factor; Mitosis; Proto-Oncogenes; Swine; Thyroid Gland; Thyrotropin

1987
Effects of epidermal growth factor on transferrin receptor phosphorylation and surface expression in malignant epithelial cells.
    Journal of cellular physiology, 1987, Volume: 132, Issue:3

    The transferrin (Tf) receptor is a major transmembrane protein which provides iron for normal and malignant cell growth. Epidermal growth factor (EGF) has been reported to rapidly and transiently alter the number of surface Tf receptors in normal and transformed epithelial cells. To investigate mechanisms of EGF-induced changes in surface Tf display, EGF effects on surface Tf receptors were compared in two cell lines which differ in their number of EGF receptors and growth responses to EGF. In cloned A431 cells with high receptor numbers which are growth-inhibited by EGF, EGF caused a 50% decrease in Tf receptor expression after 30 min. In contrast, EGF induced a rapid, transitory increase (within 5 min) in the number of surface Tf receptors on KB carcinoma cells which returned to basal levels by 15 min. The observed changes in Tf receptor display were due to altered receptor distribution and not changes in ligand affinity or total cellular transferrin receptor pools. Anti-EGF receptor monoclonal antibody blocked effects of EGF on transferrin receptor expression. Since the antibody is internalized and causes EGF receptor down-regulation, effects on transferrin receptor expression were independent of these events. EGF-induced alterations in Tf receptor display occurred even when cells were pretreated with colchicine, suggesting that changes in surface Tf binding were not mediated by cytoskeletal components. Na orthovanadate, which mimics some early cellular effects of EGF, duplicated EGF's effects on A431 Tf receptors, but had no effect on KB cells, suggesting these responses occur by differing mechanisms. To determine whether EGF caused changes in Tf receptor phosphorylation, 32P-labelled Tf receptors were immunoprecipitated after EGF treatment. After exposure to EGF, A431 cells showed no change in Tf phosphorylation, but KB cells showed a transient, 6-fold increase in transferrin receptor phosphorylation on serine residues. In both A431 and KB cells, phorbol ester (PMA) also increased phosphorylation on transferrin receptors, but had little effect on surface Tf receptor expression. In malignant cell lines, EGE induces rapid, variable changes in transferrin receptor expression and phosphorylation which differ from the effects of PMA. These early responses to EGF appear to differ with the cell type and correlate poorly with alterations in Tf receptor phosphorylation. These results suggest Tf receptor phosphorylation does not regulate Tf receptor display in

    Topics: Animals; Antibodies; Binding Sites; Carcinoma, Squamous Cell; Cell Line; Cell Transformation, Neoplastic; Epidermal Growth Factor; Epithelium; ErbB Receptors; Methionine; Phosphorylation; Receptors, Transferrin; Tetradecanoylphorbol Acetate; Vanadates

1987
Epidermal-growth-factor-dependent transformation by a human EGF receptor proto-oncogene.
    Science (New York, N.Y.), 1987, Dec-04, Volume: 238, Issue:4832

    The epidermal growth factor (EGF) receptor gene EGFR has been placed in a retrovirus vector to examine the growth properties of cells that experimentally overproduce a full-length EGF receptor. NIH 3T3 cells transfected with the viral DNA or infected with the corresponding rescued retrovirus developed a fully transformed phenotype in vitro that required both functional EGFR expression and the presence of EGF in the growth medium. Cells expressing 4 x 10(5) EGF receptors formed tumors in nude mice, while control cells did not. Therefore, the EGFR retrovirus, which had a titer on NIH 3T3 cells that was greater than 10(7) focus-forming units per milliliter, can efficiently transfer and express this gene, and increased numbers of EGF receptors can contribute to the transformed phenotype.

    Topics: Animals; Cell Transformation, Neoplastic; Cells, Cultured; DNA, Recombinant; Epidermal Growth Factor; ErbB Receptors; Fibroblasts; Genetic Vectors; Harvey murine sarcoma virus; Humans; Male; Mice; Mice, Nude; Neoplasms, Experimental; Proto-Oncogene Mas; Proto-Oncogenes; Recombinant Proteins

1987
Overexpression of the human EGF receptor confers an EGF-dependent transformed phenotype to NIH 3T3 cells.
    Cell, 1987, Dec-24, Volume: 51, Issue:6

    The epidermal growth factor receptor (EGFR) gene is frequently amplified and/or overexpressed in human malignancies. To investigate the biological effects of its overexpression, we constructed a eukaryotic vector containing human EGFR cDNA. Introduction of this construct led to reconstitution of functional EGF receptors in NR6 mutant cells, which are normally devoid of this receptor. Transfection of NIH 3T3 resulted in no significant alterations in growth properties. However, EGF addition led to the formation of densely growing transformed foci in liquid culture and colonies in semisolid medium. NIH 3T3-EGFR clonal lines, which expressed the EGF at 500- to 1000-fold levels over control NIH 3T3 cells, demonstrated a marked increase in DNA synthesis in response to EGF. Thus EGF receptor overexpression appears to amplify normal EGF signal transduction. Finally, high levels of EGFR expression, which conferred a transformed phenotype to NIH 3T3 cells in the presence of ligand, were demonstrated in representative human tumor cell lines that contained amplified copies of the EGFR gene.

    Topics: Animals; Cell Division; Cell Line, Transformed; Cell Transformation, Neoplastic; Epidermal Growth Factor; ErbB Receptors; Gene Amplification; Humans; Phenotype; Transfection

1987
Gangliosides as bimodal regulators of cell growth.
    Proceedings of the National Academy of Sciences of the United States of America, 1987, Volume: 84, Issue:1

    The B subunit of cholera toxin, which binds specifically to several molecules of ganglioside galactosyl-(beta 1----3)-N-acetylgalactosyminyl(beta 1----4)-[N- acetylneuraminyl(alpha 2----3)]-galactosyl(beta 1----4)glucosyl(beta 1----1) ceramide (GM1) on the cell surface, stimulated DNA synthesis and cell division in quiescent, nontransformed mouse 3T3 cells in a dose-dependent manner. In addition, the B subunit potentiated the response of the 3T3 cells to other mitogens, such as epidermal growth factor, platelet-derived growth factor, and insulin. This synergistic effect indicates that the B subunit does not act identically to any of these growth factors but probably modulates a common effector system crucial for cell proliferation. In distinct contrast, the B subunit inhibited the growth of ras-transformed 3T3 cells as well as rapidly dividing normal 3T3 cells. Thus, the same cells, depending on their state of growth, exhibited a bimodal response to the B subunit. We conclude that endogenous gangliosides may be bimodal regulators of positive and negative signals for cell growth.

    Topics: Animals; Cell Line; Cell Transformation, Neoplastic; Cells, Cultured; Cholera Toxin; DNA Replication; Drug Synergism; Epidermal Growth Factor; Gangliosides; Growth Substances; Insulin; Kinetics; Macromolecular Substances; Mice; Mice, Inbred BALB C; Mice, Inbred Strains; Oncogenes

1987
Establishment and preliminary growth characteristics of a transformed mouse cerebral microvessel endothelial cell line.
    Laboratory investigation; a journal of technical methods and pathology, 1986, Volume: 54, Issue:5

    A clone of spontaneously transformed mouse cerebral microvessel endothelial cells has been characterized and its growth characteristics have been investigated. The clone has been designated TEn and has been derived from parent nontransformed cerebral microvessel endothelial cells after multiple passages in culture. These transformed endothelial cells maintained characteristics of other transformed cells: anchorage-independent growth in soft agar, nude mouse tumorigenicity, and growth to high saturation density in culture with lack of contact inhibition. Although transformed, these cells still maintained endothelial functions and markers. These cells respond to both epidermal growth factor and platelet-derived growth factor. Although the TEn cells utilize serum factors for growth, they have maintained in completely serum-free media for up to 3 months. Additionally, serum-free media conditioned by these cells contained a growth factor-like activity that was mitogenic for the nontransformed parent endothelial cells. The TEn cells' maintenance of endothelial functions, growth in serum-free media, production of growth factor-like activity, and the response to well-characterized purified mitogens (epidermal growth factor and platelet-derived growth factor) suggest that these cells may be useful in studying growth control of cerebral microvessel-derived endothelial cells as well as in studies of growth factor production by transformed cells.

    Topics: Animals; Brain; Cell Division; Cell Line; Cell Transformation, Neoplastic; Clone Cells; Endothelium; Epidermal Growth Factor; Factor VIII; Female; gamma-Glutamyltransferase; Lectins; Mice; Mice, Nude; Microcirculation; Microscopy, Electron; Neoplasm Transplantation; Plant Lectins; Platelet-Derived Growth Factor; Receptors, Cell Surface; Receptors, Thrombin

1986
Enhancement by 1 alpha,25-dihydroxyvitamin D3 of chemically induced transformation of BALB 3T3 cells without induction of ornithine decarboxylase or activation of protein kinase C1.
    Cancer research, 1986, Volume: 46, Issue:2

    We reported previously that 1 alpha,25-dihydroxyvitamin D3 [1 alpha,25(OH)2D3], a hormonally active form of vitamin D3, markedly enhanced methylcholanthrene-induced transformation of BALB 3T3 A31-1-1 cells. When the cells were treated with methylcholanthrene (1 microgram/ml) for 72 h and then with 1 alpha,25(OH)2D3 (5 ng/ml) for 2 wk, the transformation frequency was 1.95 +/- 0.73 (SD) foci/dish in 8 independent experiments, which was about 20 times that in cultures treated with methylcholanthrene only. Even at a physiological concentration in plasma, i.e., 0.05 ng/ml, 1 alpha,25(OH)2D3 enhanced the transformation frequency significantly (P less than 0.001). 1 alpha,25(OH)2D3 was not cytotoxic but slightly inhibited growth of the cells. Cells treated with 1 alpha,25(OH)2D3 were thin and became arranged in a meshwork with wide intercellular spaces. These morphological changes were reversible. 1 alpha,25(OH)2D3 induced DNA synthesis in quiescent BALB 3T3 cells dose and time dependently, but this effect was less than that of 12-O-tetradecanoylphorbol-13-acetate. Unlike 12-O-tetradecanoylphorbol-13-acetate, 1 alpha,25(OH)2D3 did not interfere with the binding of epidermal growth factor or phorbol dibutyrate. 1 alpha,25(OH)2D3 did not induce ornithine decarboxylase. Moreover, it did not activate protein kinase C in quiescent BALB 3T3 cells or this enzyme isolated from mouse brain. BALB 3T3 cells and their transformants contain a specific cytosol receptor for 1 alpha,25(OH)2D3, but the binding sites of the transformants were fewer and had lower affinity than those of untransformed BALB 3T3 cells. These effects of 1 alpha,25(OH)2D3 were specific, because other derivatives of vitamin D3 induced the same effects only at 200 times or more higher concentrations.

    Topics: Animals; Caenorhabditis elegans Proteins; Calcitriol; Carrier Proteins; Cell Line; Cell Survival; Cell Transformation, Neoplastic; DNA; Drug Synergism; Enzyme Activation; Enzyme Induction; Epidermal Growth Factor; ErbB Receptors; Methylcholanthrene; Mice; Ornithine Decarboxylase; Protein Kinase C; Receptors, Cell Surface; Receptors, Drug; Receptors, Immunologic; Tetradecanoylphorbol Acetate

1986
Partial purification of transforming growth factors from human milk.
    Cancer research, 1986, Volume: 46, Issue:2

    Crude, delipidated milk and the acid:ethanol extracts of primary human breast tumors contain several activities that biologically resemble transforming growth factors (TGFs) in that they promote the anchorage-independent growth of normal rat kidney and Mm5mt/c1 mouse mammary tumor cells in soft agar. Three major TGF species with isoelectric points (pl) of about 4.0, 6.0-6.5, and 7.0 have been detected in both tumors and milk. The pl 4.0 species from milk has been purified about 10,000-fold by isoelectric focusing and high-performance liquid chromatography. This species, designated milk-derived growth factor II (MDGFII), coelutes from gel filtration columns with an authentic human epidermal growth factor standard when using a low ionic strength eluting buffer. However, on the same column, MDGFII is completely resolved from human epidermal growth factor with high ionic strength eluting buffers. Nevertheless, MDGFII purified by the latter technique still competes with 125I-epidermal growth factor for receptor binding to A431 cell membranes. Additionally the TGF activity of MDGFII present in the pl 4.0 fraction of milk is markedly inhibited by anti-epidermal growth factor receptor antibody preparations. Consequently MDGFII appears to be an alpha-TGF. MDGFII is a pepsin-sensitive, disulfide reducing agent-sensitive, heat-stable protein that may be physiologically important for the mammary gland or the neonate.

    Topics: Animals; Breast Neoplasms; Cell Transformation, Neoplastic; Chromatography, High Pressure Liquid; Epidermal Growth Factor; ErbB Receptors; Female; Growth Substances; Humans; Isoelectric Point; Mammary Neoplasms, Experimental; Mice; Milk Proteins; Milk, Human; Peptides; Receptors, Cell Surface; Transforming Growth Factors

1986
Effect of phorbol esters on the activity of purified transforming growth factors.
    Cancer research, 1986, Volume: 46, Issue:4 Pt 1

    Application of either phorbol esters or transforming growth factors to normal cells in culture may induce the appearance of a malignant phenotype. Since the mechanism of transformation by both chemical tumor promoters and growth factors is unknown, it was of interest to investigate whether phorbol esters might potentiate the activity of transforming growth factors. The combination of beta-transforming growth factor (beta-TGF) and epidermal growth factor (EGF) causes the normally anchorage-dependent cells (NRK-49F) to form colonies in soft agar. In the presence of maximally stimulating concentrations of EGF and suboptimally stimulating concentrations of beta-TGF, phorbol 12-myristate-13-acetate (PMA) enhances the soft agar colony growth of NRK-49F cells in a concentration-dependent manner. However, PMA alone or in combination with either EGF or beta-TGF does not stimulate soft agar growth. In contrast to the stimulation of PMA, 4 alpha-phorbol 12,13-didecanoate, an inactive phorbol ester, does not potentiate the effects of EGF plus beta-TGF on soft agar growth. PMA does not stimulate the growth of NRK-49F cells in monolayer, nor does it further potentiate the monolayer growth induced by EGF with or without beta-TGF. Because the addition to NRK-49F cells of compounds which potentiate beta-TGF activity increases EGF receptor number, the effects of PMA on the EGF receptor were studied. A short exposure to PMA (30 min) induces a 50% decrease in EGF receptors of NRK cells whether or not they have been exposed to beta-TGF. Scatchard analysis shows that this decrease involves primarily high affinity EGF receptors. However, cells treated with PMA for longer periods of time (4, 6, and 24 h) show no change in EGF or beta-TGF receptor binding. PMA therefore must potentiate the activity of purified TGFs without causing an increase in EGF or beta-TGF receptor binding.

    Topics: Animals; Cell Transformation, Neoplastic; Cells, Cultured; Drug Synergism; Epidermal Growth Factor; ErbB Receptors; Kidney; Peptides; Phorbols; Rats; Receptors, Cell Surface; Tetradecanoylphorbol Acetate; Transforming Growth Factors

1986
Transforming growth factor-beta and retinoic acid modulate phenotypic transformation of normal rat kidney cells induced by epidermal growth factor and platelet-derived growth factor.
    The Journal of biological chemistry, 1986, Apr-15, Volume: 261, Issue:11

    In this study we have investigated the ability of epidermal growth factor (EGF), platelet-derived growth factor (PDGF), and transforming growth factor-beta (TGF beta) together with retinoic acid (RA) at saturating concentrations to induce phenotypic transformation of normal rat kidney (NRK) cells in a growth factor-defined medium. This medium contains serum in which all growth factor activity has been chemically inactivated, thereby eliminating the effects of growth factors from serum in the assay. It is shown that neither TGF eta nor a ligand binding to the EGF receptor is essential for phenotypic transformation of NRK cells, since anchorage-independent growth is also induced by EGF in combination with RA and by PDGF in combination with RA and TGF beta. Our data indicate strong similarities between TGF beta and RA in their ability to act as modulators for phenotypic transformation. In addition, both agents enhance the number of EGF receptors in NRK cells, without affecting the number of PDGF receptors. On the other hand, TGF beta has mitogenic effects on a number of non-transformed cell lines, such as Swiss 3T3 fibroblasts, particularly when assayed in the absence of insulin, whereas RA is mitogenic for these cells only in the presence of insulin. These data demonstrate that phenotypic transformation of NRK cells requires specific combinations of polypeptide growth factors and modulating agents, but that this process can be induced under many more conditions than previously described. Moreover, our data point toward both parallels and differences in the activities of TGF beta and RA.

    Topics: Animals; Cell Line; Cell Transformation, Neoplastic; Epidermal Growth Factor; ErbB Receptors; Fibroblasts; Humans; Insulin; Kidney; Mice; Mitosis; Peptides; Platelet-Derived Growth Factor; Rats; Receptors, Cell Surface; Transforming Growth Factors; Tretinoin

1986
Cell-mediated co-action of transforming growth factors: incubation of type beta with normal rat kidney cells produces a soluble activity that prolongs the ruffling response to type alpha.
    The Journal of cell biology, 1986, Volume: 102, Issue:4

    Intense, continuous ruffling is a characteristic of many transformed cells, but untransformed cells ruffle intensely only briefly after exposure to growth factors. We reported previously that cells of a normal rat kidney (NRK) cell line transformed by Kirsten murine sarcoma virus secrete their own ruffle-inducing agent(s) that cause sustained ruffling in either themselves or untransformed NRK cells. In the present study, we examined the roles of the transforming growth factors TGF-alpha and TGF-beta in the induction and maintenance of ruffling in untransformed NRK cells and observed the following: TGF-alpha caused a transient epidermal growth factor (EGF)-like response, which could be blocked by prior exposure of cells to EGF or by antiserum directed against the COOH-terminus of TGF-alpha. TGF-beta caused no ruffling and did not itself prolong TGF-alpha ruffling. A new, buffer-soluble (transferable) mediator activity produced by incubation of TGF-beta with NRK cells for 6-h extended the duration of maximal TGF-alpha-induced ruffling by several-fold. This study demonstrates that TGF-alpha alone causes an EGF-like, transient ruffling response, but neither TGF-alpha or TGF-beta alone, nor the two together, cause transformation-associated sustained ruffling. Rather, TGF-alpha acts in concert with a new, TGF-beta-dependent activity. This new activity appears to inhibit normal cellular off-regulation of TGF-alpha-induced ruffling. Inhibition of the cellular off-regulation of a growth factor response could play a key role in the unregulated growth associated with malignancy.

    Topics: Animals; Cell Line; Cell Transformation, Neoplastic; Culture Media; Epidermal Growth Factor; Growth Substances; Kidney; Kinetics; Kirsten murine sarcoma virus; Peptides; Rats; Sarcoma Viruses, Murine; Structure-Activity Relationship; Transforming Growth Factors

1986
An agent or agents produced by virus-transformed cells cause unregulated ruffling in untransformed cells.
    The Journal of cell biology, 1986, Volume: 102, Issue:4

    KNRK cells (a normal rat kidney [NRK] cell line transformed by Kirsten murine sarcoma virus) in sparse culture exhibit a highly ruffled morphology, but the cause of this ruffling is unknown. In this study, we have demonstrated that the continuous, excess ruffling on KNRK cells is caused by one or more soluble agents secreted by the KNRK cells themselves. To do this study, an assay for ruffling responses in live cell cultures was defined, and its reproducibility was demonstrated. This assay permitted observation of the kinetics of ruffling responses (percentage of cells ruffled as a function of time after stimulation). This method was used to compare the kinetics of ruffling induced by insulin, epidermal growth factor, fibroblast growth factor, glucose, and KNRK cell conditioned medium (CM). Ruffling was elicited on NRK cells by each of the polypeptide mitogens and nutrients, but, in each case, this ruffling subsided spontaneously within an hour. CM from KNRK cells also caused ruffling movements on untransformed NRK cells, but this ruffling continued for at least 20 h. This response was largely blocked by premixing the KNRK cell CM with rabbit IgG against rat transforming growth factor, type alpha, (TGF-alpha). KNRK cells made quiescent (ruffle free) by a pH shift (from 7.4 to 8.4) responded to insulin, glucose, and KNRK cell CM with kinetics similar to those observed for each of these factors in NRK cells. The unusual feature for the ruffle-inducing agent(s) produced by KNRK cells was that this activity was not subject, in either NRK or KNRK cells, to the cellular off-regulation that limits the responses to insulin or glucose. Thus, the continuous ruffling of KNRK cells is caused by their own unregulated ruffle-inducing agent or agents, which appear to include TGF-alpha. This work also demonstrates that kinetic analysis of cellular responses to exogenous factors can provide new insights into the regulatory mechanisms involved in the normal limitation of these responses.

