thymosin-beta(4) and Neoplasm-Metastasis

thymosin-beta(4) has been researched along with Neoplasm-Metastasis* in 17 studies

Reviews

3 review(s) available for thymosin-beta(4) and Neoplasm-Metastasis

ArticleYear
Roles and mechanisms of β-thymosins in cell migration and cancer metastasis: an update.
    Cancer investigation, 2013, Volume: 31, Issue:2

    β-thymosins, including thymosin β4 (Tβ4), Tβ10, and Tβ15, are a family of highly conserved 5 kDa peptides. They are involved not only in normal cell migration, but also in tumor metastasis. However, the molecular mechanisms of β-thymosins to regulate cell migration and other functions are not fully understood. Recently, this important area is under active investigation worldwide. Many new discoveries have been made from molecular biology and cell culture models as well as animal models and human diseases. This timely review provides the most updated information about functional roles and molecular mechanisms of β-thymosins in normal tissues and disease conditions.

    Topics: Actins; Animals; Cell Movement; Epithelial-Mesenchymal Transition; Humans; Neoplasm Metastasis; Neoplasms; Thymosin

2013
Thymosin β4: a potential molecular target for tumor therapy.
    Critical reviews in eukaryotic gene expression, 2012, Volume: 22, Issue:2

    Thymosin β4 (Tβ4), a 5 kDa protein, has been demonstrated to play an important role in a variety of biological activities, such as actin sequestering, cellular motility, migration, inflammation, and damage repair. Recently, several novel findings provided compelling evidence that Tβ4 played a key role in facilitating tumor metastasis and angiogenesis. It has been found that Tβ4 expressed increasingly in a number of metastatic tumors, which was associated with an increased expression of a known angiogenic factor, vascular endothelial growth factor. Thus, Tβ4 provided a potential target of opportunity for cancer management, especially for cancer metastasis therapy.

    Topics: Animals; Cell Movement; Cell Survival; Cell Transformation, Neoplastic; Drug Resistance, Neoplasm; Epithelial-Mesenchymal Transition; Gene Expression Regulation, Neoplastic; Humans; Inflammation; Neoplasm Metastasis; Neoplastic Stem Cells; Neovascularization, Pathologic; Thymosin; Wound Healing

2012
beta-Thymosins, small acidic peptides with multiple functions.
    The international journal of biochemistry & cell biology, 2001, Volume: 33, Issue:3

    The beta-thymosins are a family of highly conserved polar 5 kDa peptides originally thought to be thymic hormones. About 10 years ago, thymosin beta(4) as well as other members of this ubiquitous peptide family were identified as the main intracellular G-actin sequestering peptides, being present in high concentrations in almost every cell. beta-Thymosins bind monomeric actin in a 1:1 complex and act as actin buffers, preventing polymerization into actin filaments but supplying a pool of actin monomers when the cell needs filaments. Changes in the expression of beta-thymosins appear to be related to the differentiation of cells. Increased expression of beta-thymosins or even the synthesis of a beta-thymosin normally not expressed might promote metastasis possibly by increasing mobility of the cells. Thymosin beta(4) is detected outside of cells in blood plasma or in wound fluid. Several biological effects are attributed to thymosin beta(4), oxidized thymosin beta(4), or to the fragment, acSDKP, possibly generated from thymosin beta(4). Among the effects are induction of metallo-proteinases, chemotaxis, angiogenesis and inhibition of inflammation as well as the inhibition of bone marrow stem cell proliferation. However, nothing is known about the molecular mechanisms mediating the effects attributed to extracellular beta-thymosins.

    Topics: Actins; Amino Acid Sequence; Animals; Binding Sites; Cell Differentiation; Cell Movement; Humans; Molecular Sequence Data; Neoplasm Metastasis; Neoplasms; Neovascularization, Pathologic; Oxidation-Reduction; RNA, Messenger; Thymosin; Wound Healing

2001

Other Studies

14 other study(ies) available for thymosin-beta(4) and Neoplasm-Metastasis

ArticleYear
In vivo CRISPR-Cas9 knockout screening using quantitative PCR identifies thymosin beta-4 X-linked that promotes diffuse-type gastric cancer metastasis.
    Molecular carcinogenesis, 2021, Volume: 60, Issue:9