    Topics: Animals; Cell Line; Cell Transformation, Neoplastic; Culture Media; Epidermal Growth Factor; Fibroblast Growth Factors; Glucose; Insulin; Kidney; Kinetics; Kirsten murine sarcoma virus; Rats; Sarcoma Viruses, Murine; Vitamins

1986
Human fibroblast collagenase. Complete primary structure and homology to an oncogene transformation-induced rat protein.
    The Journal of biological chemistry, 1986, May-15, Volume: 261, Issue:14

    We have determined the complete sequence of the cDNA clone representing the full size human skin collagenase mRNA. Collagenase is synthesized in preproenzyme form, Mr 54,092, with a 19 amino acid long signal peptide. The primary secretion products of the enzyme consist of a minor glycosylated form, Mr 57,000, and a major unmodified polypeptide of predicted Mr 51,929. Proteolytic activation of human skin procollagenase results in removal of 81 amino acid residues from the amino-terminal portion of the proenzyme. Both potential N-glycosylation sites are contained within the proteolytically activated form of the enzyme. The primary structure of the coding region of the presented clone is homologous to an oncogene-induced rat protein whose function is still unknown, although preliminary observations suggest that it is not rat skin collagenase.

    Topics: Amino Acid Sequence; Animals; Base Sequence; Cell Transformation, Neoplastic; Cells, Cultured; Collagenases; DNA; Enzyme Precursors; Epidermal Growth Factor; Fibroblasts; Humans; Microbial Collagenase; Neoplasm Proteins; Oncogenes; Rats; RNA, Messenger; Skin

1986
Cooperation of mitogenic growth factors with polyoma virus middle T antigen in transformation of secondary cultured rat cells.
    Biochemical and biophysical research communications, 1986, May-14, Volume: 136, Issue:3

    Platelet derived growth factor cooperated with middle T antigen in inducing growth in agarose medium of secondary cultured rat embryo cells transfected with a polyoma virus middle T antigen cDNA clone. In contrast, epidermal growth factor and a conditioned medium containing transforming growth factor did not stimulate the colony-forming efficiency of such cells in the agarose medium.

    Topics: Animals; Antigens, Polyomavirus Transforming; Antigens, Viral, Tumor; Cell Transformation, Neoplastic; Cells, Cultured; Epidermal Growth Factor; Female; Fibroblasts; Oncogene Proteins, Viral; Peptides; Platelet-Derived Growth Factor; Polyomavirus; Pregnancy; Rats; Rats, Inbred F344; Sepharose; Transfection; Transforming Growth Factors

1986
8th San Antonio Breast Cancer Symposium--Plenary lecture. Autocrine and paracrine growth regulation of human breast cancer.
    Breast cancer research and treatment, 1986, Volume: 7, Issue:2

    We consider the hypothesis that estrogen control of hormone dependent breast cancer is mediated by autocrine and paracrine growth factors secreted by the breast cancer cells themselves. Though we show direct, unmediated effects of estrogen on specific cell functions, we also provide evidence that human breast cancer cells secrete a collection of growth factors (IGF-I, TGF alpha, TGF beta, a PDGF-like competency factor, and at least one new epithelial colony stimulating factor). Some of these are estrogen-regulated in hormone dependent cells, and are constitutively increased in cells which acquire independence either spontaneously or by ras transfection. Collectively, the secreted growth factors are capable of promoting tumor formation by MCF-7 cells in nude mice, though not to the same extent as estrogens. There would seem to be potential for clinical intervention in the autocrine and paracrine control of breast cancer cells, including some cells which are no longer dependent on estrogens.

    Topics: Breast Neoplasms; Cell Division; Cell Line; Cell Transformation, Neoplastic; Epidermal Growth Factor; Epithelium; ErbB Receptors; Estrogens; Female; Growth Substances; Humans; Insulin-Like Growth Factor I; Neoplasm Invasiveness; Organic Chemicals; Peptides; Platelet-Derived Growth Factor; Receptors, Cell Surface; Receptors, Estrogen; Transforming Growth Factors; Tumor Stem Cell Assay

1986
Transformation of rat hepatocytes by transfection with simian virus 40 DNA to yield proliferating differentiated cells.
    Cancer research, 1986, Volume: 46, Issue:8

    Cultured hepatocytes from adult Fischer 344 rats were transformed by virion or cloned simian virus 40 (SV40) DNA using the calcium phosphate method. Transformation by SV40 occurred in either serum-supplemented medium or chemically defined medium (CDM). The frequency was greatest in serum-supplemented medium but transformants did not remain differentiated. In contrast, SV40 transformants developed less frequently in CDM, but retained differentiated functions. The frequency of transformation was enhanced by treatments that stimulated cell proliferation, in particular supplementing CDM with epidermal growth factor. Hepatocytes transformed in CDM were epithelial in morphology, secreted albumin, transferrin, hemopexin, and expressed the enzyme glucose-6-phosphatase, all characteristics of normal liver. Transformants did not produce detectable levels of alpha-fetoprotein, a marker of fetal or abnormal liver. We conclude that (a) hepatocytes can be transformed by transfection with SV40 DNA; (b) the frequency of transformation is enhanced by stimulating DNA synthesis; and (c) the transformed cells retain specific functions of normal hepatocytes in situ. Using this system it will be possible to study transformation of hepatocytes by viral and cellular oncogenes and to determine their effects on hepatocellular differentiation.

    Topics: Animals; Antigens, Viral; Cell Differentiation; Cell Division; Cell Transformation, Neoplastic; Culture Media; Dimethyl Sulfoxide; Epidermal Growth Factor; Glucose-6-Phosphatase; Liver Neoplasms, Experimental; Male; Oncogenes; Rats; Rats, Inbred F344; Serum Albumin; Simian virus 40; Transfection; Transferrin

1986
Differences in sequences encoding the carboxyl-terminal domain of the epidermal growth factor receptor correlate with differences in the disease potential of viral erbB genes.
    Proceedings of the National Academy of Sciences of the United States of America, 1986, Volume: 83, Issue:16

    Eleven recently isolated erbB-transducing viruses as well as avian erythroblastosis virus (AEV)-R (ES4) and AEV-H have been characterized for the type of disease they cause, their ability to transform fibroblasts in culture, their ability to cause disease in pedigrees of chicken that differ in susceptibility to erbB-induced erythroblastosis, and the structure of their erbB genes. Differences in each of the biological parameters correlated with differences in erbB sequences encoding the C-terminal domain of the epidermal growth factor receptor (EGFR). Seven viruses were strain restricted in their ability to induce erythroblastosis and did not transform fibroblasts. These seven viruses contained v-erbB genes encoding the complete C terminus of the EGFR. AEV-R and AEV-H were not pedigree restricted in their ability to induce erythroblastosis and could transform fibroblasts. These viruses contain v-erbB genes that lack codons for the immediate C terminus of the EGFR. Three viruses caused angiosarcoma and one caused fibrosarcoma. The angiosarcoma and fibrosarcoma-inducing viruses were not strain restricted and did not cause erythroblastosis. The v-erbB genes of each of these viruses contained extensive internal deletions or 3' truncations in sequences encoding the C-terminal domain of the EGFR.

    Topics: Alpharetrovirus; Animals; Avian Leukosis; Avian Leukosis Virus; Base Sequence; Cell Line; Cell Transformation, Neoplastic; Chick Embryo; Chickens; Epidermal Growth Factor; ErbB Receptors; Genes; Genes, Viral; Leukemia, Erythroblastic, Acute; Receptors, Cell Surface; Transduction, Genetic

1986
Induction of anchorage-independent growth by epidermal growth factor and altered sensitivity to type beta transforming growth factor in partially transformed rat kidney cells.
    Cancer research, 1986, Volume: 46, Issue:11

    A partially transformed cell line (NRK-PT14) was isolated from normal rat kidney (NRK) cells. Like NRK cells, NRK-PT14 cells required epidermal growth factor for anchorage-independent growth, but lost the additional requirement for exogenous type beta transforming growth factor (TGF-beta). Compared to NRK cells, NRK-PT14 cells did not secrete elevated levels of TGF-beta, but exhibited an altered response to this growth factor. Monolayer growth of NRK cells in a serum-free medium was inhibited by TGF-beta, whereas growth of NRK-PT14 cells was stimulated by TGF-beta. In addition, TGF-beta stimulated epidermal growth factor binding to high affinity sites in NRK cells, but decreased epidermal growth factor binding to NRK-PT14 cells during growth of the cells in serum-free medium. These qualitative changes in the response to TGF-beta may be representative of an intermediate stage in the spontaneous transformation of NRK cells.

    Topics: Animals; Cell Cycle; Cell Line; Cell Transformation, Neoplastic; Culture Media; Epidermal Growth Factor; ErbB Receptors; Mice; Mice, Nude; Neoplasms, Experimental; Peptides; Rats; Sarcoma Viruses, Murine; Transforming Growth Factors

1986
Differential responsiveness of myc- and ras-transfected cells to growth factors: selective stimulation of myc-transfected cells by epidermal growth factor.
    Molecular and cellular biology, 1986, Volume: 6, Issue:3

    To identify functional relationships between oncogenes and growth factors, we compared the effects of transfected myc and ras oncogenes on the responsiveness of Fischer rat 3T3 cells to three growth factors: epidermal growth factor (EGF), platelet-derived growth factor (PDGF), and transforming growth factor-beta (TGF-beta). Control cells did not grow in soft agar under any conditions. ras-Transfected cells grew in soft agar under all conditions tested and were insensitive to the stimulatory effects of exogenous growth factors. These cells secreted elevated levels of both EGF-like factors and TGF-beta, suggesting that the lack of responsiveness of these cells to exogenous growth factors arose from autocrine stimulation. myc-Transfected cells displayed conditional anchorage-independent growth: they formed numerous colonies in soft agar in the presence of EGF but relatively few colonies in the presence of PDGF or TGF-beta. Secretion of EGF-like factors and TGF-beta by these cells was not elevated above that of control cells. These results suggest a model for the mechanism of cooperation between myc and ras oncogenes in which ras-like genes induce growth factor production, while myc-like genes increase the responsiveness of cells to these factors.

    Topics: Animals; Antigens, Polyomavirus Transforming; Antigens, Viral, Tumor; Cell Division; Cell Transformation, Neoplastic; Cells, Cultured; Epidermal Growth Factor; Genes; Genes, Viral; Growth Substances; Oncogene Proteins, Viral; Oncogenes; Peptides; Platelet-Derived Growth Factor; Polyomavirus; Rats; Rats, Inbred F344; Transfection; Transforming Growth Factors

1986
c-erbB activation in avian leukosis virus-induced erythroblastosis: multiple epidermal growth factor receptor mRNAs are generated by alternative RNA processing.
    Molecular and cellular biology, 1986, Volume: 6, Issue:9

    Avian leukosis virus-induced erythroblastosis results from the specific interruption of the host oncogene, c-erbB, by the insertion of an intact provirus. This insertion results in the expression of two size classes (3.6 and 7.0 kilobases [kb]) of truncated c-erbB transcripts which are initiated in the 5' long terminal repeat of the integrated provirus. Through sequence analysis of erbB cDNA clones we have previously shown that the 3.6-kb activated erbB mRNA contains portions of viral gag and env genes fused to c-erbB sequences (T.W. Nilsen, P.A. Maroney, R.G. Goodwin, F.M. Rottman, L.B. Crittenden, M.A. Raines, and H.-J. Kung, Cell 41:719-726, 1985). In this report we show that the 7-kb mRNA differs from the shorter activated c-erbB mRNA in the length of its 3' untranslated sequence such that the longer mRNA has an extremely long (4.3 kb) 3' untranslated sequence. Additionally, we demonstrate that activated c-erbB mRNA precursors can be processed by alternative splicing to yield mRNAs with viral gag sequences fused directly to c-erbB sequences. Finally, blot hybridization evidence suggests that the two size classes of activated c-erbB mRNA in erythroblastic tissue represent truncated versions of the two c-erbB mRNAs present in normal tissue.

    Topics: Animals; Avian Leukosis Virus; Base Sequence; Cell Transformation, Neoplastic; Cloning, Molecular; DNA; Epidermal Growth Factor; Erythroblasts; Oncogenes; RNA Splicing; RNA, Messenger; Transcription, Genetic

1986
Purification and biological properties of type beta transforming growth factor from mouse transformed cells.
    Cancer research, 1986, Volume: 46, Issue:10

    Transforming growth factor type beta (TGF beta) has been purified from serum-free culture fluids of transformed mouse L-929 cells which are capable of continual growth in serum-free medium in the absence of any exogenously added polypeptide growth factors. TGF beta has been purified to homogeneity as judged by NH2-terminal amino acid sequence analysis. Analysis of the purified polypeptide by gel electrophoresis indicates that TGF beta is composed of two polypeptide chains of Mr 12,500 cross-linked by disulfide bonds. TGF beta was characterized by its ability to induce anchorage-dependent normal rat kidney (NRK) cells to grow in soft agar in the presence of epidermal growth factor (EGF). TGF beta was also able to enhance both EGF-induced DNA synthesis and cell proliferation on growth-arrested NRK cells in monolayer cultures under serum-free conditions. We also show that in mouse melanoma B-16 cells under serum-free conditions TGF beta stimulates both DNA synthesis in monolayer cultures and anchorage-independent growth in soft agar. Paradoxically, the anchorage-independent growth in the presence of serum of many human cell lines, including melanomas, and mammary, prostatic, vulvar, and lung carcinomas is inhibited by TGF beta at saturating concentrations similar to those that stimulate colony formation of the rodent cell lines L-929 and B-16 under serum-free conditions. The peculiar action of TGF beta is further revealed by the observations that while EGF and TGF beta synergize to induce inhibition of anchorage-independent growth of A-431 human vulvar carcinoma cells, their effects on the anchorage-independent growth of one human lung carcinoma cell line (A-549) and two human prostatic carcinoma cell lines (PC-3 and DU-145) are antagonistic. Moreover, we show that in the rodent and human cell lines TGF beta interacts with specific cellular receptors which may mediate the actions of TGF beta. We conclude that the expression of both TGF beta and TGF beta receptors by L-929 cells and the stimulation of growth of L-929 cells in serum-free medium by TGF beta suggests that TGF beta may be important for maintaining the transformed state of this tumor cell line, and the way in which a cell responds to TGF beta is dependent on the presence or absence of growth factors contained in the serum.

    Topics: Amino Acid Sequence; Breast; Cell Division; Cell Line; Cell Transformation, Neoplastic; DNA; Epidermal Growth Factor; Growth Substances; Humans; Kidney; Melanoma; Molecular Weight; Neoplasm Proteins; Neoplasms; Peptides; Transforming Growth Factors

1986
Retinoic acid restores normal growth control to a transformed mouse embryo fibroblast cell line.
    Cancer letters, 1986, Volume: 33, Issue:1

    The effects of retinoic acid on a transformed mouse embryo fibroblast cell line (AKR-MCA) were examined. Treatment with retinoic acid restored a non-transformed phenotype to this transformed cell line in a dose dependent manner. Retinoic acid (RA) treated AKR-MCA cells showed a non-transformed morphology, a slower growth rate, and did not grow with anchorage independence. A 38,000 Da protein was phosphorylated to a high degree in the AKR-MCA transformed cell line compared to the non-transformed AKR-2B cell line. RA treatment greatly reduced the level of phosphorylation of this protein in AKR-MCA cells. Growth arrested AKR-MCA cells showed a mitogenic response to nutrient replenishment, but not to epidermal growth factor (EGF). Treatment of AKR-MCA cells with RA restored their ability to respond to EGF while the response to nutrient replenishment was lost. This pattern of growth control was similar to that of the non-transformed AKR-2B cells.

    Topics: Animals; Cell Cycle; Cell Line; Cell Transformation, Neoplastic; DNA Replication; Epidermal Growth Factor; Mice; Phosphoproteins; Tretinoin

1986
Potentiation of growth factor activity by exogenous c-myc expression.
    Proceedings of the National Academy of Sciences of the United States of America, 1986, Volume: 83, Issue:21

    The c-myc oncogene has been implicated in deregulation of cell growth in neoplastic cells and response to "competence-inducing" growth factors in normal cells. In the latter case, expression of c-myc has been shown to be associated with the transition from the G0 to the G1 phase of the cell cycle induced by platelet-derived growth factor (PDGF). In the work reported here, we have introduced the c-myc coding region, in a retroviral vector, into mouse and rat cells. We show that under conditions of anchorage-independent growth, constitutive c-myc expression increases the response of rodent cells to PDGF, as well as to other growth factors of both the competence-inducing and "progression" classes. These effects of the myc product are observed whether or not an exogenous ras gene has also been introduced into the same cells. Possible models for the influence of myc on growth responses are discussed.

    Topics: Cell Line; Cell Transformation, Neoplastic; Epidermal Growth Factor; Growth Substances; Models, Biological; Peptide Biosynthesis; Platelet-Derived Growth Factor; Proto-Oncogenes; Transforming Growth Factors

1986
Growth control in cerebral microvessel-derived endothelial cells.
    Brain research, 1986, Oct-01, Volume: 384, Issue:1

    Endothelial (En) cells derived from the cerebral microvasculature were examined for their growth control properties. These cells were shown to be growth responsive to fetal bovine serum and arrested growth if serum was removed. They lend themselves to studies in chemically defined media since they survive well in serum-free medium with little or no proliferation. These cerebral microvessel En cells also respond to epidermal growth factor (EGF) and platelet-derived growth factor (PDGF), unlike some other endothelial cells. Similar to other endothelial cells, the En cells responded to fibroblast growth factor (FGF). The response to EGF and PDGF occurred in serum-free media, but only if both agents were present. The cells further responded mitogenically to conditioned media obtained from transformed endothelial cells. The En cells were found to bind EGF and displayed two orders of affinity as determined by Scatchard analysis, depending upon whether the cells were in confluent or subconfluent conditions. The data indicate that some endothelial cells respond to EGF and PDGF and that one mechanism for growth control of endothelial cells may be an ability to regulate high-affinity growth factor receptors for epidermal growth factor.

    Topics: Animals; Brain; Cell Count; Cell Division; Cell Transformation, Neoplastic; Cells, Cultured; Dose-Response Relationship, Drug; Endothelium; Epidermal Growth Factor; Flow Cytometry; Growth Substances; Mice; Mice, Inbred Strains; Microcirculation; Platelet-Derived Growth Factor; Serum Albumin, Bovine; Time Factors

1986
Activation of proto-oncogene expression by growth regulatory signals.
    Current topics in microbiology and immunology, 1986, Volume: 132

    Topics: Animals; Cell Transformation, Neoplastic; Cells, Cultured; Epidermal Growth Factor; Genes, Regulator; Mice; Proto-Oncogenes; Tetradecanoylphorbol Acetate

1986
PDGF-like growth factor induces EGF-potentiated phenotypic transformation of normal rat kidney cells in the absence of TGF beta.
    Biochemical and biophysical research communications, 1986, Dec-30, Volume: 141, Issue:3

    Using a growth factor defined assay for anchorage-independent growth (van Zoelen, E.J.J., van Oostwaard, Th.M.J., van der Saag, P.T. and de Laat, S.W. (1985) J. Cell. Physiol. 123, 151- 160, we have studied the ability of polypeptide growth factors produced by Neuro-2A neuroblastoma cells to induce anchorage-independent growth of normal rat kidney cells. Neuro-2A cells produce and secrete a PDGF-like growth factor in addition to TGF beta, which can be fully separated from each other by means of reverse-phase HPLC. Using a new, very sensitive technique for detection of TGF beta in growth factor samples based on its additional ability to act as a growth inhibitory factor, it is shown that the PDGF-like growth factor does not contain any detectable TGF beta. Still this neuroblastoma derived PDGF-like growth factor is able to induce anchorage-independent growth of NRK cells, particularly in the additional presence of EGF. It is concluded that under growth factor defined assay conditions TGF beta is not essential for phenotypic transformation of NRK cells.

    Topics: Animals; Cell Division; Cell Line; Cell Transformation, Neoplastic; Chromatography, High Pressure Liquid; Epidermal Growth Factor; Growth Substances; Kidney; Mice; Neuroblastoma; Peptides; Platelet-Derived Growth Factor; Rats; Transforming Growth Factors

1986
The involvement of activated ras genes in determining the transformed phenotype.
    Proceedings of the Royal Society of London. Series B, Biological sciences, 1985, Oct-22, Volume: 226, Issue:1242

    Activated ras oncogenes have been identified in a wide range of tumours. All examples of ras gene activation in tumours so far result from amino acid substitution at Gly12 or Gln61. To learn more about how mutations in ras genes lead to transformation, we have analysed transforming growth factor production in NIH/3T3 cells transformed by each of the three ras genes. These results show that the transformed phenotype of these cells results from a combination of the presence of the mutant ras protein and TGF alpha production. In a second series of experiments we have shown that the mutation of a ras gene in a tumour cell line can lead to tumour progression towards a more aggressive phenotype.

    Topics: Animals; Cell Transformation, Neoplastic; Cells, Cultured; Clone Cells; Epidermal Growth Factor; ErbB Receptors; Leukemia L5178; Mice; Mice, Inbred Strains; Mutation; Nucleic Acid Hybridization; Oncogenes; Peptides; Phenotype; Receptors, Cell Surface; Transfection; Transforming Growth Factors

1985
Neuroblastoma cells express c-sis and produce a transforming growth factor antigenically related to the platelet-derived growth factor.
    Molecular and cellular biology, 1985, Volume: 5, Issue:9

    Mouse neuroblastoma Neuro-2A cells produce transforming growth factors during exponential growth in a defined hormone-free medium, which, on Bio-Gel columns in 1 M HAc, elute at a molecular size of 15 to 20 kilodaltons (kDa). These neuroblastoma-derived transforming growth factors have strong mitogenic activity, but they do not compete with epidermal growth factor for receptor binding (E. J. J. van Zoelen, D. R. Twardzik, T. M. J. van Oostwaard, P. T. van der Saag, S. W. de Laat, and G. J. Todaro, Proc. Natl. Acad. Sci. U.S.A. 81:4085-4089, 1984). In this study approximately 80% of the mitogenic activity was immunoprecipitated by antibodies raised against platelet-derived growth factor (PDGF). Immunoblotting indicated a true molecular size of 32 kDa for this PDGF-like growth factor. Analysis of poly(A)+ RNA from Neuro-2A cells demonstrated the expression of the c-sis oncogene in this cell line, whereas in vitro translation of the RNA yielded a 20-kDa protein recognized by anti-PDGF antibodies. Separation by reverse-phase high-pressure liquid chromatography demonstrated the presence of two distinct mitogenic activities in neuroblastoma-derived transforming growth factor preparations, one of which is antigenically related to PDGF. Both activities had the ability to induce anchorage-independent growth in normal rat kidney cells, both in the presence and in the absence of epidermal growth factor. It is concluded that Neuro-2A cells express c-sis with concomitant production and secretion of a PDGF-like growth factor, which plays a role in the induction of phenotypic transformation on normal rat kidney cells.

    Topics: Animals; Cell Line; Cell Transformation, Neoplastic; Chromatography, High Pressure Liquid; Epidermal Growth Factor; Mice; Neuroblastoma; Oncogene Proteins v-sis; Peptide Biosynthesis; Peptides; Platelet-Derived Growth Factor; Poly A; Rats; Retroviridae Proteins; RNA, Messenger; Transforming Growth Factors

1985
Elevated growth factor levels in transformed mouse embryo cells treated with N,N-dimethylformamide.
    Cancer research, 1985, Volume: 45, Issue:1

    The transformed mouse embryo fibroblast cell line AKR-MCA, produces several transforming growth factor (TGF) activities which can be identified in cell extracts and serum-free conditioned medium. Treatment of these transformed cells with 1% N,N-dimethylformamide resulted in a more normal phenotype and an increased level of TGFs in cell extracts and conditioned medium. In addition, an 11-fold increase in an epidermal growth factor receptor-competing activity was observed in cell extracts and conditioned medium compared to control untreated cells. Fractionation of the conditioned medium on Bio-Gel P-100 showed that the same size classes of TGFs were present in N,N-dimethylformamide-treated as well as untreated cells. The increased EGF-receptor-competing activity was eluted in two peaks at Mr 6500 and Mr 4000. The Mr 6500 peak did not coelute with a TGF peak in the N,N-dimethylformamide-treated cells.

    Topics: Animals; Binding, Competitive; Cell Line; Cell Transformation, Neoplastic; Chromatography, Gel; Dimethylformamide; Embryo, Mammalian; Epidermal Growth Factor; ErbB Receptors; Female; Growth Substances; Humans; Mice; Mice, Inbred AKR; Molecular Weight; Peptide Biosynthesis; Peptides; Placenta; Pregnancy; Receptors, Cell Surface; Transforming Growth Factors

1985
Lack of correlation between loss of anchorage-independent growth and levels of transformation-specific p53 protein in retinoic acid-treated F9 embryonal carcinoma cells.
    Experimental cell research, 1985, Volume: 156, Issue:1

    It has been shown that differentiated derivatives of retinoic acid (RA)-treated F9 embryonal carcinoma cells become non-malignant. In the present study it is asked whether this loss of malignancy is due to cellular differentiation. Because the ability of cells to grow in suspension correlates with in vivo tumorigenicity, we determined the time course of the loss of this property, after RA treatment, with relation to the differentiation to parietal endoderm and the acquisition of normalcy in several common transformation-specific properties of F9 cells. Our results show that pretreatment with RA for 24 h caused 80% inhibition of anchorage-independent growth in F9 cells, and this inhibition reached its highest level (98%) after pretreatment with RA for 48 h and longer. However, all other observed transformation-related properties, and the levels of plasminogen activator (marker for parietal endoderm) remained unaltered at this early post-treatment stage. These observations suggest that the loss of malignancy is a relatively early event in the biochemical pathways involved in the RA-induced differentiation of F9 cells. Furthermore, our data show that the presence of elevated levels of p53 alone may not be sufficient to maintain the anchorage-independent growth and the rapid proliferation of F9 cells.

    Topics: Actins; Animals; Bucladesine; Cell Adhesion; Cell Differentiation; Cell Division; Cell Line; Cell Transformation, Neoplastic; Deoxyglucose; Embryonal Carcinoma Stem Cells; Epidermal Growth Factor; ErbB Receptors; Fibronectins; Mice; Neoplasm Proteins; Neoplastic Stem Cells; Phosphoproteins; Plasminogen Activators; Receptors, Cell Surface; Teratoma; Tretinoin; Tumor Suppressor Protein p53

1985
Phenotypic transformation of normal rat kidney cells in a growth-factor-defined medium: induction by a neuroblastoma-derived transforming growth factor independently of the EGF receptor.
    Journal of cellular physiology, 1985, Volume: 123, Issue:2

    Polypeptide growth factor activity in serum can be destroyed by treatment with dithiothreitol. When such growth-factor-inactivated serum is used as a supplement of culture media instead of regular serum, normal rat kidney (NRK) cells become quiescent unless defined polypeptide growth factors like insulin and epidermal growth factor (EGF) are added. On this basis a growth-factor-defined medium has been developed for NRK cells, which permits cell proliferation as rapidly as in media supplemented with serum, even at low cell densities. Moreover, cells can be serially passaged in this medium. NRK cells can be induced to grow in semisolid media when incubated with transforming growth factors. The growth-factor-defined medium permits soft agar growth experiments of NRK cells, without interference from polypeptide growth factors in serum. Using this assay system we have shown that EGF alone is unable to induce any degree of anchorage-independent growth in NRK cells. However, a recently identified transforming growth factor from mouse neuroblastoma cells which does not compete with EGF for receptor binding is able to induce progressively growing colonies of NRK cells in soft agar, even without additional EGF.

    Topics: Agar; Animals; Blood; Cell Division; Cell Line; Cell Transformation, Neoplastic; Clone Cells; Culture Media; Epidermal Growth Factor; ErbB Receptors; Growth Substances; Kidney; Mice; Neuroblastoma; Peptides; Phenotype; Rats; Receptors, Cell Surface; Transforming Growth Factors

1985
Multistep change in epidermal growth factor receptors during spontaneous neoplastic progression in Chinese hamster embryo fibroblasts.
    Cancer research, 1985, Volume: 45, Issue:5

    Whole Chinese hamster embryo lineages have been shown to undergo multistep spontaneous neoplastic progression during serial passage in culture. We have studied the binding, internalization, and degradation of 125I-labeled epidermal growth factor at four different stages of transformation. The whole Chinese hamster embryo cells lost cell surface epidermal growth factor receptors gradually during the course of neoplastic progression until only 10% of the receptor number present in the early-passage cells (precrisis) were retained in the late-passage cells (tumorigenic). No differences in internalization rates, chloroquine sensitivity, or ability to degrade hormone between the various passage levels were seen. No evidence for the presence in conditioned medium of transforming growth factors which might mask or down-regulate epidermal growth factor receptor was obtained. These results suggest that a reduction in cell surface epidermal growth factor receptor might be an early event during spontaneous transformation in whole Chinese hamster embryo cells.

    Topics: Animals; Cell Count; Cell Transformation, Neoplastic; Cells, Cultured; Chloroquine; Cricetinae; Cricetulus; Embryo, Mammalian; Epidermal Growth Factor; ErbB Receptors; Fibroblasts; Iodine Radioisotopes; Receptors, Cell Surface; Temperature

1985
Stimulation by insulin-like growth factors is required for cellular transformation by type beta transforming growth factor.
    The Journal of biological chemistry, 1985, Apr-25, Volume: 260, Issue:8

    Medium conditioned by BRL-3A cells, a known source of insulin-like growth factor II (IGF-II), induced phenotypic transformation (anchorage-independent proliferation) of mouse BALB/c 3T3 fibroblasts but not rat NRK-49F fibroblasts, in the presence of 10% calf serum. A specific radioreceptor assay and a bioassay indicated that BRL-3A conditioned medium contained 0.5-1 ng/ml of type beta transforming growth factor (beta TGF). Purified IGF-II and beta TGF acting together reconstituted the transforming activity of BRL-3A conditioned medium on BALB/c 3T3 cells. Insulin was 5-10% as potent as IGF-II in supporting the transforming action of beta TGF on BALB/c 3T3 cells. NRK-49F cells were phenotypically transformed by beta TGF in the presence of EGF and 10% calf serum as the sole source of IGFs. However, transformation of NRK-49F cells under these conditions was inhibited by addition of purified IGF-binding protein. Addition of an excess of IGF-II prevented the inhibitory action of IGF-binding protein. The different sensitivity of the two cell lines to IGFs was correlated with lower levels of type I IGF receptor and higher levels of type II IGF receptor in NRK-49F cells as compared with BALB/c 3T3 cells. The results suggest that cellular stimulation by IGFs is a prerequisite for transformation of rodent fibroblasts by beta TGF. We propose that transformation of fibroblasts by beta TGF requires concomitant stimulation by the set of growth factors that support normal cell proliferation.

    Topics: Animals; Cell Line; Cell Transformation, Neoplastic; Chromatography, Gel; Dose-Response Relationship, Drug; Epidermal Growth Factor; Fibroblasts; Insulin; Mice; Mice, Inbred BALB C; Peptides; Phenotype; Rats; Receptor, Insulin; Receptors, Cell Surface; Receptors, Somatomedin; Somatomedins; Transforming Growth Factors

1985
A beta-type transforming growth factor, present in conditioned cell culture medium independent of cell transformation, may derive from serum.
    Journal of cellular biochemistry, 1985, Volume: 27, Issue:4

    An alpha-type transforming growth factor (TGF alpha) is produced at high levels by rat embryo cells transformed by the Snyder-Theilen strain of feline sarcoma virus (FeSV). Addition of 2 ng mouse epidermal growth factor (mEGF) during purification identified the presence of a second, EGF-dependent growth factor of the TGF beta type (TGF beta) in this conditioned medium. This factor had an approximate Mr of 12,000 and eluted at 37% acetonitrile during high performance liquid chromatography. This extracellular type of TGF beta activity also was present in conditioned medium of rat cells after infection with a transformation defective strain of Abelson leukemia virus, and hence expression of this growth factor activity was independent of cell transformation. Moreover, the presence of an EGF-dependent, 12,000 Mr clonogenic activity in extracts of bovine serum alone suggests serum as an origin for the B-type transforming growth factor initially observed in conditioned medium of Snyder-Theilen FeSV transformed cells. This does not, however, preclude the possibility that TGF beta is also secreted by the transformed rat embryo cells themselves.

    Topics: Animals; Blood; Cell Line; Cell Transformation, Neoplastic; Cell Transformation, Viral; Chromatography, High Pressure Liquid; Culture Media; Embryo, Mammalian; Epidermal Growth Factor; Fibroblasts; Molecular Weight; Peptides; Rats; Sarcoma Viruses, Feline; Transforming Growth Factors

1985
Purification and characterization of a low molecular weight transforming growth factor from the urine of melanoma patients.
    The Journal of biological chemistry, 1985, Aug-05, Volume: 260, Issue:16

    A low Mr human transforming growth factor (TGF) present in melanoma patients' urine has been purified approximately 200,000-fold to apparent homogeneity. Initial purification of an acid-soluble fraction of urine was achieved by Bio-Gel P-30 gel filtration chromatography in 1 M acetic acid. TGF activities were demonstrated in the Mr ranges of 30,000 and 6,000-10,000. These competed with epidermal growth factor (EGF) for binding to A431 membrane receptors and induced anchorage-independent growth of untransformed fibroblasts. The low Mr TGF activity obtained from P-30 chromatography was purified to apparent homogeneity by two sequential reverse-phase high performance liquid chromatography steps with a mu Bondapak C18 column first using a linear gradient of acetonitrile going from 0-60% in 120 min and then by rechromatography of the activity over the same column using a shallower gradient of acetonitrile going from 20-40% in 160 min. The isoelectric point of the melanoma patient-derived urinary TGF was determined to be 6.2, which is distinct from that for human EGF. Amino acid composition analysis of the purified urinary TGF (uTGF) revealed that it is composed of at least 42 amino acid residues with a minimum estimated Mr of 4,545. Compositional analysis further revealed distinct similarities and differences between the uTGF, human EGF and TGFs secreted by various transformed human and rodent cell lines.

    Topics: Amino Acids; Animals; Binding, Competitive; Carcinoma, Squamous Cell; Cell Line; Cell Membrane; Cell Transformation, Neoplastic; Epidermal Growth Factor; ErbB Receptors; Humans; Kidney; Kinetics; Melanoma; Molecular Weight; Neoplasm Proteins; Peptides; Rats; Receptors, Cell Surface; Transforming Growth Factors

1985
Ectopic peptides released by a human melanoma cell line that modulate the transformed phenotype.
    Proceedings of the National Academy of Sciences of the United States of America, 1985, Volume: 82, Issue:15

    Cells derived from a human melanoma strain by low-serum selection in monolayer were found to be capable of growth in semisolid medium, forming colonies ranging from tight to loose or dispersed. These phenotypes were found to be stable after cloning and retesting. Examination of the biological activities of ectopic peptides produced by a clone that gives rise to loose dispersed colonies revealed the presence of a transforming growth factor beta (TGF-beta; apparent Mr 14,700) and a single TGF-alpha species produced at an unusually high concentration (2.2 ng/ml). Coeluted with the TGF-alpha at approximately 22,500 was a mitogenic activity and a previously undescribed activity capable of modulating the phenotype of induced normal rat kidney fibroblast (NRK-49F) colonies in semisolid medium. This activity can modulate the usual tight colony to express a loose or dispersed phenotype characteristic of the producer cells. Both the possible role these ectopic peptides play in the expression of the transformed phenotype by the tumor cells producing them and the possible correlation between the Mr 22,500 epidermal growth factor-like peptide released by this particular tumor line and high molecular weight epidermal growth factor-like peptides found in the urine of cancer patients are discussed.

    Topics: Animals; Cell Line; Cell Transformation, Neoplastic; Epidermal Growth Factor; ErbB Receptors; Humans; Kidney; Melanoma; Molecular Weight; Neoplasm Proteins; Peptides; Radioligand Assay; Rats; Receptors, Cell Surface; Transforming Growth Factors

1985
Two distinct mechanisms involving growth factors employed in subversion of growth regulation by oncogenes.
    Progress in medical virology. Fortschritte der medizinischen Virusforschung. Progres en virologie medicale, 1985, Volume: 32

    Topics: Amino Acid Sequence; Animals; Cell Transformation, Neoplastic; Cell Transformation, Viral; Epidermal Growth Factor; ErbB Receptors; Growth Substances; Humans; Oncogenes; Platelet-Derived Growth Factor; Receptors, Cell Surface

1985
Growth factors and cancer.
    The Australian and New Zealand journal of surgery, 1985, Volume: 55, Issue:2

    Recent advances in protein chemistry and genetic engineering have revealed new information about the molecular lesions involved in the induction and maintenance of cancer cells. It is now known that a single base change in the DNA of human cells leads to cancer. The normal pathway of proliferation and differentiation is perturbed by changes to molecules involved in the intracellular biochemical pathways controlled by growth factors. Some cancer cells appear to produce their own growth factor, others have higher concentrations of growth factor receptors on their surface and others have mutated versions of the intracellular proteins linked to the growth factor receptors. This increased understanding of growth control in normal and neoplastic cell populations is gradually providing a foundation for new approaches to cancer therapy.

    Topics: Animals; Cell Line; Cell Transformation, Neoplastic; Cell Transformation, Viral; DNA, Neoplasm; Epidermal Growth Factor; Growth Substances; Hematopoietic Stem Cells; Humans; Neoplasms; Neoplastic Stem Cells; Nerve Growth Factors; Platelet-Derived Growth Factor; Receptors, Cell Surface

1985
Loss of epidermal growth factor receptors and release of transforming growth factors do not correlate with sarcoma virus-transformation in clonally-related NIH/3T3-derived cell lines.
    The Biochemical journal, 1985, Jul-01, Volume: 229, Issue:1

    Transformation of NIH/3T3 cells by Kirsten murine sarcoma virus (MSV) caused a dramatic reduction in the number of cell-surface receptors for epidermal growth factor (EGF). However, the number of EGF receptors remained at a very low level in a non-tumourigenic revertant cell line isolated from the virus-transformed cells, indicating that an increase in EGF receptors is not a requirement for the phenotypic reversion of Kirsten MSV-transformed 3T3 cells. Serum-free conditioned medium from normal and virus-transformed cell lines contained similar amounts of cell growth-promoting activity as assayed by the ability to stimulate DNA synthesis in quiescent Swiss 3T3 cell cultures. However, the concentrated conditioned medium from these cell lines showed no evidence of beta-transforming growth factor (TGF) activity as assayed by promotion of anchorage-independent growth of untransformed normal rat kidney (NRK) fibroblasts in agarose. The cellular release of alpha-TGF activity was assayed by measuring the ability of concentrated conditioned medium to inhibit the binding of 125I-EGF to Swiss 3T3 cells. Conditioned medium protein from the virus-transformed cell line inhibited 125I-EGF binding but only to the same extent as conditioned medium protein prepared from the untransformed cell line. The alpha-TGF secretion by these cell lines was estimated to be 30-45-fold lower than the level of alpha-TGF released by a well-characterized alpha-TGF-producing cell line (3B11). These results suggest that the induction of TGF release is not a necessary event in the transformation of NIH/3T3 cells by Kirsten MSV.

    Topics: Animals; Cell Line; Cell Transformation, Neoplastic; Culture Media; DNA; Epidermal Growth Factor; ErbB Receptors; Fibroblasts; Kirsten murine sarcoma virus; Mice; Neoplasm Proteins; Peptides; Receptors, Cell Surface; Sarcoma, Experimental; Transforming Growth Factors

1985
Inositol phospholipid and phosphatidic acid metabolism in response to membrane receptor activation.
    The Proceedings of the Nutrition Society, 1985, Volume: 44, Issue:2

    Topics: Animals; Calcium; Cell Division; Cell Membrane; Cell Transformation, Neoplastic; Epidermal Growth Factor; Humans; Hydrolysis; Models, Chemical; Phosphatidic Acids; Phosphatidylinositol Phosphates; Phosphatidylinositols; Rats; Receptors, Cell Surface; Receptors, Cholinergic

1985
The action of oncogenes in the cytoplasm and nucleus.
    Science (New York, N.Y.), 1985, Nov-15, Volume: 230, Issue:4727

    As many as 40 distinct oncogenes of viral and cellular origin have been identified to date. Many of these genes can be grouped into functional classes on the basis of their effects on cellular phenotype. These groupings suggest a small number of mechanisms of action of the oncogene-encoded proteins. Some data suggest that, in the cytoplasm, these proteins may regulate levels of critical second messenger molecules; in the nucleus, these proteins may modulate the activity of the cell's transcriptional machinery. Many of the gene products can also be related to a signaling pathway that determines the cell's response to growth-stimulating factors. Because some of these genes are expressed in nongrowing, differentiated cells, the encoded proteins may in certain tissues mediate functions that are unrelated to cellular growth control.

    Topics: Animals; Birds; Cell Nucleus; Cell Transformation, Neoplastic; Chickens; Cytoplasm; Deltaretrovirus; DNA Tumor Viruses; Drosophila; Epidermal Growth Factor; Growth Substances; Guanosine Triphosphate; Humans; Mutation; Neoplasms; Oncogenes; Platelet-Derived Growth Factor; Polyomavirus; Proto-Oncogenes; Rats; Repetitive Sequences, Nucleic Acid; Retroviridae; Simian virus 40; Transcription, Genetic

1985
Ectopic peptides released by a human melanoma cell line that modulate the transformed phenotype.
    Ciba Foundation symposium, 1985, Volume: 116

    Cells derived from a human melanoma strain by low serum selection in monolayer were found to be capable of growth in semi-solid medium, forming colonies ranging from tight, to loose, or dispersed. These phenotypes were found to be stable on cloning and retesting. Examination of the serum-free media conditioned by these clones indicates that the clones release activities capable of inducing agar colonies, in an indicator cell line (NRK, 49F), that express phenotypes similar to those of the melanoma clones used to produce the serum-free conditioned media (SF-CM). These SF-CM contained a transforming growth factor (TGF) beta (apparent Mr 14 700) and a single, apparently high Mr TGF-alpha species. Co-eluting with the TGF-alpha at an Mr of approximately 22 500 was a previously undescribed activity capable of modulating the phenotype of the NRK agar colonies induced by the combination of TFGs alpha and beta. This new activity can modulate the usual tight colony to express a loose or dispersed phenotype characteristic of the producer cells. The possible role these ectopic peptides play in the expression of the transformed phenotype by the tumour cells producing them, and the possible correlation between the 22 500 Mr EGF-like peptide (TGF-alpha) released by this particular tumour line and high Mr EGF-like peptides found in the urine of cancer patients, are both discussed.