    Gastric cancer (GC) is histologically classified into intestinal-type gastric cancer (IGC) and diffuse-type gastric cancer (DGC), and the latter is poorly differentiated and highly metastatic. In this study, using quantitative real-time polymerase chain reaction, we described a complete protocol for in vivo CRISPR-Cas9-based knockout screening of essential genes for DGC metastasis. We functionally screened 30 candidate genes using our mouse DGC models lacking Smad4, p53, and E-cadherin. Pooled knockout mouse DGC cells were transplanted into a spleen of syngeneic immunocompetent mice to study clonal advantages in context of a complex process of liver metastasis. Tmsb4x (thymosin beta-4 X-linked), Hmox1, Ifitm3, Ldhb, and Itgb7 were identified as strong candidate genes that promote metastasis. In particular, Tmsb4x enhanced DGC metastasis and stomach organoid-generated tumor growth in in vivo transplantation models. Tmsb4x promoted tumor clonogenicity and anoikis resistance. In situ hybridization analysis showed that Tmsb4x is highly expressed in E-cadherin-negative mouse DGC models compared with mouse IGC and intestinal cancer models. E-cadherin deficiency also increased Tmsb4x expression in stomach organoids via Wnt signaling activation. Collectively, these results demonstrate that Tmsb4x promotes DGC metastasis. In addition, this experimental system will aid in the identification of novel target genes responsible for DGC metastasis.

    Topics: Animals; Biomarkers, Tumor; CRISPR-Cas Systems; Disease Models, Animal; Gene Expression; Gene Knockout Techniques; Humans; Mice; Neoplasm Metastasis; Real-Time Polymerase Chain Reaction; Signal Transduction; Stomach Neoplasms; Thymosin

2021
A loss of profilin-1 in late-stage oral squamous cell carcinoma.
    Journal of oral pathology & medicine : official publication of the International Association of Oral Pathologists and the American Academy of Oral Pathology, 2017, Volume: 46, Issue:7

    The genes for PFN1 and TMSB4 are both highly expressed in oral tissue and both encode actin monomer binding proteins thought to play a role in cell motility and possibly other crucial parts of tumor progression.. Oral brush cytology of epithelium from oral squamous cell carcinoma (OSCC) was used to measure PFN1 and TMSB4 mRNA in OSCC, while immunohistochemical analysis of tissue was used to check protein levels.. High but variable expression of mRNAs encoding these two proteins was observed suggesting they may contribute to tumor characteristics in a subset of OSCCs. Both proteins were highly expressed in normal appearing basal epithelium, in the cytoplasm, and perinuclear area, while expression was minimal in upper epithelial layers. In OSCCs, expression of these proteins varied. In tumors classified as later stage, based on size and/or lymph node involvement, PFN1 levels were lower in tumor epithelium. A control gene, KRT13, showed expression in normal differentiated basal and suprabasal oral mucosa epithelial cells and as reported was lost in OSCC cells.. Loss of PFN1 in tumor cells has been associated with lymph node invasion and metastasis in other tumor types, strengthening the argument that the protein has the potential to be a tumor suppressor in late-stage OSCC.

    Topics: Aged; Carcinoma, Squamous Cell; Cell Line, Tumor; Female; Gene Expression Regulation, Neoplastic; Humans; Keratin-13; Lymphatic Metastasis; Male; Mouth Mucosa; Mouth Neoplasms; Neoplasm Metastasis; Neoplasm Staging; Profilins; RNA, Messenger; Thymosin

2017
Thymosin β4 promotes hepatoblastoma metastasis via the induction of epithelial-mesenchymal transition.
    Molecular medicine reports, 2015, Volume: 12, Issue:1

    Hepatoblastoma (HB) is the most common malignant hepatic tumor in children and complete surgical resection offers the highest possibility for cure in this disease. Tumor metastasis is the principle obstacle to the development of efficient treatments for patients with HB. The present study aimed to measure the expression levels of thymosin β4 (Tβ4) in liver samples from patients with HB and to investigate the involvement of Tβ4 in HB metastasis. The expression of Tβ4 was significantly higher in liver samples from patients with metastatic HB and in the HepG2 metastatic HB cell line, compared with that in adjacent healthy liver samples and in the L02 healthy hepatic cell line. By contrast, the expression levels of epithelial-cadherin (E-cadherin) and cytosolic accumulation of β-catenin, the two most prominent markers involved in epithelial-mesenchymal transition (EMT), were reduced in liver specimens from patients with metastatic HB compared with that of healthy adjacent control tissue. HepG2 cells were transfected with small interfering-RNA in order to downregulate Tβ4 gene expression. This resulted in a reduced cell migratory capacity compared with control cells. Tβ4 gene expression knockdown significantly inhibited transforming growth factor β1-mediated-EMT in vitro by upregulating the expression of E-cadherin. The results of the present study suggested that Tβ4 may promote HB metastasis via the induction of EMT, and that Tβ4 may therefore be a target for the development of novel treatments for patients with HB.