    Topics: Animals; Cell Line; Cell Transformation, Neoplastic; Culture Media; Epidermal Growth Factor; ErbB Receptors; Humans; Melanoma; Molecular Weight; Peptides; Phenotype; Radioligand Assay; Rats; Receptors, Cell Surface; Transforming Growth Factors

1985
Selection of transformed cells in serum-free media.
    In vitro cellular & developmental biology : journal of the Tissue Culture Association, 1985, Volume: 21, Issue:12

    NIH3T3 cells grow in a serum-free basal nutrient medium supplemented with fibronectin, transferrin, insulin, epidermal growth factor (EGF) and high density lipoprotein (HDL). The individual omission from the serum-free medium of insulin, EGF, or HDL results in greatly reduced cell growth. These growth-restrictive conditions can be used to select for cells transformed with SV40, the polyomavirus middle T antigen gene, the activated human ras gene, and the mouse c-myc gene.

    Topics: Animals; Antigens, Polyomavirus Transforming; Antigens, Viral, Tumor; Blood; Cell Division; Cell Line; Cell Transformation, Neoplastic; Cell Transformation, Viral; Culture Media; Epidermal Growth Factor; Fibronectins; Insulin; Lipoproteins, HDL; Mice; Mice, Inbred C3H; Oncogene Proteins, Viral; Oncogenes; Simian virus 40; Transferrin

1985
Transforming growth factors from a human tumor cell: characterization of transforming growth factor beta and identification of high molecular weight transforming growth factor alpha.
    Biochemistry, 1985, Oct-08, Volume: 24, Issue:21

    Intracellular transforming growth factors (TGFs) were extracted from a human rhabdomyosarcoma cell line and purified to apparent homogeneity by using gel filtration, cation-exchange, and high-performance liquid chromatography. Two types of transforming growth factor activities, TGF-alpha and TGF-beta, were detected. The intracellular polypeptides which belonged to the TGF-alpha family required TGF-beta for full activity in inducing nonneoplastic normal rat kidney fibroblasts to grow in soft agar. These peptides also bound to the membrane receptor for epidermal growth factor. As determined by sodium dodecyl sulfate-polyacrylamide gels, the apparent molecular weight of these intracellular TGF-alpha's was 18 000. Intracellular TGF-beta required either epidermal growth factor or TGF-alpha for stimulation of soft agar growth. The intracellular TGF-beta was purified to homogeneity as judged by a single peak after reverse-phase high-performance liquid chromatography and a single band on a sodium dodecyl sulfate-polyacrylamide gel. The intracellular TGF-beta from the human tumor cell line was similar in all respects tested (migration on sodium dodecyl sulfate-polyacrylamide gels, stimulation of soft agar growth, binding to the membrane receptor for TGF-beta, and amino acid composition) to intracellular TGF-beta from normal human placenta.

    Topics: Amino Acids; Animals; Binding, Competitive; Cell Line; Cell Transformation, Neoplastic; Chromatography, High Pressure Liquid; Chromatography, Ion Exchange; Epidermal Growth Factor; ErbB Receptors; Female; Humans; Kidney; Kinetics; Molecular Weight; Peptides; Placenta; Pregnancy; Rats; Receptors, Cell Surface; Rhabdomyosarcoma; Transforming Growth Factors

1985
Effect of retinoic acid on the late-stage promotion of transformation in JB6 mouse epidermal cells in culture.
    Cancer research, 1985, Volume: 45, Issue:5

    beta-All-trans-retinoic acid (RA) inhibited the anchorage-independent growth of JB6 cells induced by either mezerein or alpha-epidermal growth factor (alpha-EGF) (a purified fraction of epidermal growth factor). The inhibition was dose dependent for alpha-EGF as well as for RA. Mezerein-induced growth in soft agar was inhibited to a greater extent by RA than was alpha-EGF-induced growth in soft agar, at similar colony yields. The extent of inhibition of anchorage-dependent growth induced by RA was similar for nontransformed JB6 cells and for alpha-EGF-transformed cells, so that transformation was shown not to influence the sensitivity of cells to retinoid inhibition of anchorage-dependent growth. RA was as effective at inhibiting anchorage-independent growth when it was applied after promoter-induced transformation as when it was applied during promoter-induced transformation. Therefore, the antiproliferative effect of RA, without an additional antitransformation effect, was sufficient to account for the reduced colony yield. These results suggest that the antipromoting action of retinoids in JB6 cells may occur by limiting proliferation, the regulation of which may be coupled with the state of differentiation of cells.

    Topics: Animals; Cell Division; Cell Transformation, Neoplastic; Cells, Cultured; Diterpenes; Epidermal Growth Factor; Epidermis; Mice; Skin Neoplasms; Terpenes; Tretinoin

1985
Selective phosphorylation of cytosol proteins associated with transformation and restoration of normal phenotype in AKR mouse embryo fibroblasts.
    Cancer research, 1985, Volume: 45, Issue:5

    Phosphoproteins from cytosol preparations of methylcholanthrene-transformed AKR mouse (AKR-MCA) cells were compared to those of their untransformed counterparts, AKR-2B cells, by two-dimensional electrophoresis following an in vitro 32P phosphorylation procedure using endogenous kinases and substrates. Five proteins were phosphorylated in the AKR-MCA cells which were not observed in the AKR-2B cells, while six proteins were phosphorylated in the untransformed cells which were not observed in the malignant cells. Treatment of AKR-MCA cells with 1% N,N-dimethylformamide induced the reversion of the malignant cells to a phenotype similar to that of untransformed AKR-2B cells (S. Chakrabarty et al., Cancer Res., 44: 2181, 1984). Treatment of AKR-MCA cells with dimethyl formamide resulted in the restoration of five of the AKR-2B-associated phosphorylations and abolished 2 of the AKR-MCA-associated phosphorylations. AKR-2B cells have been shown to respond to transforming growth factors with reversible phenotypic transformation (R. F. Tucker et al., Cancer Res., 43: 1581, 1983). Transforming growth factor treatment of AKR-2B cells induced all five of the AKR-MCA-associated phosphoproteins and the loss of all six of the AKR-2B phosphoproteins. Epidermal growth factor treatment of AKR-2B cells resulted in the phosphorylation of several proteins which were not observed in either AKR-MCA or untreated AKR-2B cells. Some, but not all, of the AKR-2B-associated phosphorylations were also observed in epidermal growth factor-treated cells. The results of these studies demonstrated qualitative and/or quantitative changes in cytosolic protein kinase-phosphatase activities between transformed and normal AKR-2B cells. Treatment of AKR-MCA cells with dimethylformamide resulted in the restoration of some of the normal AKR-2B cell-associated protein kinase-phosphatase activities.

    Topics: Animals; Cell Transformation, Neoplastic; Cells, Cultured; Cytosol; Dimethylformamide; Embryo, Mammalian; Epidermal Growth Factor; Fibroblasts; Methylcholanthrene; Mice; Mice, Inbred AKR; Peptides; Phenotype; Phosphoproteins; Phosphorylation; Protein Kinases; Proteins; Transforming Growth Factors

1985
Transforming growth factors induce markers of neoplasia in cultured adult rat bladder.
    Carcinogenesis, 1985, Volume: 6, Issue:4

    Transforming growth factors alpha and beta (TGF-alpha and TGF-beta) isolated from normal mouse kidney induced gross morphological changes in rat urothelial cells maintained in organ culture. These morphological effects are similar to those observed after long-term treatment of rat bladder organ cultures with the carcinogen N-methyl-N-nitrosourea (MNU) or the promoting agents sodium saccharin and sodium cyclamate. Cultures were treated continuously with 5-25 micrograms/ml of Bio-Gel P-30-purified TGF containing both TGF-alpha and TGF-beta between days 1 and 14 in culture, or with 5 micrograms/ml from days 28 to 42. Controls received 1-10 ng/ml epidermal growth factor (EGF) or control medium. Untreated controls retained a normal urothelium throughout the period of study. Mature superficial-type cells covered most of the surface and less mature forms appeared on the cut sides and damaged areas where cells followed the normal pattern of urothelial differentiation. EGF at 5 and 10 ng/ml caused necrosis of the entire urothelium but at 1 and 2 ng/ml had minimal effects on histology and scanning electron microscopical appearance up to 14 days in culture. Crude P-30-purified TGFs induced a series of dose-related changes from 4 days, which were maximal at 8 days and persisted or decreased between 8 and 14 days. These included hyperplasia, loss of epithelial polarity, hyperchromasia and elongation of basal cells between the overlying cell layers to reach the culture surface. Scanning electron microscopy showed the appearance at the culture surface of immature cells with gross surface abnormalities including large numbers of blebs, stubby microvilli and long pleomorphic microvilli. Immature cells on the sides of the culture and in damaged areas developed similar features. At crude TGF doses of 10 micrograms/ml many superficial cells were rounded, some became cystic and epithelial necrosis was observed. Cultures treated with h.p.l.c.-purified TGF-beta at 80 ng/ml in the presence of 2 ng/ml EGF showed similar effects to those treated with 5 micrograms/ml P-30-purified TGF. Fully differentiated cultures treated from 28 to 42 days with crude TGF, showed changes similar to those seen in early cultures. However, histological changes, particularly basal cell elongation were more widespread and there was an abnormal development of globular processes between the membrane ridges of mature superficial cells. Neither crude TGF nor EGF stimulated growth in soft agar of isolated e

    Topics: Animals; Cell Transformation, Neoplastic; Epidermal Growth Factor; Male; Methylnitrosourea; Mice; Microscopy, Electron, Scanning; Organ Culture Techniques; Peptides; Rats; Rats, Inbred F344; Transforming Growth Factors; Urinary Bladder

1985
Gradation of carcinogen-induced capacity for anchorage-independent growth in cultured rat liver epithelial cells.
    Cancer research, 1985, Volume: 45, Issue:9

    The effect of epidermal growth factor (EGF) on the capacity for anchorage-independent growth of chemically treated rat hepatic epithelial cells has been investigated. We have performed the studies using 16 clonally derived cell strains which represented single cell-derived subpopulations of a heterogeneous rat hepatic epithelial cell line that had been tumorigenically transformed by 11 repeated treatments with N-methyl-N'-nitro-N-nitrosoguanidine. The results can be summarized as follows. (a) Secondary clonal subpopulations isolated from the colonies formed by these strains in soft agar subsequently and invariably acquired markedly enhanced colony-forming efficiencies as compared to their parental strains. (b) EGF could enhance or induce colony-forming ability in soft agar in all of these cell strains. (c) The magnitudes of enhancement of the colony-forming efficiencies by EGF in soft agar could not be correlated with the absolute EGF-binding capacity of these cell strains. (d) The enhancement or induction of the colony-forming ability by EGF was either reversible or irreversible, partially correlating with the expression of gamma-glutamyl transpeptidase activity by the strains. These findings indicate that the cellular capacity of liver epithelial cells to grow anchorage independently in soft agar medium can be graded according to the pattern of response of EGF induction of colony-forming ability. These grades may reflect the level of neoplastic transformation of these cells. Moreover during the multistep transformation of rat hepatic epithelial cells by a chemical carcinogen, EGF can be used to reveal the presence of altered cells which have acquired partial capacity for the anchor-age-independent growth property. This property may constitute an additional identifiable early step of the neoplastic transformation of these cells.

    Topics: Animals; Cell Transformation, Neoplastic; Cells, Cultured; Epidermal Growth Factor; Liver; Liver Neoplasms, Experimental; Methylnitronitrosoguanidine; Rats

1985
Interaction of epidermal growth factor with initiators and complete carcinogens in the C3H10T1/2 cell culture system.
    Journal of cellular biochemistry, 1985, Volume: 28, Issue:1

    Unlike 12-O-tetradecanoylphorbol-13-acetate, epidermal growth factor (EGF) could not promote the appearance of type III foci from initiated C3H10T1/2 cells. At appropriate concentrations, EGF induced the formation of type II colonies in the absence of any initiator. At higher concentrations, EGF suppressed the induction of both type II and type III colonies elicited by methylcholanthrene.

    Topics: Animals; Carcinogens; Cell Transformation, Neoplastic; Cells, Cultured; Cocarcinogenesis; Epidermal Growth Factor; Fibroblasts; Methylcholanthrene; Mice; Mice, Inbred C3H; Tetradecanoylphorbol Acetate; Tumor Stem Cell Assay

1985
Growth factor production during multistage transformation of epithelium in vitro. I. Partial purification and characterisation of the factor(s) from a fully transformed epithelial cell line.
    Journal of cellular physiology, 1985, Volume: 125, Issue:1

    Transforming growth factor (TGF)-like activity is characterised from one of a series of salivary epithelial cell lines, CSG 211, chemically transformed in vitro. In this transformation system, we can demonstrate multiple stages in the acquisition of a malignant phenotype by normal diploid ductal epithelial cells from male mouse submandibular gland. The fully transformed, tumorigenic cell TGF-like activity in serum-free supernatants resembles no other well-characterised growth factor and has an apparent molecular weight (Mr) of 14 kd. There is also evidence of a higher Mr activity, which is separable by anion exchange chromatography. We show that the premalignant, nontumorigenic progenitor cells of this line do not produce demonstrable TGF-like activity and that this property is therefore acquired as CSG 211 cells become carcinoma producing.

    Topics: Animals; Cell Line; Cell Transformation, Neoplastic; Chromatography, Gel; Chromatography, Ion Exchange; DNA Replication; Epidermal Growth Factor; Epithelial Cells; Fibroblasts; Lectins; Male; Mice; Microscopy, Phase-Contrast; Molecular Weight; Peptide Biosynthesis; Plant Lectins; Rabbits; Rats; Salivary Glands; Time Factors; Transforming Growth Factors

1985
Interrelation between cellular rRNA content and regulation of the cell cycle of normal and transformed mouse cell lines.
    Cell biology international reports, 1985, Volume: 9, Issue:11

    The relation between cellular rRNA content, as a measure of cell size, and the regulation of the cell cycle has been investigated for swiss 3T3 and the spontaneously transformed swiss 3T6 cell line. It is shown that the characteristic of percent of quiescent cells stimulated into the cell cycle versus cellular rRNA content is basically different for 3T3 and 3T6 cells: 3T3 cells do not enter the cell cycle below a certain threshold of cellular rRNA content, whereas 3T6 cells start proliferation without a substantial increase of rRNA. These data are interpreted as consistent with transformation of 3T6 cells being in essence their uncoupling from the requirement of normal cells of passing over a threshold of cellular rRNA content (cell size) before initiating DNA-replication.

    Topics: Animals; Blood; Cattle; Cell Cycle; Cell Division; Cell Transformation, Neoplastic; Cells, Cultured; Culture Media; Epidermal Growth Factor; Insulin; Kinetics; Mice; RNA, Ribosomal

1985
Human transforming growth factor-alpha: precursor structure and expression in E. coli.
    Cell, 1984, Volume: 38, Issue:1

    Transforming growth factor-alpha (TGF-alpha) is secreted by many human tumors and can induce the reversible transformation of nontransformed cell lines. Using long synthetic deoxyoligonucleotides as hybridization probes we isolated an exon coding for a portion of TGF-alpha from a human genomic DNA library. Utilizing this exon as a probe, a cell line derived from a human renal cell carcinoma was identified as a source of TGF-alpha mRNA. A cloned TGF-alpha cDNA was isolated from a cDNA library prepared using RNA from this cell line, and was found to encode a precursor polypeptide of 160 amino acids. The 50 amino acid mature TGF-alpha produced by expression of the appropriate coding sequence in E. coli binds to the epidermal growth factor receptor and induces the anchorage independence of normal mammalian cells in culture.

    Topics: Adenocarcinoma; Amino Acid Sequence; Base Sequence; Cell Line; Cell Transformation, Neoplastic; Cloning, Molecular; DNA; Epidermal Growth Factor; ErbB Receptors; Escherichia coli; Genes; Humans; Kidney Neoplasms; Neoplasm Proteins; Nucleic Acid Hybridization; Peptides; Plasmids; Receptors, Cell Surface; RNA, Messenger; Transforming Growth Factors

1984
Latent beta-transforming growth factor in nontransformed and Kirsten sarcoma virus-transformed normal rat kidney cells, clone 49F.
    Cancer research, 1984, Volume: 44, Issue:12 Pt 1

    Normal rat kidney cells of the clone 49F and their Ki-MSV-transformed counterparts spontaneously release the same transforming growth factor (TGF) activity in an inactive form. By acidification followed by neutralization prior to assay, this TGF activity is unmasked and promotes anchorage-independent growth of the NRK-49F indicator cells in the presence of epidermal growth factor. The TGF activity released by both cell types has an apparent molecular weight of 9,000 under acidic conditions, does not compete for binding to epidermal growth factor receptors, is heat resistant but dithiothreitol and trypsin sensitive, and therefore is of the beta-TGF class.

    Topics: Animals; Cell Line; Cell Transformation, Neoplastic; Clone Cells; Epidermal Growth Factor; ErbB Receptors; Kidney; Kirsten murine sarcoma virus; Molecular Weight; Peptides; Rats; Receptors, Cell Surface; Sarcoma Viruses, Murine; Transforming Growth Factors

1984
Production of somatomedin-like activity by human adult tumor-derived, transformed, and normal cell cultures and by cultured rat hepatocytes: effects of culture conditions and of epidermal growth factor (urogastrone).
    Canadian journal of biochemistry and cell biology = Revue canadienne de biochimie et biologie cellulaire, 1984, Volume: 62, Issue:12

    We have measured the production of a basic-somatomedin-like activity (SLA) by a variety of human tumor-derived, transformed, and normal postnatal cell cultures; and we have compared the production of SLA by these cell types with the production of SLA by adult rat hepatocytes cultured in serum-free medium. Cells derived from a human epidermoid carcinoma (KB), a pancreatic carcinoma (Panc-1), a Simian virus 40 transformed adult human skin-derived cell line (SV40 fibroblasts), and a normal adult human skin-derived fibroblast line released SLA when cultured in a serum-free growth medium. No SLA was recovered from the culture medium of human choriocarcinoma-derived cells (BeWo) or of a human lymphoblastoid cell line (IM-9). The production of SLA by rat hepatocytes cultured in serum-free medium appeared to exceed the production of SLA by the other cell cultures. In cultures of KB cells, SV40 fibroblasts, and rat hepatocytes, the production of SLA depended on the frequency with which the growth medium was renewed; in general, the highest rates of SLA production were observed when the medium was renewed every 48-72 h. The presence of mouse epidermal growth factor (urogastrone) (EGF-URO) in the serum-free culture medium stimulated the production of SLA by KB cells and by rat hepatocytes, but did not increase SLA production by normal or by SV40-transformed human skin-derived fibroblasts. We conclude that tumor-derived cells are capable of producing somatomedin-like activity and that the production of SLA by such cells can be subject to controls (nutrient availability, EGF-URO stimulation) that regulate SLA production, either by normal adult tissues, like liver, or by a variety of normal embryonic tissues.

    Topics: Animals; Cell Line; Cell Transformation, Neoplastic; Cell Transformation, Viral; Cells, Cultured; Epidermal Growth Factor; Humans; Liver; Mice; Neoplasms; Simian virus 40; Somatomedins

1984
Immunohistochemical demonstration of epidermal growth factor and nerve growth factor in experimental carcinogenesis in the mouse submandibular gland.
    Virchows Archiv. B, Cell pathology including molecular pathology, 1984, Volume: 45, Issue:4

    Immunohistochemical demonstration of epidermal growth factor (EGF) and nerve growth factor (NGF) was made during chemical carcinogenesis in the mouse submandibular gland. The granular convoluted tubule cells in the normal male submandibular gland contained larger amounts of EGF and NGF than in the female. The initial phase and early stages in chemical carcinogenesis showed degranulation of the granular convoluted tubule cells with a marked decrease in EGF and NGF. Premalignant lesions such as duct-like structures and multicystic lesions showed variable staining for EGF and were usually negative for NGF. Material secreted into the luminal spaces revealed increased staining for EGF and NGF. Scattered tumor cells of the poorly differentiated squamous-cell carcinoma type and desquamated tumor cells contained abundant EGF, but not NGF. No positive reaction for EGF or NGF was found in the induced squamous-cell carcinoma cells.

    Topics: Animals; Carcinoma, Squamous Cell; Cell Transformation, Neoplastic; Cytoplasmic Granules; Epidermal Growth Factor; Female; Male; Mice; Nerve Growth Factors; Salivary Gland Neoplasms; Submandibular Gland Neoplasms

1984
The proteins of oncogenes.
    Scientific American, 1984, Volume: 251, Issue:2

    Topics: Animals; Annexins; Cell Transformation, Neoplastic; Cell Transformation, Viral; DNA; Epidermal Growth Factor; ErbB Receptors; Guanosine Triphosphate; Humans; Membrane Proteins; Neoplasm Proteins; Nucleoproteins; Oncogene Protein pp60(v-src); Oncogenes; Phosphotyrosine; Platelet-Derived Growth Factor; Protein Binding; Protein Biosynthesis; Protein Kinases; Receptors, Cell Surface; Retroviridae; Tyrosine; Viral Proteins

1984
Rat transforming growth factor type 1: structure and relation to epidermal growth factor.
    Science (New York, N.Y.), 1984, Mar-09, Volume: 223, Issue:4640

    The complete amino acid sequence of rat transforming growth factor type 1 has been determined. This growth factor, obtained from retrovirus-transformed fibroblasts, is structurally and functionally related to mouse epidermal growth factor and human urogastrone. Production of this polypeptide by various neoplastic cells might contribute to the continued expression of the transformed phenotype.