    Topics: beta Catenin; Cadherins; Cell Proliferation; Child; Child, Preschool; Epithelial-Mesenchymal Transition; Female; Gene Expression Regulation, Neoplastic; Hep G2 Cells; Hepatoblastoma; Humans; Infant; Male; Neoplasm Metastasis; Thymosin

2015
Hypoxia/reoxygenation-experienced cancer cell migration and metastasis are regulated by Rap1- and Rac1-GTPase activation via the expression of thymosin beta-4.
    Oncotarget, 2015, Volume: 6, Issue:12

    Signaling by small guanosine triphosphatases (GTPase), Rap1/Rac1, is one of the major pathways controlling cancer cell migration and tumor metastasis. Thymosin beta-4 (Tβ4), an actin-sequestering protein, has been shown to increase migration of cancer cells. Episodes of hypoxia and re-oxygenation (H/R) are an important phenomenon in tumor microenvironment (TME). We investigated whether Tβ4 could play as an intermediary to crosstalk between Rac1- and Rap1- GTPase activation under hypoxia/reoxygenation (H/R) conditions. Inhibition of Tβ4 expression using transcription activator-like effector nucleases (TALEN) significantly decreased lung metastasis of B16F10 cells. Rac1 and Rap1 activity, as well as cancer cell migration, increased following induction of Tβ4 expression in normoxia- or H/R-experienced cells, but were barely detectable in Tβ4-depleted cells. Rap1-regulated Rac1 activity was decreased by a dominant negative Rap1 (Rap1N17), and increased by 8-(4-chloro-phenylthio)-2'-O-methyladenosine-3',5'-cyclic monophosphate (CPT), a Rap1 activator. In contrast, a Rac1-specific inhibitor, NSC23766, and dominant negative Rac1 (Rac1N17) enhanced Tβ4 expression and aberrant Rap1 activity. While NSC23766 and Rac1N17 incompletely inhibited tumor metastasis in vivo, and H/R-experienced cancer cell migration in vitro, more efficient attenuation of cancer cell migration was accomplished by simultaneous inactivation of Rap1 and Rac1 with Rap1N17 and Rac1N17, respectively. These data suggest that a combination therapy targeting both Rap1 and Rac1 activity may be an effective method of inhibiting tumor metastasis.

    Topics: Aminoquinolines; Animals; Cell Hypoxia; Cell Line, Tumor; Cell Movement; Enzyme Activation; Gene Expression Regulation, Neoplastic; GTP Phosphohydrolases; HeLa Cells; Humans; Male; Melanoma, Experimental; Mice; Mice, Inbred C57BL; Neoplasm Metastasis; Neuropeptides; Oxygen; Pyrimidines; rac1 GTP-Binding Protein; Shelterin Complex; Signal Transduction; Telomere-Binding Proteins; Thymosin; Tumor Microenvironment; Two-Hybrid System Techniques

2015
A thymosin beta15-like peptide promotes intersegmental myotome extension in the chicken embryo.
    Histochemistry and cell biology, 2014, Volume: 141, Issue:3