    Topics: Amino Acid Sequence; Animals; Cell Line; Cell Transformation, Neoplastic; DNA; Epidermal Growth Factor; ErbB Receptors; Humans; Idoxuridine; Mice; Peptide Biosynthesis; Peptides; Rats; Receptors, Cell Surface; Structure-Activity Relationship; Transforming Growth Factors

1984
Nonfunctional epidermal growth factor receptor in cells transformed by Kirsten sarcoma virus.
    Biochemical and biophysical research communications, 1984, Jan-30, Volume: 118, Issue:2

    The cell membrane receptor for epidermal growth factor (EGF) appears to be a glycoprotein of Mr 170,000 and mediates the mitogenic and metabolic responses of cells with EGF receptors (EGF-R). Normal rat kidney (NRK) have about 3 X 10(5) EGF-R per cell. Upon transformation of NRK cells by Kirsten sarcoma virus, the transformed derivative (KNRK) loses the ability to bind 125I-EGF. Membranes from NRK and KNRK cells were included in EGF-dependent phosphorylation reactions to search for evidence of the EGF-R. A phosphorylated protein of Mr 170,000 was detected in both NRK and KNRK membranes. The Mr 170,000 protein was identified to be EGF-R by immunoprecipitation with monoclonal antibody to the receptor. Furthermore, two-dimensional peptide mapping using trypsin and chymotrypsin digestions of the iodinated receptors from both NRK and KNRK cells showed essentially identical patterns. These data indicate that the EGF-R is present in KNRK cells with apparently the same protein structure as the NRK counterpart.

    Topics: Animals; Cell Line; Cell Membrane; Cell Transformation, Neoplastic; Epidermal Growth Factor; ErbB Receptors; Kidney; Kinetics; Kirsten murine sarcoma virus; Molecular Weight; Ornithine Decarboxylase; Peptide Fragments; Rats; Receptors, Cell Surface; Sarcoma Viruses, Murine

1984
Neuroblastoma cells produce transforming growth factors during exponential growth in a defined hormone-free medium.
    Proceedings of the National Academy of Sciences of the United States of America, 1984, Volume: 81, Issue:13

    Mouse neuroblastoma Neuro-2A cells have been cultured in a chemically defined serum-free medium consisting of a 1:1 mixture of Dulbecco's modified Eagle's medium and Ham's F-12 medium, supplemented with 30 nM selenite and 10 micrograms of transferrin per ml. In this medium, which does not contain any externally added polypeptide growth factor, cells proliferate rapidly with a doubling time of approximately equal to 10 hr. During exponential growth in this serum-free medium, Neuro-2A cells secrete a 15- to 20-kDa transforming growth factor with strong mitogenic action and the ability to induce anchorage-independent growth on nontransformed cells. This neuroblastoma-derived transforming growth factor (ND-TGF) is acid and heat stable but is sensitive to treatment with trypsin or dithiothreitol. However, it does not compete with epidermal growth factor (EGF) for receptor binding and does not require EGF receptors for its mitogenic activity. Experiments on the effects of EGF on ND-TGF-induced soft agar growth of normal rat kidney cells indicate that Neuro-2A cells secrete an EGF-potentiated TGF in addition to ND-TGF. It is suggested that Neuro-2A cells can proliferate in the absence of externally added growth factors as a result of autocrine production of polypeptide growth factors.

    Topics: Animals; Cell Division; Cell Line; Cell Transformation, Neoplastic; Cells, Cultured; Culture Media; DNA Replication; Epidermal Growth Factor; ErbB Receptors; Kinetics; Mice; Neoplasm Proteins; Neuroblastoma; Peptide Biosynthesis; Peptides; Receptors, Cell Surface; Transforming Growth Factors

1984
Presence of transforming growth factors in human breast cancer cells.
    Cancer research, 1984, Volume: 44, Issue:9

    Conditioned medium (CM) from a human mammary carcinoma cell line, MCF-7, and ten individual clones derived from these cells was examined for the presence of transforming growth factors (TGFs). Concentrated CM from all of the MCF-7 cell lines was found to stimulate the anchorage-independent growth of normal rat kidney cells in soft agar and to inhibit the binding of epidermal growth factor (EGF) to mouse NIH/3T3 fibroblasts and to A431 human epidermoid carcinoma cell membranes. The soft agar stimulating activity was heat stable but sensitive to treatment with dithiothreitol. EGF receptors were measured on the MCF-7 cell lines to determine whether the amount of TGFs associated with the CM from the various cell lines was correlated with the level of EGF receptors being expressed on these cells. Moreover, the intrinsic cloning efficiency of these lines in soft agar was measured to ascertain if any correlation might exist between the level of TGFs associated with these cells and the ability of these cell lines to form colonies in soft agar. Although all the MCF-7 cell lines had approximately the same number of EGF receptors per cell, ranging from 3 to 6 X 10(3) sites/cell, CM from these lines varied in potency with respect to inducing the growth of normal rat kidney cells as colonies in soft agar and in inhibiting the binding of EGF to NIH/3T3 cells. Likewise, the level of TGFs associated with the CM from the various clones showed no correlation with the ability of these individual lines to grow as colonies in soft agar. TGF activity was also detected in acid-ethanol extracts prepared from MCF-7 cells propagated in nude mice as tumors and in the extracts from two transplantable human mammary adenocarcinomas, Clouser I and II. In addition, approximately 50% of the normal rat kidney colonies formed in response to the Clouser II tumor extracts exhibited a branching morphology in contrast to spherical colonies produced by Clouser I or MCF-7 extracts. These results demonstrate that human mammary carcinoma cells from both established cell lines and cells maintained in nude mice as tumors contain TGF-like activities. Furthermore, the variation in TGFs associated with the CM from the MCF-7 clones suggests that the parent MCF-7 cell line contains a heterogeneous population of cells.

    Topics: Animals; Breast Neoplasms; Cell Cycle; Cell Line; Cell Transformation, Neoplastic; Cells, Cultured; Epidermal Growth Factor; ErbB Receptors; Female; Humans; Kidney; Kinetics; Mice; Mice, Nude; Neoplasm Transplantation; Peptides; Rats; Receptors, Cell Surface; Transforming Growth Factors; Transplantation, Heterologous

1984
Neoplastic human fibroblast proteins are related to epidermal growth factor precursor.
    Proceedings of the National Academy of Sciences of the United States of America, 1984, Volume: 81, Issue:17

    We report the amino acid composition of two polypeptides, p788 and p789. These polypeptides are reliable markers for neoplastic transformation in human fibroblasts. Their compositions are unusually rich in cysteine and serine. Because the recently reported amino acid sequence of mouse epidermal growth factor precursor (prepro-EGF) is also rich in those two amino acids and because the role of p788 and p789 as markers for neoplastic transformation is consistent with the fact that epidermal growth factor has been shown to play some role in transformation, we investigated the hypothesis that p788 and p789 are related to prepro-EGF. We compared the amino acid composition of p788 with that of all possible interior domains of prepro-EGF of appropriate length. We found that the composition of p788 is remarkably similar to that of residues 630-880 of prepro-EGF. The similarity is sufficiently strong to support the conclusion that it reflects amino acid sequence homology.

    Topics: Amino Acid Sequence; Animals; Cell Line; Cell Transformation, Neoplastic; Epidermal Growth Factor; Fibroblasts; Humans; Mice; Neoplasm Proteins; Protein Precursors; Species Specificity; Structure-Activity Relationship

1984
Effects of nickel sulfate on growth and differentiation of normal human bronchial epithelial cells.
    Carcinogenesis, 1984, Volume: 5, Issue:12

    Epidemiological studies have shown that inhalation of nickel compounds enhances the risk of human respiratory cancer. Cultures of normal human bronchial epithelial cells were continuously exposed to a dose (5-20 micrograms/ml) of NiSO4 that reduced their colony forming efficiency by 30-80%. After 40 days of incubation, the cultures consisted of large, squamous cells; mitotic cells were not observed. The cells were then maintained in medium without NiSO4. After 40-75 total days of incubation, colonies of mitotic cells appeared at a rate of 1 colony per 100 000 cells originally at risk; no colonies appeared in control cultures or in cultures exposed to less than 5 micrograms NiSO4/ml for 90 days. Twelve NiSO4-altered cell cultures isolated from five experiments have been expanded into mass cultures. Most of the cell lines have an increased population doubling potential (greater than 50 divisions). Some exhibit aberrations in the squamous (terminal) differentiation process whereas others have lost the requirement for epidermal growth factor for clonal growth. Aneuploidy and marker chromosomes have also been noted. However, none of these NiSO4-altered cell cultures are anchorage independent nor do they produce tumors upon injection into athymic nude mice.

    Topics: Bronchi; Cell Division; Cell Transformation, Neoplastic; Dose-Response Relationship, Drug; Epidermal Growth Factor; Epithelium; Humans; In Vitro Techniques; Nickel; Tetradecanoylphorbol Acetate

1984
Acquisition of a limited lifespan by differentiating cells derived from PC13 embryonal carcinoma cells.
    Journal of cell science, 1984, Volume: 72

    Retinoic acid (RA) has previously been shown to induce the differentiation of mouse embryonal carcinoma (EC) cells to endoderm-like cells that have a slower rate of proliferation and are nontumorigenic. These cells also acquire the ability to respond to a range of exogenous growth factors. We have analysed the change in growth phenotype for PC13 EC cells using video recordings and autoradiography. We have shown that the endoderm-like cells have a longer cell cycle time than their undifferentiated counterparts (five cell divisions after exposure to RA the differentiated cells had a median cell cycle time of 1800 min compared to 800 min for control cells). The endoderm-like cells also have a progressively decreasing probability of dividing again and this indicates that the differentiation process is accompanied by the acquisition of a limited life-span. The characteristics of mortal cells are well documented, and the endoderm-like cells demonstrate the properties of such cells. In addition, we have confirmed the observation that epidermal growth factor (EGF) can stimulate the proliferation of the endoderm-like cells and have shown, using autoradiography, that 92% of these cells express EGF receptors. Using video recordings, we have demonstrated that the effect of EGF is to shorten the cell cycle of the differentiating cells. We have also shown that EGF can enhance the survival of the endoderm-like cells and thereby prolong their life-span. It is known that EGF and other growth factors can prolong the life-span of mortal cells derived from normal tissues, but we have demonstrated that EGF can have this effect on the differentiated derivatives of a tumour cell.

    Topics: Animals; Cell Cycle; Cell Division; Cell Survival; Cell Transformation, Neoplastic; Clone Cells; Culture Media; Epidermal Growth Factor; Mice; Neoplasms, Experimental; Teratoma; Tretinoin; Video Recording

1984
Growth factors: mechanism of action and relation to oncogenes.
    Cell, 1984, Volume: 37, Issue:1

    Topics: Animals; Cell Transformation, Neoplastic; Epidermal Growth Factor; Growth Substances; Humans; Insulin; Mitosis; Neoplasms; Oncogenes; Platelet-Derived Growth Factor; Protein Kinases; Protein-Tyrosine Kinases

1984
Alteration of the chemotactic response of NIH/3T3 cells to PDGF by growth factors, transformation, and tumor promoters.
    Cell, 1984, Volume: 36, Issue:2

    The platelet-derived growth factor (PDGF) is a potent chemoattractant for cells that respond to PDGF as a mitogen. The chemotactic response of these cells to PDGF is inversely related to their rate of proliferation, with quiescent cells exhibiting a 25-fold greater chemotactic response than exponentially growing cells. Factors that stimulate the growth of quiescent cells (EGF, FGF, PDGF, and serum) decrease the cells' migratory response to PDGF but not to fibronectin, suggesting that the decreased migration is not due to a general paralysis of cell motility. Transformed lines of NIH/3T3 cells lose their ability to respond to PDGF as a chemoattractant but can still migrate in response to fibronectin. Similarly, after treatment of 3T3 cells with the tumor-promoter phorbol myristate acetate, which induces a transformation-like phenotype, the cells no longer respond to PDGF as a chemoattractant but retain their migratory response to fibronectin. Thus it appears that the growth state of the cells can alter their migratory response to PDGF. These data suggest that growth factors, transformation, and tumor promoters specifically alter the cells' ability to respond to the PDGF-mediated chemotactic signal. It appears that both transformation and tumor promoters accomplish this by altering PDGF-binding to the cell surface.

    Topics: Animals; Cell Transformation, Neoplastic; Cells, Cultured; Chemotaxis; Epidermal Growth Factor; Epithelium; Fibroblast Growth Factors; Fibroblasts; Growth Substances; Kirsten murine sarcoma virus; Mice; Phorbols; Platelet-Derived Growth Factor; Tetradecanoylphorbol Acetate

1984
Reduced trisialoganglioside synthesis in chemically but not mos-transformed mouse epidermal cells.
    Cancer research, 1984, Volume: 44, Issue:4

    A specific decrease in the net de novo synthesis ([1-14C]-glucosamine incorporation) of cell surface trisialoganglioside (GT) occurs in preneoplastic mouse JB6 epidermal cells in response to tumor-promoting phorbol esters, mezerein, or epidermal growth factor, all of which promote neoplastic transformation in JB6 cells, but not in response to the bladder promoter sodium cyclamate, a nonpromoter in JB6 cells. The ganglioside showing elevated synthesis after mezerein or epidermal growth factor exposure is monosialoganglioside 1, whereas disialoganglioside 1b synthesis is elevated after phorbol ester exposure. Primary mouse epidermal cells and putatively initiated epidermal cell lines selected for their resistance to induction of terminal differentiation by high calcium are resistant to promotion of anchorage-independent transformation by 2-week exposure to 12-O-tetradecanoylphorbol-13-acetate. In both cell types, little or no decrease in GT synthesis occurs in response to short-term 12-O-tetradecanoylphorbol-13-acetate exposure, thus extending further our previous observation that this GT response is restricted to promotable cells. A decreased synthesis of GT also occurs consistently in cell lines transformed by 12-O-tetradecanoylphorbol-13-acetate or N-methyl-N-nitro-nitrosoguanidine as compared with their nontransformed counterparts but not in cell lines transformed by a cloned integrated murine sarcoma provirus containing the oncogenic sequence v-mos. Thus, reduced cell surface GT synthesis may be important both in the induction and in the maintenance of the chemically transformed but not viral oncogene mos-transformed phenotype in mouse epidermal cells.

    Topics: Animals; Carbon Radioisotopes; Carcinogens; Cell Line; Cell Transformation, Neoplastic; Diterpenes; Epidermal Growth Factor; Gangliosides; Glucosamine; Mice; Phorbol Esters; Phorbols; Skin; Terpenes; Tetradecanoylphorbol Acetate

1984
Restoration of normal growth control and membrane antigen composition in malignant cells by N,N-dimethylformamide.
    Cancer research, 1984, Volume: 44, Issue:5

    The effects of the differentiation agent, N,N-dimethylformamide (DMF), on malignant AKR-MCA cells were studied. The properties of DMF-treated AKR-MCA cells were compared to those of the normal parental AKR-2B mouse embryo fibroblasts. AKR-MCA cells grown in 1% DMF were found to be more similar to their normal counterparts than to untreated AKR-MCA cells by several criteria. These criteria included the loss of the transformed morphology, a 2-fold reduction of doubling time, a 10-fold reduction of saturation density, and the complete loss of the ability to grow with anchorage independence. The expression of high-molecular-weight membrane antigens (Mr 110,000 to 450,000), which was found to be greatly reduced in AKR-MCA cells in comparison to normal AKR-2B cells, was restored by treatment of AKR-MCA cells with DMF. The expression of a low-molecular-weight AKR-MCA cell-associated membrane antigen, on the other hand was found to be suppressed. Studies on the mitogenic response of these cells indicated that AKR-MCA and AKR-2B cells may be regulated by different types of growth control. Growth-arrested AKR-MCA cells did not respond to epidermal growth factor, but responded to nutrient replenishment. AKR-2B cells, on the other hand, responded to epidermal growth factor, but did not respond to nutrient replenishment. Treatment of AKR-MCA cells with DMF restored their ability to respond to epidermal growth factor, while their ability to respond to nutrient replenishment was lost. The results of this study indicated that DMF treatment induced the normalization of malignant AKR-MCA cells with regard to membrane antigen composition and growth control properties.

    Topics: Animals; Antigens, Neoplasm; Cell Division; Cell Membrane; Cell Transformation, Neoplastic; Cells, Cultured; Dimethylformamide; Embryo, Mammalian; Epidermal Growth Factor; Fibroblasts; Methylcholanthrene; Mice; Mice, Inbred AKR; Neoplasms, Experimental

1984
[The effect of growth factors on cell cultures].
    Acta histochemica. Supplementband, 1983, Volume: 27

    As sources for cell growth factors with polypeptide nature sera, organs and conditioned culture media can be used. A few samples of such factors are listed in this report. Their main effects on cultured cells consist in the stimulation of DNA-synthesis and proliferation. The knowledge on the mechanism of action of these factors is still incomplete. Some aspects of this question are discussed. The development of chemically defined culture media will be a fundamental prerequisite for further progress in the elucidation of the problems of growth regulation on the cellular level. Certain discrepancies between the effects of the growth factors in vivo and in vitro might arise by the fact that the cells in culture are not provided by adequate substrata when growing on glass or plastic and thus having a cell shape not typical for the in vivo situation. The cell shape may have an influence on the mitogenic response of the cells.

    Topics: Alpha-Globulins; Animals; Cell Division; Cell Transformation, Neoplastic; Cells, Cultured; Endocytosis; Epidermal Growth Factor; Fibroblast Growth Factors; Fibroblasts; Growth Substances; Insulin; Mice; Nerve Growth Factors; Peptides; Platelet-Derived Growth Factor; Somatomedins

1983
A subpopulation of cultured human keratinocytes which is resistant to the induction of terminal differentiation-related changes by phorbol, 12-myristate, 13-acetate: evidence for an increase in the resistant population following transformation.
    Carcinogenesis, 1983, Volume: 4, Issue:7

    We have studied the action of phorbol, 12-myristate, 13-acetate (PMA) on human keratinocytes grown with lethally-irradiated 3T3 cells using medium supplemented with hydrocortisone, cholera toxin and epidermal growth factor. Normal keratinocyte cultures show a heterogeneous response to PMA; 90-93% of the colony-forming cells lose their colony-forming ability and form cornified envelopes when treated for 24 h with doses of 100 nM or less, but the remainder are resistant to doses of 1000 nM. The resistant cells are the precursors of the sensitive ones and heterogeneity is restored to those cells and their progeny after 8 days culture in the absence of PMA. Cultures of 3 squamous cell carcinoma lines, a SV40-transformed human keratinocyte line, and three clones of these lines were found to contain 3-17 times more PMA-resistant keratinocytes than the normal strains, and the size of the PMA-resistant fraction in each line was inversely related to the competence of that line to lose colony-forming efficiency when placed in suspension culture (which is the first detectable change in an ordered programme of events resembling terminal differentiation of the keratinocyte). The number of cells with cornified envelopes in surface cultures of normal human keratinocytes increased from approximately 3% in control cultures to approximately 70% in those treated for 6 days with 100 nM PMA. The transformed human keratinocyte cultures showed a 3-25-fold smaller increase in cornified envelope formation when treated with 100 nM PMA, and the increase in envelope formation by each line when exposed to this dose of PMA was related to the competence of that line to lose cloning efficiency in suspension culture. No relationship was found between the ability of any human keratinocyte strain or line we studied to metabolically inactivate PMA and their resulting response to the compound. The results are discussed in relation to the mechanism of action of PMA as a promoter of epidermal carcinogenesis.

    Topics: Animals; Carcinoma, Squamous Cell; Cell Differentiation; Cell Line; Cell Transformation, Neoplastic; Cells, Cultured; Child; Cholera Toxin; Epidermal Growth Factor; Female; Fetus; Humans; Hydrocortisone; Keratins; Male; Mice; Phorbols; Pregnancy; Simian virus 40; Skin; Skin Physiological Phenomena; Tetradecanoylphorbol Acetate

1983
Cancer as a problem in intercellular communication: regulation by growth-inhibiting factors (Chalones).
    Progress in nucleic acid research and molecular biology, 1983, Volume: 29

    Topics: Animals; Cell Communication; Cell Differentiation; Cell Transformation, Neoplastic; Epidermal Growth Factor; Growth Inhibitors; Humans; Neoplasms

1983
BALB and Kirsten murine sarcoma viruses alter growth and differentiation of EGF-dependent balb/c mouse epidermal keratinocyte lines.
    Cell, 1983, Volume: 32, Issue:2

    Clonal BALB/c mouse epidermal keratinocyte (BALB/MK) cell lines were established in tissue culture. Despite their aneuploid nature, the lines were nontumorigenic, and retained in vitro properties similar to those of primary diploid keratinocytes. These included the constitutive expression of keratin and terminal differentiation in response to a calcium concentration greater than 1.0 mM in the medium. The cells also demonstrated an absolute requirement for nanomolar concentrations of epidermal growth factor (EGF) for their proliferation. BALB or Kirsten murine sarcoma viruses are acute transforming retroviruses, which have been shown to transform fibroblastic and hematopoietic cells. Infection of BALB/MK or its clonal sublines with either virus leads to the rapid acquisition of EGF-independent growth. The cells concomitantly lose their sensitivity to calcium-induced terminal differentiation. Thus these retroviruses can rapidly confer upon epithelial keratinocytes in culture growth properties that resemble those of malignant epidermoid carcinoma cells.