    Beta-thymosins constitute a group of small actin-sequestering peptides. These highly conserved peptides are involved in cytoskeleton dynamics and can influence different cell properties such as motility, substrate adhesion, shape and chemotaxis. As a marker for tumour metastasis, the mammalian thymosin beta15 is believed to have an important diagnostic relevance in cancer prognosis, although little is known about its physiological function. In order to study the role of thymosin beta15(avian) in embryogenesis, we cloned the chicken and quail orthologues of thymosin beta15 and used the chicken as a model for vertebrate development. Avian thymosin beta15, the first known non-mammalian thymosin beta15-like gene, encodes a peptide that possesses a cysteine at position one after the methionine which is a significant difference compared to its mammalian counterparts. Thymosin beta15(avian) expression starts at an early stage of development. The expression pattern changes rapidly with development and differs from that of the related thymosin beta4 gene. The most prominent expression domain is seen in developing muscles of limbs and trunk. Gain-of-function experiments revealed that thymosin beta15(avian) has a function in normal myotome development. Ectopic over-expression of thymosin beta15(avian) leads to premature elongation of myotome cells trespassing segment borders. We conclude that thymosin beta15(avian) has a still undescribed function in promoting myocyte elongation.

    Topics: Amino Acid Sequence; Animals; Chick Embryo; Cloning, Molecular; Electroporation; Molecular Sequence Data; Muscle Cells; Muscle Development; Muscle, Skeletal; Neoplasm Metastasis; Retroviridae; Thymosin

2014
Thymosin beta 4 induces colon cancer cell migration and clinical metastasis via enhancing ILK/IQGAP1/Rac1 signal transduction pathway.
    Cancer letters, 2011, Sep-28, Volume: 308, Issue:2

    Thymosin β(4) (Tβ(4)) overexpression increases cell migration and tumor metastasis. Hence, understanding the mechanism of cancer cell migration induced by Tβ(4) may provide means to inhibit their metastasis. We demonstrated higher Rac1 activities and expression levels of IQGAP1 and ILK in highly migrated Tβ(4)-overexpressing SW480 cells. In addition, IQGAP1 formed a complex with ILK and knockdown of Tβ(4) simultaneously reduced ILK and IQGAP1 protein levels as well as their migration ability. These findings suggest that Tβ(4) increases migration of colon cancer cells via activating Rac1 by elevating IQGAP1/ILK complexes and IHC results illustrated a similar mechanism occurring in vivo.

    Topics: Animals; Blotting, Western; Cell Line, Tumor; Cell Movement; Colonic Neoplasms; Humans; Mice; Neoplasm Metastasis; Protein Serine-Threonine Kinases; rac1 GTP-Binding Protein; ras GTPase-Activating Proteins; Reverse Transcriptase Polymerase Chain Reaction; Signal Transduction; Thymosin; Tumor Cells, Cultured; Up-Regulation

2011
Actin-sequestering protein, thymosin beta-4, is a novel hypoxia responsive regulator.
    Clinical & experimental metastasis, 2010, Volume: 27, Issue:8

    Angiogenesis is induced by soluble factors such as vascular endothelial growth factor (VEGF) released from tumor cells in hypoxia. It enhances solid tumor growth and provides an ability to establish metastasis at peripheral sites by tumor cell migration. Thymosin beta-4 (TB4) is an actin-sequestering protein to control cytoskeletal reorganization. Here, we investigated whether angiogenesis and tumor metastasis are dependent on hypoxia conditioning-induced TB4 expression in B16F10 melanoma cells. TB4 expression in B16F10 cells was increased by hypoxia conditioning in a time-dependent manner. In addition, we found an increase of angiogenesis and HIF-1α expression in TB4-transgenic (Tg) mice as compared to wildtype mice. When wound healing assay was used to assess in vitro tumor cell migration, hypoxia conditioning for 1 h enhanced B16F10 cell migration. When TB4 expression in B16F10 cells was inhibited by the infection with small hairpin (sh) RNA of TB4 cloned in lentiviral vector, tumor cell migration was retarded. In addition, hypoxia conditioning-induced tumor cell migration was reduced by the infection of lentiviral shRNA of TB4. HIF-1α stabilization and the expression of VEGF isoform 165 and 121 in hypoxia were also reduced by the infection of lentiviral shRNA of TB4 in B16F10 cells. We also found an increase of tumor growth and lung metastasis count in TB4-Tg mice as compared to wildtype mice. Collectively, hypoxia conditioning induced tumor cell migration by TB4 expression-dependent HIF-1α stabilization. It suggests that TB4 could be a hypoxia responsive regulator to control tumor cell migration in angiogenesis and tumor metastasis.