    Topics: Animals; Calcium; Cell Differentiation; Cell Division; Cell Line; Cell Transformation, Neoplastic; Cell Transformation, Viral; Clone Cells; Epidermal Cells; Epidermal Growth Factor; Kirsten murine sarcoma virus; Mice; Mice, Inbred BALB C; Sarcoma Viruses, Murine

1983
Embryonal carcinoma cell growth and differentiation. Production of and response to molecules with transforming growth factor activity.
    Experimental cell research, 1983, Volume: 143, Issue:1

    Transforming growth factors are known to induce anchorage-independent growth of non-transformed cells, and are released by a variety of cells, including MSV-transformed cells. This study demonstrates that the differentiated cells derived from F9 and PC-13 embryonal carcinoma cells, but not the parental cells themselves, respond by increased growth to several factors released by MSV-transformed cells, including partially purified sarcoma growth factor. The chemical properties of the growth-promoting activity are shown to match the chemical properties of the transforming growth factors released by MSV-transformed cells. Furthermore, F9 and PC-13 embryonal carcinoma cells, which do not respond to factors released by MSV-transformed cells, are shown to release factors with transforming growth factor activity. Based on the close relationship between mouse embryonal carcinoma cells and cells of early mouse embryos, it is suggested that molecules with transforming growth factor activity may play a role during the early stages of mammalian development.

    Topics: Animals; Binding, Competitive; Cell Differentiation; Cell Division; Cell Line; Cell Transformation, Neoplastic; Epidermal Growth Factor; ErbB Receptors; Mice; Neoplasms; Peptides; Receptors, Cell Surface; Transforming Growth Factors

1983
Comparison of intra- and extracellular transforming growth factors from nontransformed and chemically transformed mouse embryo cells.
    Cancer research, 1983, Volume: 43, Issue:4

    Multiple transforming growth factors (TGFs) have been isolated from the serum-free conditioned media and acid-ethanol cell extracts of nontransformed AKR-2B and chemically transformed AKR-MCA mouse cells. The TGF activity was analyzed using Bio-Gel P-60 chromatography in 1 M acetic acid and tested for colony-stimulating activity in soft agar using AKR-2B, AKR-2B (clone 84A), NRK, and AKR-MCA cells as indicators. Both intracellular and extracellular TGF activity from AKR-MCA and AKR-2B cells show a major peak of AKR-2B and epidermal growth factor-potentiated NRK colony-stimulating activity that coelute in the 13,000 +/- 2,000 molecular weight region. In the 24,000 +/- 7,000 molecular weight range, AKR-MCA cells produce intracellular and extracellular NRK colony-stimulating activity that is not potentiated by epidermal growth factor, while the intracellular and extracellular NRK colony-stimulating activity produced by AKR-2B cells requires added epidermal growth factor for colony formation. Also important in determining the growth and morphological characteristics of a cell line, besides the production of a TGF, is the ability of a cell to respond to TGF activity. We have shown that the transformed AKR-MCA cells form more colonies than AKR-2B cells in response to certain TGF activities. This suggests that the increased responsiveness of AKR-MCA cells to TGFs may be important in determining its phenotype.

    Topics: Animals; Biological Assay; Cell Line; Cell Transformation, Neoplastic; Embryo, Mammalian; Epidermal Growth Factor; ErbB Receptors; Mice; Mice, Inbred AKR; Molecular Weight; Peptides; Receptors, Cell Surface; Transforming Growth Factors

1983
Teleocidin is not mitogenic for 12-O-tetradecanoylphorbol-13-acetate non-proliferative variants of 3T3 cells.
    Carcinogenesis, 1983, Volume: 4, Issue:4

    Teleocidin and 12-O-tetradecanoylphorbol-acetate (TPA), two potent tumor promoters, are also excellent mitogens for 3T3 cells. I now demonstrate that teleocidin is unable to stimulate mitogenesis in TPA non-proliferative 3T3 variants that retain TPA receptors.

    Topics: Alkaloids; Animals; Carcinogens; Cell Division; Cell Survival; Cell Transformation, Neoplastic; Cells, Cultured; Epidermal Growth Factor; ErbB Receptors; Genetic Variation; Lyngbya Toxins; Mice; Phorbols; Receptors, Cell Surface; Tetradecanoylphorbol Acetate

1983
Transforming growth factors from neoplastic and nonneoplastic tissues.
    Federation proceedings, 1983, Volume: 42, Issue:9

    Transforming growth factors (TGFs) are a heterogeneous family of polypeptides that induce anchorage-independent growth in nonneoplastic anchorage-dependent cells. They have been found in many tissues, both neoplastic and nonneoplastic. All TGFs isolated thus far are of low molecular weight (6000-25,000), are acid and heat stable, and are inactivated by reagents that reduce disulfide bonds. TGFs have been classified as type alpha or type beta based on their interactions with the receptor for epidermal growth factor (EGF) and their requirement for EGF (or an EGF-like polypeptide) for functional activity. TGF-alpha and TGF-beta act synergistically. TGF-alpha induces phosphorylation of tyrosine in the EGF receptor. TGF-beta, isolated from bovine sources, accelerates experimental wound healing in rats.

    Topics: Animals; Cell Adhesion; Cell Division; Cell Transformation, Neoplastic; Cell Transformation, Viral; Drug Stability; Epidermal Growth Factor; ErbB Receptors; Humans; Mice; Molecular Weight; Neoplasms; Neoplasms, Experimental; Peptides; Rats; Receptors, Cell Surface; Transforming Growth Factors

1983
Phosphorus-31 nuclear magnetic resonance analysis of epidermal growth factor action in A-431 human epidermoid carcinoma cells and SV-40 virus transformed mouse fibroblasts.
    Biochemistry, 1983, Apr-26, Volume: 22, Issue:9

    Topics: Adenosine Diphosphate; Adenosine Triphosphate; Animals; Carcinoma, Squamous Cell; Cell Line; Cell Transformation, Neoplastic; Epidermal Growth Factor; Fibroblasts; Humans; Kinetics; Magnetic Resonance Spectroscopy; Mice; NAD; Simian virus 40

1983
Transforming growth factors produced by retrovirus-transformed rodent fibroblasts and human melanoma cells: amino acid sequence homology with epidermal growth factor.
    Proceedings of the National Academy of Sciences of the United States of America, 1983, Volume: 80, Issue:15

    Transforming growth factors (TGFs) were purified from serum-free medium conditioned by retrovirus-transformed Fisher rat embryo fibroblasts, mouse 3T3 cells, and two human melanoma cell lines. The purification of each TGF was monitored in a radioreceptor assay based on receptor crossreactivity with mouse submaxillary gland epidermal growth factor (mEGF) and was achieved by gel permeation chromatography of the acid-soluble TGF-containing activity, followed by reverse-phase high-pressure liquid chromatography with sequential use of acetonitrile and 1-propanol in the presence of aqueous trifluoroacetic acid. The amino-terminal sequences of rat, mouse, and human TGFs were determined. Extensive sequence homology was found among TGF polypeptides from different species and cell types. Alignment of the amino acid sequences of rat TGF, mEGF, and human urogastrone (hEGF) reveals statistically significant sequence homology. The reported results suggest that TGFs that compete for binding to the cellular EGF receptor and EGF may have evolved from a common progenitor.

    Topics: Amino Acid Sequence; Animals; Cell Line; Cell Transformation, Neoplastic; Cells, Cultured; Embryo, Mammalian; Epidermal Growth Factor; Fibroblasts; Genes; Humans; Melanoma; Mice; Moloney murine leukemia virus; Neoplasm Metastasis; Peptides; Rats; Rats, Inbred F344; Retroviridae; Sarcoma Viruses, Feline; Transforming Growth Factors

1983
Isolation and partial purification of a new class of transforming growth factors from an avian sarcoma virus-transformed rat cell line.
    Cancer research, 1983, Volume: 43, Issue:12 Pt 1

    Transformation of rat cells by avian sarcoma viruses induced the release of growth factors into serum-free conditioned medium. An avian sarcoma virus-transformed rat cell line, 77N1, produced and released a polypeptide growth factor, classified as a transforming growth factor (TGF), which transiently promotes anchorage-dependent BALB3T3 A31 cells to form progressively growing colonies in soft agar. The TGF was isolated and partially purified from an extract of 77N1 cells by ion-exchange chromatography on a diethylaminoethyl Sephacel column followed by ammonium sulfate precipitation. The TGF was assumed to have a molecular weight of 11,000 from gel filtration on Sephadex G-50. This TGF did not compete with epidermal growth factor for binding to cell membrane receptors and was not potentiated by epidermal growth factor. The TGF was trypsin and dithiothreitol sensitive as well as heat and acid labile, indicating that it was different from previously reported TGFs of similar molecular weight and thus belonged to a new class of TGFs.

    Topics: Animals; Avian Sarcoma Viruses; Biological Assay; Cell Line; Cell Transformation, Neoplastic; Clone Cells; DNA Replication; Epidermal Growth Factor; ErbB Receptors; Kidney; Peptides; Rats; Receptors, Cell Surface; Transforming Growth Factors

1983
Bromine residue at hydrophilic region influences biological activity of aplysiatoxin, a tumor promoter.
    Science (New York, N.Y.), 1983, Dec-16, Volume: 222, Issue:4629

    Aplysiatoxin and debromoaplysiatoxin, which are isolated from the seaweed, Lyngbya gracilis, differ in their chemical structure only by the presence or absence of a bromine residue in the hydrophilic region. The function and the structure-activity relation of the hydrophilic region are not known. Aplysiatoxin increased malignant transformation, stimulated DNA synthesis, and inhibited the binding of phorbol-12,13-dibutyrate and epidermal growth factor to cell receptors. Debromoaplysiatoxin inhibited the binding of these two substances as strongly as aplysiatoxin but did not increase malignant transformation or stimulate DNA synthesis. These results indicate that a slight change in the chemical structure of the hydrophilic region of aplysiatoxin affects its abilities to increase cell transformation and stimulate DNA synthesis and that the abilities of the tumor promoters to inhibit the binding of phorbol-12,13-dibutyrate and epidermal growth factor are dissociable from their abilities to increase cell transformation and stimulate DNA synthesis under some circumstances.

    Topics: Animals; Caenorhabditis elegans Proteins; Carcinogens; Carrier Proteins; Cell Line; Cell Transformation, Neoplastic; Chemical Phenomena; Chemistry; DNA; Epidermal Growth Factor; ErbB Receptors; Lactones; Lyngbya Toxins; Mice; Phorbol 12,13-Dibutyrate; Phorbol Esters; Protein Kinase C; Receptors, Cell Surface; Receptors, Drug; Structure-Activity Relationship

1983
High-molecular-weight transforming growth factor activity in the urine of patients with disseminated cancer.
    Cancer research, 1983, Volume: 43, Issue:1

    Urine from 22 patients with a variety of disseminated cancers and from an equivalent number of nonmalignant controls of similar age and sex was tested for the presence of transforming growth factor (TGF) activity as measured by the ability to promote the growth in soft agar of nontransformed indicator cells. Cancer patients included those with carcinomas of the lung, breast, colon, and ovary, as well as melanomas and sarcomas. The nonmalignant controls included both normals and individuals with a variety of inflammatory and infectious disorders. Aliquots of unfrozen urine were acid extracted, chromatographed on a Bio-Gel P-30 column, and then tested for TGF activity using normal rat kidney fibroblasts and epidermal growth factor (EGF)-competing activity with human carcinoma A431 cells. These assays revealed that a high-molecular-weight TGF activity (Mr 30,000 to 35,000) which coelutes with EGF-competing activity was present in 18 of 22 cancer patients but present in only five of 22 nonmalignant controls (p less than 0.01). In contrast, a low-molecular-weight TGF activity (Mr 6000 to 8000) which does not coelute with EGF-competing activity was found in all urines tested. These results indicate that an EGF-related, high-molecular-weight TGF activity is found in the urine of cancer patients and may be a useful tumor marker. Unlike other tumor markers described previously, high-molecular-weight TGF activity has a biological activity which is related to the expression of the transformed phenotype.

    Topics: Binding, Competitive; Carcinoma; Cell Transformation, Neoplastic; Chromatography, Gel; Epidermal Growth Factor; Humans; Molecular Weight; Neoplasms; Peptides; Transforming Growth Factors

1983
Transforming growth factor-beta in human platelets. Identification of a major storage site, purification, and characterization.
    The Journal of biological chemistry, 1983, Jun-10, Volume: 258, Issue:11

    Acidic ethanol extracts of human platelets induced non-neoplastic normal rat kidney fibroblasts to undergo anchorage-independent growth. Less than 100 ng/ml of the crude extract elicits 50% of the maximal biological response when assayed in the presence of epidermal growth factor (2.5 ng/ml). In the absence of epidermal growth factor, the potency of the extract decreased 1,000-fold. These results show that platelets contain a type beta transforming growth factor. The specific activity of the platelet extract is 100-fold greater than that of other non-neoplastic tissues. The growth factor was purified to homogeneity by sequential gel filtration on Bio-Gel P-60 columns, first in the absence and then in the presence of urea. As determined by sodium dodecyl sulfate-polyacrylamide gel electrophoresis, this transforming growth factor-beta is a protein of 25,000 daltons. It is composed of two 12,500-dalton subunits held together by disulfide bonds. These results, as well as its amino acid composition and its lack of strong mitogenic activity, show that this protein is distinct from platelet-derived growth factor. When completely purified, transforming growth factor-beta elicits 50% of its maximal biological response at concentrations less than 5 x 10(-12) M.

    Topics: Amino Acids; Animals; Biological Assay; Blood Platelets; Cell Line; Cell Transformation, Neoplastic; Epidermal Growth Factor; Humans; Kidney; Macromolecular Substances; Peptides; Rats; Transforming Growth Factors

1983
Epidermal growth factor-like transforming growth factor. I. Isolation, chemical characterization, and potentiation by other transforming factors from feline sarcoma virus-transformed rat cells.
    The Journal of biological chemistry, 1983, Nov-25, Volume: 258, Issue:22

    An acid-stable transforming growth factor (TGF) that interacts with epidermal growth factor (EGF) receptors and is structurally related to EGF was isolated from serum-free culture fluids of Snyder-Theilen feline sarcoma virus-transformed rat embryo (FeSV-Fre) cells. Purification of this EGF-like TGF (eTGF) was achieved by molecular filtration chromatography and successive reverse-phase high pressure liquid chromatography steps on octadecyl support eluted with acetonitrile and 1-propanol gradients, respectively. Rat eTGF consists of a 7.4-kD single polypeptide chain that co-migrates with biological activity in dodecyl sulfate-polyacrylamide electrophoresis gels. Like preparations of a related TGF from human melanoma cells (Marquardt, H., and Todaro, G.J. (1982) J. Biol. Chem. 257, 5220-5225), but unlike EGF from rat, human, or mouse, rat eTGF has phenylalanine and lacks methionine. However, the sequence of the first 30 amino acid residues in rat eTGF is H2N-Val-Val-Ser-His-Phe-Asn-Lys-Cys-Pro-Asp-Ser-His-Thr-Gln-Tyr-Cys-Phe-His-Gly - Thr-(x)-Arg-Phe-Leu-Val-Gln-Glu-Glu-(Lys)-(Lys)-, which is significantly (20% and 28%) homologous to the NH2-terminal region of mouse EGF and human EGF, respectively. In addition to eTGF, molecular filtration chromatography of acid-soluble extracts from medium conditioned by FeSV-Fre cells resolved a 14-kD transforming factor(s) apparently devoid of intrinsic mitogenic activity but able to elicit a strong anchorage-independent growth response in the presence of eTGF or EGF. These results show that: 1) a 7.4-kDa TGF structurally and functionally related to EGF has been isolated from FeSV-Fre cells and 2) the full anchorage-independent growth-promoting activity of medium conditioned by FeSV-Fre cells is due to the coordinate action of at least two types of factors, the 7.4-kDa eTGF and a second 14-kDa transforming factor(s).

    Topics: Amino Acid Sequence; Animals; Cell Division; Cell Line; Cell Transformation, Neoplastic; Cells, Cultured; Embryo, Mammalian; Epidermal Growth Factor; Kidney; Oncogenic Viruses; Peptides; Rats; Rats, Inbred F344; Transforming Growth Factors

1983
Fibroblast growth factor stimulates anchorage independent growth in agar of BHK21/13 and SV3T3 cells.
    Cell biology international reports, 1982, Volume: 6, Issue:5

    Topics: Animals; Blood; Cell Adhesion; Cell Division; Cell Line; Cell Transformation, Neoplastic; Cell Transformation, Viral; Clone Cells; Cricetinae; Culture Media; Epidermal Growth Factor; Fibroblast Growth Factors; Insulin; Kidney; Peptides

1982
Effects of 12-O-tetradecanoylphorbol-13-acetate on fibroblasts from individuals genetically predisposed to cancer.
    Cancer research, 1982, Volume: 42, Issue:9

    The purpose of the present study was to determine whether skin fibroblasts from individuals, either with an inherited predisposition to cancer or with genetic disorders usually associated with a high risk of cancer, can be oncogenically transformed in vitro by a tumor promoter alone. The effects of chronic and limited applications of 12-O-tetradecanoylphorbol-13-acetate (TPA) on several properties that are associated with transformation were examined using skin fibroblasts from individuals with polyposis coli, a familial cancer syndrome, xeroderma pigmentosum, Fanconi's anemia, and trisomy 21. The results of this study show that TPA treatment induces similar changes on cellular morphology, growth rate, saturation density, epidermal growth factor binding, and cytoskeleton in fibroblasts from both normal and genetically predisposed individuals. None of these cell lines, however, acquired anchorage-independent growth or unlimited growth potential in culture after chronic application of TPA. These observations suggest clearly that skin fibroblasts from individuals with either a genetic predisposition to or a high risk of cancer may not exist in a preneoplastic or "initiated" state susceptible to oncogenic transformation by TPA alone and that the mechanism of genetically determined cancer induction may be different from that of chemical carcinogenesis.

    Topics: Cell Adhesion; Cell Division; Cell Line; Cell Transformation, Neoplastic; Cytoskeleton; Disease Susceptibility; Down Syndrome; Epidermal Growth Factor; Fanconi Anemia; Female; Fibroblasts; Humans; Intestinal Polyps; Male; Neoplasms; Phorbols; Tetradecanoylphorbol Acetate; Trypsin; Xeroderma Pigmentosum

1982
Isolation from murine sarcoma cells of novel transforming growth factors potentiated by EGF.
    Nature, 1982, Feb-04, Volume: 295, Issue:5848

    Topics: Animals; Cell Division; Cell Transformation, Neoplastic; Drug Synergism; Epidermal Growth Factor; ErbB Receptors; Growth Substances; Mice; Receptors, Cell Surface; Sarcoma, Experimental

1982
Human transforming growth factor. Production by a melanoma cell line, purification, and initial characterization.
    The Journal of biological chemistry, 1982, May-10, Volume: 257, Issue:9

    A low molecular weight human transforming growth factor (hTGFs) was isolated from serum-free medium conditioned by a human metastatic melanoma tumor line, A2058. The purification of hTGFs was achieved by gel permeation chromatography of the acid-soluble growth-promoting activity on Bio-Gel P-10 in 1 M acetic acid, followed by reverse phase high pressure liquid chromatography on muBondapak C18 support using a linear gradient of acetonitrile in 0.045% trifluoroacetic acid, and subsequently by rechromatography of the human transforming growth factor-containing fractions with a linear 1-propanol gradient in 0.035% trifluoroacetic acid on the same column. The estimated molecular weight of hTGFs is 7400. It is a single chain polypeptide with three intrachain disulfide bridges and no free sulfhydryl groups. Lacking tyrosine and methionine, but containing three phenylalanine residues, hTGFs is unlike human and mouse epidermal growth factor (EGF). hTGFs competes with 125I-labeled EGF for binding to A431 human carcinoma cells completely and equivalently, and thus is functionally related to EGF. In contrast, hTGFs enabled normal anchorage-dependent rat kidney cells to grow in soft agar, whereas EGF did not stimulate growth of these cells in semisolid medium. The half-maximal growth-stimulating response of hTGFs was reached with one EGF-competing unit or 1.1 ng of hTGFs.

    Topics: Amino Acids; Cell Line; Cell Transformation, Neoplastic; Epidermal Growth Factor; ErbB Receptors; Humans; Kinetics; Melanoma; Peptide Biosynthesis; Peptides; Receptors, Cell Surface; Transforming Growth Factors

1982
A new tumor-promoting agent, dihydroteleocidin B, markedly enhances chemically induced malignant cell transformation.
    Science (New York, N.Y.), 1982, Apr-30, Volume: 216, Issue:4545

    Teleocidin, which was isolated from mycelia of Streptomyces, is a potent tumor promoter in mouse skin. The catalytically hydrogenated compound dihydroteleocidin B markedly enhanced malignant cell transformation induced by 3-methylcholanthrene or ultraviolet radiation. Dihydroteleocidin B was at least 100 times more effective in enhancing transformation than 12-O-tetradecanoyl phorbol-13-acetate, the strongest promoter known until now, whereas both promoters showed equal capacities to induce early membrane effects and DNA synthesis.