    Topics: Animals; Cell Hypoxia; Humans; Lung Neoplasms; Melanoma, Experimental; Mice; Mice, Transgenic; Neoplasm Metastasis; Neovascularization, Pathologic; Reverse Transcriptase Polymerase Chain Reaction; Thymosin; Tumor Cells, Cultured

2010
Elevation of intracellular cyclic AMP inhibits NF-kappaB-mediated thymosin beta4 expression in melanoma cells.
    Experimental cell research, 2009, Nov-15, Volume: 315, Issue:19

    Thymosin beta4 (Tbeta4) is a major actin-sequestering protein that has been implicated in the growth, survival, motility, and metastasis of certain tumors and is considered an indicator for malignant progression. Therefore, identifying compounds that can downregulate Tbeta4 expression is very important for the development of anti-cancer chemotherapies. In this study, we investigated the effects of elevated cAMP on Tbeta4 expression and the metastatic potential of murine B16 melanoma cells. In addition, we also dissected the mechanism underlying cAMP-mediated Tbeta4 suppression. We found that treatment with the cAMP-inducing compounds alpha-MSH (alpha-melanocyte stimulating hormone) and IBMX (3-isobutyl-1-methylxanthine) significantly suppressed Tbeta4 expression and regulated EMT-associated genes through the suppression of NF-kappaB activation in B16F10 cells. Along with decreased Tbeta4 expression, the in vitro invasiveness and anchorage-independent growth in a semi-solid agar of these cells were also inhibited. In animal experiments, the metastatic potential of the alpha-MSH- or IBMX-treated B16F10 melanoma cells was decreased compared to untreated control cells. Collectively, our data demonstrate that elevated intracellular cAMP significantly suppresses Tbeta4 expression and reduces MMP-9 activity, which leads to decreased metastatic potential. Moreover, suppression of NF-kappaB activation by alpha-MSH or IBMX is critical for inhibiting Tbeta4 expression.

    Topics: Animals; Cell Proliferation; Cells, Cultured; Cyclic AMP; Gene Expression Regulation, Neoplastic; Matrix Metalloproteinase 9; Melanoma; Melanoma, Experimental; Mice; Neoplasm Metastasis; NF-kappa B; Thymosin

2009
Overexpression of the thymosin beta-4 gene is associated with increased invasion of SW480 colon carcinoma cells and the distant metastasis of human colorectal carcinoma.
    Oncogene, 2004, Aug-26, Volume: 23, Issue:39

    Cell-matrix and cell-cell adhesive interactions play important roles in the normal organization and stabilization of the cell layer in epithelial tissue. Alterations in the expression and function of these adhesion systems that cause a switch to a migratory phenotype in tumor invasion and metastasis are critical for the malignant conversion of epithelial cells. Thymosin beta-4 (Tbeta-4) is the major actin-sequestering protein that has been shown to be upregulated in a wide variety of human carcinomas and has been implicated to be involved in altering the motility of certain tumors. We have recently demonstrated that the growth rate, colony formation in soft agar, and motility, all good indicators for malignant progression, of SW480 colon carcinoma cells are dramatically increased by enforced Tbeta-4 expression. To test the hypothesis that overexpression of this G-actin sequestering peptide also promotes tumor invasion, we examined not only the invasion capability of Tbeta-4-overexpressing SW480 cells, but also the expression levels of Tbeta-4 as well as several proteins that participate in different stages of tumor progression in matched samples of human primary colorectal adenocarcinoma and liver metastases from several patients. A marked increase on the invasiveness in Tbeta-4-overexpressing SW480 cells with increased levels and activity of matrix metalloproteinase-7 (MMP-7) was observed. Furthermore, the levels of Fas as well as the susceptibility to Fas ligand-mediated apoptosis in Tbeta-4-overexpressing cells were significantly decreased. Interestingly, the levels of Tbeta-4 mRNA, beta-catenin, c-Myc, and MMP-7 in metastatic liver lesions were relatively higher, whereas the levels of E-cadherin and Fas were significantly lower than those in the matched primary colorectal tumors. These results suggest that upregulation of Tbeta-4, by promoting the disruption of cell-cell adhesion and a consequential activation of the beta-catenin signaling, could be a key event in the acquisition of growth advantages as well as invasive phenotypes in human colorectal carcinomas.