    Topics: Alkaloids; Animals; Carcinogens; Cell Division; Cell Transformation, Neoplastic; Cells, Cultured; Epidermal Growth Factor; ErbB Receptors; Lyngbya Toxins; Methylcholanthrene; Mice; Receptors, Cell Surface

1982
Autocrine secretion of peptide growth factors by tumor cells.
    National Cancer Institute monograph, 1982, Volume: 60

    Transforming growth factors (TGF) are growth-promoting polypeptides that cause phenotypic transformation and anchorage-independent growth of normal cells. They have been isolated from several human and animal carcinoma and sarcoma cells. One TGF is sarcoma growth factor (SGF) which is released by murine sarcoma virus-transformed cells. Whereas the TGF interacts with epidermal growth factor (EGF) cell membrane receptors, it is not detectable in culture fluids from cells which contain high numbers of free EGF cell membrane receptors. The SGF acts as a tumor promoter in cell culture systems, and its effect on the transformed phenotype is blocked by retinoids (vitamin A and synthetic analogs). The production of TGF by transformed cells and the responses of normal cells to the addition of TGF to the culture medium raise the possibility that cells "autostimulate" their growth by releasing factors that rebind at the cell surface. The term "autocrine secretion" has been proposed for this type of situation in which a cell secretes a hormone-like substance for which it has external cell membrane receptors. The autocrine concept may provide a partial explanation for some aspects of tumor cell progression.

    Topics: Animals; Binding Sites; Carcinogens; Cell Transformation, Neoplastic; Cell Transformation, Viral; Cells, Cultured; Epidermal Growth Factor; ErbB Receptors; Growth Substances; Humans; In Vitro Techniques; Neoplasms; Peptides; Receptors, Cell Surface; Sarcoma Viruses, Murine; Transforming Growth Factors

1982
Synergistic interaction of two classes of transforming growth factors from murine sarcoma cells.
    Cancer research, 1982, Volume: 42, Issue:11

    Transforming growth factors (TGFs) isolated from murine sarcoma virus-transformed 3T3 cells have been separated by high-pressure liquid chromatography into two subsets. One subset, called TGF alpha, competes with epidermal growth factor (EGF) for receptor sites, whereas the other, called TGF beta, does not. TGB beta, purified by high-pressure liquid chromatography, will not induce formation of large colonies of cells in soft agar in the absence of TGF alpha or EGF. However, the combined action of either TGF alpha or EGF (which by themselves are relatively ineffective in promoting growth of cells in soft agar) together with TGF beta results in a potent synergistic effect, with formation of large colonies. Chemically modified analogs of EGF also potentiate TGF beta activity to the extent that they bind to the EGF receptor. It is suggested that TGF beta may be an important mediator of the known effects of both TGF alpha and EGF on neoplastic transformation.

    Topics: Animals; Binding, Competitive; Cell Transformation, Neoplastic; Epidermal Growth Factor; ErbB Receptors; Kinetics; Mice; Peptides; Receptors, Cell Surface; Sarcoma Viruses, Murine; Sarcoma, Experimental; Transforming Growth Factors

1982
Affinity labeling of a transforming growth factor receptor that does not interact with epidermal growth factor.
    Proceedings of the National Academy of Sciences of the United States of America, 1982, Volume: 79, Issue:22

    Membrane components that interact with epidermal growth factor (EGF) and transforming growth factors (TGFs) have been identified by covalent crosslinking to their respective 125I-labeled ligands. Under appropriate conditions, disuccinimidyl suberate or hydroxysuccinimidyl p-azidobenzoate cross-link receptor-bound 125I-labeled EGF to a 140- to 170-kilodalton (kDal) receptor species in membranes from both A431 human carcinoma cells and normal rat kidney cells. 125I-Labeled sarcoma growth factor (SGF), a TGF from virally transformed mouse 3T3 cells, also can be affinity-crosslinked to the 140- to 170-kDal EGF receptor species in membranes from A431 and rat kidney cells. The labeling of this receptor is inhibited when either excess unlabeled EGF or SGF is present during incubation of membranes with either 125I-labeled EGF or 125I-labeled SGF. In contrast, a second receptor species of 60 kDal is affinity-labeled with 125I-labeled SGF but not with 125I-labeled EGF in membranes from both A431 and rat kidney cells. SGF and a TGF from virally transformed rat embryo cells inhibit the labeling of the 60-kDal species when present in excess during incubation of membranes with 125I-labeled SGF, whereas EGF is completely ineffective in inhibiting the labeling of this receptor. The data suggest that a specific 60-kDal receptor that displays high affinity for TGFs but not for EGF may mediate induction of the transformed phenotype. In addition, SGF and other TGFs interact with the 140- to 170-kDal EGF receptor that appears to mediate normal cell growth effects.

    Topics: Affinity Labels; Animals; Carcinoma; Cell Line; Cell Transformation, Neoplastic; Epidermal Growth Factor; ErbB Receptors; Humans; Kidney; Peptides; Rats; Receptors, Cell Surface; Transforming Growth Factors

1982
Promotion of x-ray transformation in vitro.
    Carcinogenesis; a comprehensive survey, 1982, Volume: 7

    Topics: Animals; Anti-Inflammatory Agents; Cell Count; Cell Division; Cell Line; Cell Transformation, Neoplastic; Cocarcinogenesis; DNA Repair; Epidermal Growth Factor; Mice; Mitomycins; Mutation; Phorbols; Radiation, Ionizing; Tetradecanoylphorbol Acetate; X-Rays

1982
Studies on antagonistic actions of TPA and retinoic acid.
    Carcinogenesis; a comprehensive survey, 1982, Volume: 7

    Topics: Animals; Cell Adhesion; Cell Division; Cell Line; Cell Transformation, Neoplastic; Clone Cells; Contact Inhibition; Epidermal Growth Factor; Mice; Phorbols; Tetradecanoylphorbol Acetate; Tretinoin

1982
Cell cycle variation in 125I-labeled epidermal growth factor binding in chemically transformed cells.
    Cancer research, 1982, Volume: 42, Issue:7

    Previous studies have shown that the nontransformed AKR-2B cells when arrested in the G1 phase of the cell cycle due to low-molecule-weight nutrient (amino acid) deficiency exhibit a 5- to 10-fold lower level of epidermal growth factor (EGF) receptor activity than do the same cells in the rapidly growing state or arrested in G1 due to growth factor deficiency. The chemically transformed AKR-MCA and C3H/MCA-58 cell lines spontaneously arrest growth in G1 due to nutrient deficiency when grown to saturation density in medium with 10% fetal bovine serum. An examination of 125I-labeled EGF binding in rapidly growing and G1-arrested AKR-MCA and C3H/MCA-58 cells showed that the G1-arrested chemically transformed cells also have a 10- to 20-fold reduction in the amount of 125I-labeled EGF binding relative to the same cells in the rapidly growing state. Stimulation of DNA synthesis in the arrested cells by the addition of serum-free medium caused a 6- to 10-fold increase in 125I-labeled EGF binding. This recovery of receptor activity was inhibited by actinomycin D and cycloheximide, suggesting that new messenger RNA synthesis as well as increased protein synthesis is necessary for the recovery of EGF binding. A comparison of EGF binding in C3H/MCA-58 cells and the nontransformed parent line (C3H/10T 1/2) in the rapidly growing state showed the same approximate level of receptor activity. However, the rapidly growing AKR-MCA cells had approximately one-tenth the amount of EGF binding as did the rapidly growing nontransformed parent line (AKR-2B). Scatchard analysis of binding data showed a 10-fold greater number of receptors in the AKR-2B cells relative to the AKR-MCA cells with a lesser difference in apparent receptor affinity. The chemically transformed BP-3T3, like the other two chemically transformed lines, was also demonstrated to arrest growth spontaneously due to nutrient deficiency with an associated 100-fold decrease in EGF binding. Rapidly growing BP-3T3 cells had only slightly less 125I-labeled EGF binding than did the nontransformed parent line (BALB-3T3) in the rapidly growing state. The data indicate that one mechanism for reduction of EGF binding in chemically transformed cells is the propensity of these cells to arrest growth in G1 at saturation density due to low-molecular-weight nutrient deficiency, a state associated with decreased EGF binding.

    Topics: Amino Acids; Animals; Cell Cycle; Cell Division; Cell Line; Cell Transformation, Neoplastic; Cycloheximide; Dactinomycin; Epidermal Growth Factor; Mice

1982
Transforming growth factor and epidermal growth factor stimulate the phosphorylation of a synthetic, tyrosine-containing peptide in a similar manner.
    The Journal of biological chemistry, 1982, Dec-25, Volume: 257, Issue:24

    A partially purified preparation of a transforming growth factor (TGF) obtained from serum-free growth medium conditioned by a human melanoma tumor line was found to stimulate the phosphorylation of a synthetic tyrosine-containing peptide. The sequence of the peptide is related to that of the known site of tyrosine phosphorylation in the Rous sarcoma virus-encoded transforming protein, pp60src. In A431 membranes, the characteristics of TGF- and epidermal growth factor (EGF)-stimulated peptide phosphorylation are nearly identical. The effects of the two growth factors are not additive, suggesting that TGF and EGF stimulate peptide phosphorylation through the same EGF receptor system. This conclusion is supported by the finding that both TGF and EGF stimulate peptide phosphorylation in wild type Swiss 3T3 cell membranes, but neither factor is effective in stimulating peptide phosphorylation in membranes prepared from EGF receptor-deficient NR6 3T3 cells.

    Topics: Animals; Carcinoma, Squamous Cell; Cell Line; Cell Membrane; Cell Transformation, Neoplastic; Cells, Cultured; Epidermal Growth Factor; Humans; Kinetics; Melanoma; Membrane Proteins; Mice; Peptides; Phosphorylation; Transforming Growth Factors; Tyrosine

1982
Characteristics of C3H/He mouse embryo cell lines established by culture with or without benzaldehyde.
    Gan, 1982, Volume: 73, Issue:4

    The characteristics of RME-5-3-1 cell line, which had been established from C3H/He mouse embryos by culture in benzaldehyde-containing medium, were compared with those of a benzaldehyde-untreated cell line, RME-5-1, derived from the same embryos as the former and with those of RME-5-1/TMT cell line, reestablished from the tumors induced by implantation of RME-5-1 cells into syngeneic mice. The characterization of these cell lines covered cell morphology, chromosome distribution, population doubling time, saturation density, fibronectin, epidermal growth factor receptor, plasminogen activator, ornithine decarboxylase, anchorage-independent growth and transplantability into mice. The results indicated that RME-5-3-1 cells had well-preserved normal phenotypes, while both RME-5-1 and RME-5-1/TMT cells showed malignant phenotypes to varying degrees.

    Topics: Animals; Benzaldehydes; Cell Line; Cell Transformation, Neoplastic; Culture Media; Embryo, Mammalian; Epidermal Growth Factor; Fibronectins; Karyotyping; Methods; Mice; Mice, Inbred C3H; Neoplasm Transplantation; Ornithine Decarboxylase; Receptors, Drug

1982
Differences in mechanisms of transformation by independent feline sarcoma virus isolates.
    Journal of virology, 1981, Volume: 38, Issue:3

    The Gardner and Snyder-Theilen isolates of feline sarcoma virus (FeSV) have previously been shown to encode high-molecular-weight polyproteins with a transforming function and an associated tyrosine-specific protein kinase activity. Cells transformed by these viruses exhibited morphological alterations, elevated levels of phosphotyrosine, and a reduced capacity for binding epidermal growth factor. In addition, polyproteins encoded by both of these FeSV isolates bound to, and phosphorylated tyrosine acceptor sites within, a 150,000-molecular-weight cellular substrate (P150). McDonough FeSV-transformed cells resembled Gardner and Snyder-Theilen FeSV transformants with respect to morphological changes and a reduced capacity for epidermal growth factor binding. in contrast to the other two FeSV isolates, however, McDonough FeSV encoded as its major translational product a high-molecular-weight polyprotein with probable transforming function but without protein kinase activity detectable under similar assay conditions. Moreover, total cellular levels of phosphotyrosine remained unaltered in McDonough FeSV-transformed cells, and the major McDonough FeSV polyprotein translational product lacked binding affinity for P150. These findings argue for differences in the mechanisms of transformation by these independently derived FeSV isolates.

    Topics: Animals; Cell Line; Cell Transformation, Neoplastic; Cell Transformation, Viral; Epidermal Growth Factor; Mink; Phosphotyrosine; Protein Kinases; Rats; Retroviridae; Sarcoma Viruses, Feline; Tyrosine; Viral Proteins

1981
Specific binding of transforming growth factor correlates with promotion of anchorage independence in EGF receptorless mouse JB6 cells.
    Biochemical and biophysical research communications, 1981, Sep-30, Volume: 102, Issue:2

    Topics: Animals; Cell Line; Cell Transformation, Neoplastic; Clone Cells; Epidermal Growth Factor; ErbB Receptors; Humans; Mice; Peptides; Receptors, Cell Surface; Skin; Tetradecanoylphorbol Acetate; Transforming Growth Factors

1981
Regulation of intracellular protein degradation by insulin and growth factors.
    Acta biologica et medica Germanica, 1981, Volume: 40, Issue:10-11

    Protein breakdown in many cell lines is inhibited by growth factors. The response is specific, rapid and additive only at suboptimal concentrations. Transformed cells are typically more sensitive to growth factors than nontransformed cells while the effectiveness of growth factors in human fibroblasts is diminished with senescence. Bovine colostrum has been shown to be an extremely rich source of growth factors as compared to serum.

    Topics: Animals; Carcinoma, Hepatocellular; Cattle; Cell Line; Cell Survival; Cell Transformation, Neoplastic; Colostrum; Epidermal Growth Factor; Fibroblast Growth Factors; Fibroblasts; Growth Substances; Humans; Insulin; Liver Neoplasms; Mice; Peptides; Proteins; Somatomedins

1981
Epidermal growth factor, like tumor promoters, enhances viral and radiation-induced cell transformation.
    Carcinogenesis, 1981, Volume: 2, Issue:3

    The polypeptide hormone epidermal growth factor (EGF) has been shown to enhance adenovirus type 5 transformation of a cloned culture of rat embryo cells (CREF) and X-ray or u.v.-light induced transformation of 10T1/2 mouse embryo cells. In both systems, the degree of enhancement was quantitatively similar to that observed in treated rats grown in the presence of the potent tumor promoting agent 12-O-tetradecanoyl-phorbol-13-acetate (TPA). An increase in viral transformation was also observed in cells continuously exposed to phorbol esters with known promoting activity on mouse skin, but not structurally related analogs, inactive or weakly active in the two-stage mouse skin carcinogenesis assay. In addition, the appearance of transformed foci was accelerated and colonies tended to be larger in cultures grown in the presence of EGF or TPA. The present studies suggest the possibility that EGF may function as an endogenous promoter of carcinogenesis and further indicates that in vitro cell transformation systems may prove useful in identifying such agents.

    Topics: Adenoviruses, Human; Animals; Carcinogens; Cell Transformation, Neoplastic; Cells, Cultured; Epidermal Growth Factor; Mice; Stimulation, Chemical

1981
New class of transforming growth factors potentiated by epidermal growth factor: isolation from non-neoplastic tissues.
    Proceedings of the National Academy of Sciences of the United States of America, 1981, Volume: 78, Issue:9

    Proteins potentiated by epidermal growth factor (EGF) to induce a transformed phenotype in non-neoplastic rat kidney fibroblasts in cell culture have been isolated from many non-neoplastic tissues of the adult mouse, including submaxillary gland, kidney, liver, muscle, heart, and brain. They resemble previously described transforming growth factors (TGFs) isolated from neoplastic cells as follows: they are extractable by acid/ethanol and are acid-stable, low molecular weight (6000-10,000) polypeptides requiring disulfide bonds for activity; and they cause anchorage-independent growth of non-neoplastic indicator cells that will not grow in soft agar in their absence. Partial purification of these TGFs from submaxillary glands of male mice shows that they are distinct from EGF. Unlike previously described extracellular TGFs, but like certain cellular TGFs from neoplastic cells, they are potentiated by EGF in their ability to promote anchorage-independent growth. The isoelectric point of the submaxillary gland TGF protein is near neutrality. Chromatography on Bio-Gel P-30 followed by high-pressure liquid chromatography has resulted in a 22,000-fold overall purification. The most purified protein is active in inducing growth in soft agar at 1 ng/ml when assayed in the presence of EGF. The data add further evidence to the concept that neoplasia may result from a quantitative, rather than qualitative, alteration in non-neoplastic biochemical processes.

    Topics: Animals; Cell Transformation, Neoplastic; Cells, Cultured; Chromatography, High Pressure Liquid; Drug Synergism; Epidermal Growth Factor; Growth Substances; Isoelectric Point; Male; Mice; Peptides; Rats; Transforming Growth Factors

1981
On deciding which factors regulate cell growth.
    Progress in clinical and biological research, 1981, Volume: 66 Pt A

    Many conditions affect the growth of animal cells in culture. Some, such as essential nutrients, are necessary for growth as precursors of macromolecules. Other substances appear to have regulatory functions. Their presence or absence determines whether the cell will continue to grow or move into a resting, quiescent state. In particular certain factors (usually provided by serum) are thought to have regulatory roles. To decide which factors are regulatory is a difficult problem. We propose here that no single criterion is sufficient to permit a decision and that the best course at present is to apply several criteria. Three such criteria are proposed. To the degree that the tests are satisfied we can tentatively decide whether or not a given factor is regulatory.

    Topics: Animals; Caffeine; Cell Cycle; Cell Division; Cell Line; Cell Transformation, Neoplastic; Cell Transformation, Viral; Cells, Cultured; Cycloheximide; Epidermal Growth Factor; Humans; Insulin; Kinetics; Lectins; Mutation; Tetradecanoylphorbol Acetate; Thrombin

1981
Changes in epidermal growth factor receptors associated with adenovirus transformation, chemical carcinogen transformation and exposure to a phorbol ester tumor promoter.
    Biochemical and biophysical research communications, 1980, Apr-29, Volume: 93, Issue:4

    Topics: Adenoviruses, Human; Animals; Carcinogens; Cell Transformation, Neoplastic; Cell Transformation, Viral; Cells, Cultured; Epidermal Growth Factor; Mice; Peptides; Phorbols; Rats; Receptors, Cell Surface; Tetradecanoylphorbol Acetate

1980
Abelson murine leukaemia virus transformation involves loss of epidermal growth factor-binding sites.
    Nature, 1980, Jul-31, Volume: 286, Issue:5772

    Malignant transformation by mammalian RNA sarcoma viruses has previously been shown to involve a reduction in receptor sites for a well characterized 6,000-molecular weight (MW) growth-promoting substance, designated epidermal growth factor (EGF). Although Abelson murine leukaemia virus (AbLV) resembles sarcoma viruses in its ability to transform embryo fibroblasts in cell culture, AbLV induces a rapid B-cell lymphoid leukaemia rather than fibrosarcomas in vivo. The major translational product of AbLV is a highly phosphorylated polyprotein of MW 120,000 which exhibits an associated tyrosine-specific protein kinase activity and probable transforming function. We show here that AbLV transformation resembles transformation by RNA sarcoma viruses with respect to the abolition of EGF-binding sites. EGF binding is restored to control levels following loss of polyprotein expression in morphological revertants of AbLV-transformed clones and remains uninfluenced in cell lines infected with transformation-defective (td) AbLV mutants encoding polyproteins deficient in protein kinase activity. These findings indicate that AbLV transformation involves a polyprotein-associated, tyrosine-specific protein kinase activity which mediates its effect through a mechanism resulting directly or indirectly in the abolition of EGF-binding sites.

    Topics: Abelson murine leukemia virus; Animals; Cell Line; Cell Transformation, Neoplastic; Cell Transformation, Viral; Epidermal Growth Factor; Leukemia Virus, Murine; Mink; Peptides; Phosphoproteins; Protein Kinases; Rats; Receptors, Cell Surface; Tyrosine

1980
Kirsten murine sarcoma virus transformed cell lines and a spontaneously transformed rat cell-line produce transforming factors.
    Journal of cellular physiology, 1980, Volume: 105, Issue:1

    We have examined culture fluids from a variety of Kirsten murine sarcoma virus (KiMSV) transformed rat and mouse cells for the presence of factors which induce normal Rat-1 cells to assume the transformed phenotype. All KiMSV transformants produced transforming factor (TF). Revertants of KiMSV transformed rat or mouse failed to release TF as did normal rat or mouse cells. Cells transformed by a temperature sensitive mutant of KiMSV produced TF at the permissive temperature but not at the nonpermissive temperature. Further, cells from a spontaneous transformant of Rat-1 cells also produced TF. TF is a small polypeptide which competes for the epidermal growth factor receptor. Its effect upon normal cells is reversible and requires de novo RNA and protein synthesis. Cells treated with TF lose the actin fibers observed in normal fibroblasts, assume a transformed cell morphology, become anchorage independent for growth, grow in low concentrations of serum, grow to a high cell density, and have an increased rate of hexose uptake.

    Topics: Actins; Animals; Cell Division; Cell Line; Cell Transformation, Neoplastic; Cell Transformation, Viral; Deoxyglucose; Epidermal Growth Factor; ErbB Receptors; Kirsten murine sarcoma virus; Peptide Biosynthesis; Rats; Receptors, Cell Surface; Transforming Growth Factors

1980
Factors which disorganize microtubules or microfilaments increase the frequency of cell transformation by polyoma virus.
    Journal of virology, 1980, Volume: 36, Issue:2

    Griseofulvin, 12-O-tetradecanoyl phorbol-13-acetate, melittin, epidermal growth factor, vinblastine, cytochalasin B, podophyllotoxin, colcemid, and colchicine were unable to transform cells but could increase from 8- to 40-fold the frequency of cell transformation by polyoma virus. The 3T3-like cells were resting at confluence and were exposed to the drug only during the 1st week after viral infection. Griseofulvin, a tumor promoter, reduced or increased the frequency of transformation depending on the dose with which the infected cells were treated. The antitumor activity of tumor promoters is discussed.

    Topics: Cell Line; Cell Transformation, Neoplastic; Cell Transformation, Viral; Colchicine; Cytochalasin B; Epidermal Growth Factor; Griseofulvin; Melitten; Mutagens; Podophyllotoxin; Polyomavirus; Tetradecanoylphorbol Acetate; Vinblastine

1980
Transforming growth factors produced by certain human tumor cells: polypeptides that interact with epidermal growth factor receptors.
    Proceedings of the National Academy of Sciences of the United States of America, 1980, Volume: 77, Issue:9

    Three different human tumor lines in culture, a rhabdomyosarcoma, a bronchogenic carcinoma and a metastatic melanoma, release proteins (transforming growth factors, TGFs) into the medium that confer the transformed phenotype on untransformed fibroblasts. These proteins are acid and heat-stable; produce profound morphologic changes in rat and human fibroblasts; and enable normal anchorage-dependent cells to grow in agar. Removal of the transforming protein results in a reversion of cell phenotype. The major activity interacts with epidermal growth factor (EGF) cell membrane receptors. The peptides from these tumor cells are similar in their action to the sarcoma growth factor (SGF) released by murine sarcoma virus-transformed rodent cells. The most anchorage-independent tumor cells released the most TGFs. EGF-related TGFs were not detectable in fluids from cultures of cells with high numbers of free EGF membrane receptors (normal human fibroblasts and human carcinomas).

    Topics: Adult; Aged; Animals; Binding, Competitive; Carcinoma, Bronchogenic; Carcinoma, Squamous Cell; Cell Division; Cell Transformation, Neoplastic; Cells, Cultured; DNA; Epidermal Growth Factor; ErbB Receptors; Female; Fibroblasts; Growth Substances; Humans; Lung Neoplasms; Male; Melanoma; Peptides; Rats; Receptors, Cell Surface; Rhabdomyosarcoma

1980
Modulation of phagocytosis by tumor promoters and epidermal growth factor in normal and transformed macrophages.
    Cancer research, 1980, Volume: 40, Issue:4

    Topics: Animals; Cell Line; Cell Transformation, Neoplastic; Diterpenes; Epidermal Growth Factor; Erythrocytes; Female; In Vitro Techniques; Macrophages; Mice; Microscopy, Electron, Scanning; Peptides; Phagocytosis; Phorbols; Pyran Copolymer; Tetradecanoylphorbol Acetate; Thioglycolates

1980
Improved conditions for murine epidermal cell culture.
    In vitro, 1980, Volume: 16, Issue:2

    An improved method for cultivating newborn mouse epidermal cells has been developed that increases the longevity, epithelial nature and efficiency of cell-line establishment. The use of Super Medium, an enriched Waymouth's formulation, increased proliferation for long periods of time, as did incubation at 31 degrees C rather than 37 degrees C. The fetal bovine serum requirement was found to be reduced at the lower temperature. An increase in labeling indices was seen when epidermal growth factor (EGF) or the cyclic nucleotides were added and the presence of EGF receptors was determined. Of the prostaglandins (PG) examined, PGE1 and PGE2 produced the greatest increase in DNA synthesis. The PG precursors, arachidonic and 8,11,14-eicosatrienoic acid, were also greatly stimulatory. The use of a lethally irradiated 3T3 feeder layer at 31 degrees C proved superior in maintenance of an epithelial morphology. Subculturable cell lines were established much more readily and reproducibly in carcinogen-treated cultures grown under the improved conditions.

    Topics: Animals; Blood; Cell Line; Cell Transformation, Neoplastic; Cells, Cultured; Culture Media; Cytological Techniques; Epidermal Cells; Epidermal Growth Factor; Mice; Prostaglandins; Temperature

1980
Synergistic effects of epidermal growth factor and thrombin on the growth stimulation of diploid Chinese hamster fibroblasts.
    Journal of cellular physiology, 1980, Volume: 105, Issue:1

    The serum supplement used in the culture of a variety of mammalian cells can be replaced by known growth factors. Diploid Chinese hamster fibroblasts (CHEF/18) will grow for several days in a medium (4F) supplemented with four growth factors: epidermal growth factor (EGF), insulin, fibroblast growth factor (FGF), and transferrin. The growth rate is only about 50% as fast as when fetal calf serum is added. This difference is eliminated by thrombin (10--100 ng/ml; 0.3--3 nM). The CHEF/18 cell line is unique in that no other cell line responds to thrombin in this concentration range. Thrombin acts synergistically with other growth factors to stimulate CHEF/18 cell growth. By itself, thrombin is only mitogenic at elevated concentrations. Thrombin can largely compensate for the absence of EGF and partly for the absence of insulin in serum-free media. Chemically and "spontaneously" transformed cell lines related to CHEF/18 have lost requirements for both EGF and thrombin, and have retained requirements for insulin and transferrin expressed by CHEF/18. No CHEF cells in this work required FGF. These results suggest that the mechanisms by which EGF and thrombin stimulate cells to grow are related.

    Topics: Animals; Cell Division; Cell Line; Cell Transformation, Neoplastic; Cricetinae; DNA; Drug Synergism; Epidermal Growth Factor; Fibroblasts; Insulin; Peptides; Thrombin; Transferrin

1980
Selective reduction in receptors for epidermal growth factor-urogastrone in chemically transformed tumorigenic Syrian hamster embryo fibroblasts.
    Cancer research, 1979, Volume: 39, Issue:10

    Topics: Animals; Benzopyrenes; Cell Line; Cell Transformation, Neoplastic; Cricetinae; Culture Media; Embryo, Mammalian; Epidermal Growth Factor; Fibroblasts; Gastrointestinal Hormones; Mesocricetus; Peptides; Receptors, Cell Surface; Thymidine

1979
Loss of epidermal growth factor requirement and malignant transformation.
    Proceedings of the National Academy of Sciences of the United States of America, 1979, Volume: 76, Issue:8

    Serum provides growth factors that regulate and limit the growth of normal cells in tissue culture. Animal cells that are malignantly transformed usually exhibit diminished serum requirements for growth in culture. We have used a defined, serum-free medium to determine which of these growth factors becomes dispensable for the growth of transformed Syrian and Chinese hamster fibroblast cells. The medium's four growth factors-epidermal growth factor (EGF), insulin, fibroblast growth factor, and transferrin-were added or omitted as desired. A decreased requirement for EGF was most closely related to tumorigenicity of chemically (ethyl methanesulfonate) transformed cells in nude mice. All lines examined retained their requirement for transferrin, which is needed throughout the growth cycle, in contrast to the other factors, which are needed primarily in G(1) phase. Lines that had lost their EGF requirement but had retained their insulin requirement were arrested in G(1) by insulin deficiency, indicating that their growth control system remained. Mutagenesis with ethyl methanesulfonate can also create requirements of the transformed cells for unknown factors in serum. We conclude that an initial step that reduces the serum requirement in culture, and in tumorigenesis, is relaxation of the growth-regulatory function of EGF.

    Topics: Blood; Cell Division; Cell Line; Cell Transformation, Neoplastic; Cell Transformation, Viral; Culture Media; Epidermal Growth Factor; Fibroblasts; Growth Substances; Insulin; Peptides; Polyomavirus; Transferrin

1979
Reduction of the calcium requirement of normal human epithelial cells by EGF.
    Experimental cell research, 1979, Volume: 121, Issue:2

    Topics: Calcium; Cell Division; Cell Line; Cell Transformation, Neoplastic; Clone Cells; Dose-Response Relationship, Drug; Epidermal Growth Factor; Humans; Kinetics; Peptides

1979
Epidermal growth factor and the control of proliferation of Balb 3T3 and benzo[a]pyrene-transformed Balb 3T3 cells.
    Journal of cellular physiology, 1979, Volume: 100, Issue:1

    Benzo[a]pyrene-transformed Balb 3T3 cells (BP3T3) exhibit "normal" growth controls at low concentrations of serum. Epidermal growth factor (EGF) stimulates DNA synthesis and cell division in both Balb 3T3 and BP3T3 cells at physiological concentrations. The growth response of BP3T3 cells to EGF is qualitatively the same as that of 3T3 cells, however, the transformed cells have a lower quantitative requirement. Both 3T3 and BP3T3 cells show a density-dependent response to EGF, but the shift in the dose response curve for BP3T3 cells at high cell density is smaller than that seen for 3T3 cells. One cause of the restricted growth of 3T3 cells at high cell density compared with BP3T3 cells is the increased concentration of growth factor needed for stimulation of 3T3 cells at higher cell densities. A lower rate of depletion of other growth factory by BP3T3 cells may also explain the smaller effect of cell density on the EGF response of these cells.

    Topics: Animals; Autoradiography; Benzopyrenes; Cell Division; Cell Transformation, Neoplastic; Cells, Cultured; DNA; Embryo, Mammalian; Epidermal Growth Factor; Humans; Immune Sera; Mice; Mice, Inbred BALB C; Peptides; Time Factors

1979
Decrease in epidermal growth factor receptor levels and production of material enhancing epidermal growth factor binding accompany the temperature-dependent changes from normal to transformed phenotype.
    Proceedings of the National Academy of Sciences of the United States of America, 1979, Volume: 76, Issue:7

    Normal rat kidney (NRK) cells infected with a temperature-sensitive mutant of Kirsten sarcoma virus (Ts cells) exhibited normal monolayer morphology identical to that observed for uninfected cells (NRK cells) at the nonpermissive temperature, 39 degrees C, but grew as multilayered foci resembling NRK cells transformed by the wild-type virus (KNRK cells) at 32 degrees C, the permissive temperature. NRK cell division was stimulated by epidermal growth factor (EGF), and these cells showed high levels of EGF receptors, as determined by 125I-labeled EGF binding. KNRK cells were unresponsive to EGF and no EGF receptors were detectable. Ts cells also were unresponsive to EGF at both temperatures, but exhibited just detectable EGF binding at 32 degrees C and 10-15% of NRK cell binding at 39 degrees C. Use of EGF added to the culture medium by these cells paralleled the receptor levels. Crossfeeding experiments among NRK, KNRK, and Ts cultures indicated that Ts cells at the permissive temperature and KNRK cells at both temperatures produced a heat-stable substance(s) which stimulated DNA synthesis in NRK cells independent of the presence of serum or of EGF. Conditioned medium from the transformed cultures also significantly enhanced EGF binding to NRK cells. These studies demonstrated a correlation between the transformed phenotype and the receptor levels of a potent cell mitogen, EGF, which was readily reversible in the Ts cultures. In addition, cultures expressing the transformed phenotype produced material that did not compete for the EGF receptor but did enhance EGF binding, in contrast to other reports involving sarcoma virus-transformed cells.

    Topics: Animals; Binding Sites; Cell Division; Cell Transformation, Neoplastic; Cells, Cultured; Epidermal Growth Factor; Kidney; Kirsten murine sarcoma virus; Mutation; Peptides; Rats; Temperature

1979
Binding, internalization, and degradation of epidermal growth factor by balb 3T3 and BP3T3 cells: relationship to cell density and the stimulation of cell proliferation.
    Journal of cellular physiology, 1979, Volume: 100, Issue:2

    Epidermal growth factor (EGF) stimulates the growth of both benzo[a]pyrene-transformed Balb 3T3 cells (BP3T3) and untransformed Balb 3T3 cells. We describe here the binding, internalization, and degradation of [125I]-EGF by BP3T3 cells and 3T3 cells. Binding of [125I]-EGF reaches a maximum after 45 to 90 minutes incubation at 37 degrees C. In both BP3T3 and 3T3 cells the extent of EGF binding required to stimulate DNA synthesis is density dependent; sparse cultures require a 15-30% occupancy to elicit a maximal response whereas dense cultures require a 70-85% occupancy. At physiological concentrations the total binding of [125I]-EGF to 3T3 cells is higher than to BP3T3 cells, and this difference increases at higher cell densities. The rate of degradation of [125I]-EGF is directly proportional to the total [125I]-EGF binding in each cell type. This supports the hypothesis that one cause of the diminished serum requirement of BP3T3 cells is a reduced rate of utilization of serum growth factors.

    Topics: Animals; Benzopyrenes; Cell Count; Cell Division; Cell Line; Cell Transformation, Neoplastic; Clone Cells; Epidermal Growth Factor; Kidney; Mice; Mice, Inbred BALB C; Peptides

1979
Tumor promoters and epidermal growth factor stimulate anchorage-independent growth of adenovirus-tranformed rat embryo cells.
    Cell, 1979, Volume: 18, Issue:3

    Previous studies indicated that the potent tumor promoter 12--0--tetradecanoyl-phorbol-13-acetate (TPA) enhances transformation of rat embryo cells (2 degrees RE) by a mutant of human Ad5 (H5ts125). This study examines the effect of TPA, its structural analogs and epidermal growth factor (EGF) on anchorage-independent growth of a cloned population of H5ts125-transformed 2 degrees RE cells (clone E11). Both TPA and EGF (approximately 10(-8) M) induced a 3--5 fold increase in agar cloning efficiency of E11 cells. In addition, macroscopic colonies appeared earlier and were larger and more diffuse. The TPA analogs phorbol--12,13--didecanoate (PDD) and ingenol--3,20--dibenzoate also enhanced growth in agar of E11 cells, whereas phorbol, 4 alpha PDD and 4--0--meTPA, which are inactive as tumor promoters, failed to enhance agar growth. In contrast to the results obtained with E11 cells, TPA, PDD or ingenol--3,20--bidenzoate failed to induce growth in agar of normal 2 degrees RE cells. Dexamethasone (10(-5)--10(-6) M), trans retinoic acid (10(-5)--10(-6) M) and the protease inhibitors leupeptin, antipain and elastatinol did not inhibit the ability of TPA to enhance the growth of E11 cells in agar. The TPA-enhanced anchorage independence was a stable property, since subclones of E11 cells isolated from TPA-agar plates had a higher agar cloning efficiency than the parental E11 cells when retested in the absence of TPA. This effect of TPA does not appear to reflect simple selection of a subpopulation of cells. When the parental E11 cells were first cloned in monolayer culture in the absence of TPA, all ten randomly picked clones showed enhanced growth in agar in the presence of TPA. In addition, prior growth of E11 cells in monolayer culture in the presence of TPA did not enhance their subsequent growth in agar. This system therefore provides an example in which TPA appears to enhance the acquisition of a stable cell property, and thus may be a useful model for studying mechanisms of tumor promotion and progression.

    Topics: Adenoviruses, Human; Animals; Cell Adhesion; Cell Division; Cell Transformation, Neoplastic; Clone Cells; Dexamethasone; Diterpenes; Embryo, Mammalian; Epidermal Growth Factor; Peptides; Phorbols; Protease Inhibitors; Rats; Tetradecanoylphorbol Acetate; Tretinoin

1979
Growth control of normal and transformed cells.
    Journal of supramolecular structure, 1979, Volume: 11, Issue:4

    Both serum factors and protein synthesis are required for normal cell growth. Swiss 3T3 cells require the serum growth factors insulin and EGF (epidermal growth factor) during the initial part of the G1 period, until they pass a restriction point about 2 h before the initiation of DNA synthesis. Concentrations of cycloheximide that inhibit protein synthesis by as much as 70% dramatically lengthen the cell cycle before the restriction point, while the cell cycle after the restriction point remains nearly constant. These results are consistent with a model in which labile proteins are required for transit of cells past the serum-sensitive restriction point. The relation of these findings to the growth control of transformed cells is discussed.

    Topics: Animals; Cell Cycle; Cell Division; Cell Line; Cell Transformation, Neoplastic; Cycloheximide; DNA; Epidermal Growth Factor; Insulin; Kinetics; Mice; Mice, Inbred BALB C; Mitosis; Protein Biosynthesis

1979
Growth factors for tumours.
    Nature, 1978, Apr-20, Volume: 272, Issue:5655

    Topics: Animals; Cell Line; Cell Transformation, Neoplastic; Cell Transformation, Viral; Epidermal Growth Factor; Humans; Neoplasms; Peptides; Protein Biosynthesis; Receptors, Cell Surface

1978
3T3 variants lacking receptors for epidermal growth factor are susceptible to transformation by Kirsten sarcoma virus.
    Nature, 1978, Jul-20, Volume: 274, Issue:5668

    Topics: Cell Line; Cell Transformation, Neoplastic; Epidermal Growth Factor; Gammaretrovirus; Mitosis; Peptides; Receptors, Drug; Sarcoma Viruses, Murine

1978
Intracellular plasminogen activator activity in growing and quiescent cells.
    Journal of cellular physiology, 1978, Volume: 97, Issue:1

    Topics: Animals; Benzopyrenes; Blood Proteins; Cell Division; Cell Line; Cell Transformation, Neoplastic; Cell Transformation, Viral; Cycloheximide; Dactinomycin; Epidermal Growth Factor; Mice; Plasminogen Activators; Simian virus 40

1978
Tumor promoters: carcinogenesis gets more complicated.
    Science (New York, N.Y.), 1978, Aug-11, Volume: 201, Issue:4355

    Topics: Animals; Carcinogens; Cell Transformation, Neoplastic; Cocarcinogenesis; Dietary Fats; Epidermal Growth Factor; Humans; Neoplasms; Ornithine Decarboxylase; Phenobarbital; Plasminogen Activators; Receptors, Drug; Saccharin; Smoking; Tetradecanoylphorbol Acetate

1978
Modulation of cell surface glycocalyx: studies on large, external, transformation-sensitive protein.
    Annals of the New York Academy of Sciences, 1978, Jun-20, Volume: 312

    Topics: Animals; Cell Communication; Cell Line; Cell Transformation, Neoplastic; Collagen; Cricetinae; Epidermal Growth Factor; Extracellular Space; Fibronectins; Fluorescent Antibody Technique; Microscopy, Electron, Scanning; Neoplasms, Experimental

1978
MSA and EGF receptors on sarcoma virus transformed cells and human fibrosarcoma cells in culture.
    Nature, 1977, Jun-09, Volume: 267, Issue:5611

    Topics: Cell Line; Cell Transformation, Neoplastic; Cells, Cultured; Epidermal Growth Factor; Fibroblasts; Fibrosarcoma; Gammaretrovirus; Growth Substances; Oncogenic Viruses; Peptides; Receptors, Drug; Sarcoma Viruses, Feline; Sarcoma Viruses, Murine

1977
Control of a cell surface major glycoprotein by epidermal growth factor.
    Science (New York, N.Y.), 1977, Aug-19, Volume: 197, Issue:4305

    When the serum concentration of the culture medium is below 0.7 per-cent, 3T3 mouse cells lose most of their large external transformation sensitive (LETS) protein at the cell surface, Subsequent addition of epidermal growth factor results in the reappearance of massive fibrillar LETS protein networks on the surface of conluent 3T3 cells. Thirteen other hormones tested do not have this effect. It appears that epidermal growth factor can control the expression of LETS protein.

    Topics: Blood; Cell Line; Cell Transformation, Neoplastic; Culture Media; Epidermal Growth Factor; Glycoproteins; Hormones; Membrane Proteins; Peptides

1977
Uptake of alpha-aminoisobutyric acid and phosphate by membrane vesicles derived from growing and quiescent fibroblasts.
    Journal of cellular physiology, 1976, Volume: 89, Issue:4

    Membrane vesicles derived principally from the plasma membrane and endoplasmic reticulum of mouse 3T3 cells transformed by Simian virus 40 take up alpha-aminoisobutyric acid (AIB) and phosphate (Pi). When NaCl is added simultaneously with AIB or Pi, uptake rises two- to three-times above the equilibrium to accumulate AIB or Pi over the control value, in the presence of a Na+ gradient, is almost lost in membrane vesicles derived from benzpyrene-transformed 3T3 cells (BP3T3) arrested in the G1 phase of the cell cycle by serum starvation. When added to the membranes with NaCl and the uptake substrate, a combination of fibroblast growth factor (FGF) and epidermal growth factor EGF restores the ability of the membranes to accumulate AIB and Pi over the control value.

    Topics: Aminoisobutyric Acids; Animals; Benzopyrenes; Biological Transport, Active; Cell Division; Cell Line; Cell Membrane; Cell Transformation, Neoplastic; Endoplasmic Reticulum; Epidermal Growth Factor; Fibroblasts; Growth Substances; Mice; Phosphates; Simian virus 40; Sodium Chloride

1976