    Topics: Cell Line, Tumor; Colorectal Neoplasms; Gene Expression; Humans; Neoplasm Invasiveness; Neoplasm Metastasis; RNA, Messenger; Thymosin

2004
MALAT-1, a novel noncoding RNA, and thymosin beta4 predict metastasis and survival in early-stage non-small cell lung cancer.
    Oncogene, 2003, Sep-11, Volume: 22, Issue:39

    Early-stage non-small cell lung cancer (NSCLC) can be cured by surgical resection, but a substantial fraction of patients ultimately dies due to distant metastasis. In this study, we used subtractive hybridization to identify gene expression differences in stage I NSCLC tumors that either did or did not metastasize in the course of disease. Individual clones (n=225) were sequenced and quantitative RT-PCR verified overexpression in metastasizing samples. Several of the identified genes (eIF4A1, thymosin beta4 and a novel transcript named MALAT-1) were demonstrated to be significantly associated with metastasis in NSCLC patients (n=70). The genes' association with metastasis was stage- and histology specific. The Kaplan-Meier analyses identified MALAT-1 and thymosin beta4 as prognostic parameters for patient survival in stage I NSCLC. The novel MALAT-1 transcript is a noncoding RNA of more than 8000 nt expressed from chromosome 11q13. It is highly expressed in lung, pancreas and other healthy organs as well as in NSCLC. MALAT-1 expressed sequences are conserved across several species indicating its potentially important function. Taken together, these data contribute to the identification of early-stage NSCLC patients that are at high risk to develop metastasis. The identification of MALAT-1 emphasizes the potential role of noncoding RNAs in human cancer.

    Topics: Carcinoma, Non-Small-Cell Lung; Gene Expression Profiling; Gene Expression Regulation, Neoplastic; Humans; In Situ Hybridization; Lung Neoplasms; Molecular Sequence Data; Neoplasm Metastasis; Neoplasm Staging; Predictive Value of Tests; Reference Values; Reverse Transcriptase Polymerase Chain Reaction; RNA, Untranslated; Survival Rate; Thymosin

2003
Thymosin beta4: a new molecular target for antitumor strategies.
    Journal of the National Cancer Institute, 2003, Nov-19, Volume: 95, Issue:22

    Topics: Actins; Animals; Antineoplastic Agents; Cell Physiological Phenomena; Cell Transformation, Neoplastic; Humans; Male; Melanoma, Experimental; Neoplasm Metastasis; Neovascularization, Pathologic; Prostatic Neoplasms; Thymosin; Up-Regulation

2003
Role of thymosin beta4 in tumor metastasis and angiogenesis.
    Journal of the National Cancer Institute, 2003, Nov-19, Volume: 95, Issue:22

    Expression of the small peptide thymosin beta4 is associated with angiogenesis induction, accelerated wound healing, and the metastatic potential of tumor cells. However, little is known about the mechanism(s) by which thymosin beta4 promotes metastasis.. Northern blot analysis and immunohistochemistry were used to examine thymosin beta4 expression in mouse melanoma B16 cell lines and in B16-F10 cells derived from metastatic mouse lung tumors, respectively. B16-F10 cells infected with adenoviruses containing a thymosin beta4 expression vector or an empty vector were injected subcutaneously and intravenously into C57BL/6 mice to evaluate tumor growth and metastatic potential, respectively. In vitro assays were used to study cell migration, invasion, matrix metalloproteinase activity, cell proliferation, and angiogenic activity of adenovirus-infected B16-F10 cells. Statistical significance of all results was analyzed by two-tailed Student's t tests.. Thymosin beta4 mRNA was expressed in primary cultured B16-F10 cells derived from lung metastases and in B16-F10 cells that had formed lung tumors after being injected into mice but not in the B16-F1, B16-F10, or B16-BL6 cell lines. The mean tumor sizes in mice 20 days after injection with B16-F10 cells infected with thymosin beta4-expressing adenovirus and with control adenovirus were 21.7 mm (95% confidence interval [CI] = 17.7 to 25.7 mm) and 13.3 mm (95% CI = 11.1 to 15.3 mm), respectively (difference = 8.4 mm; P =.036). The mean numbers of metastatic lung nodules in mice (n = 20) 2 weeks after intravenous injection with thymosin beta4-expressing adenovirus and with control adenovirus were 46.7 (95% CI = 35.0 to 57.7) and 10.9 (95% CI = 6.2 to 15.6), respectively (difference = 35.8 metastatic lung nodules, P<.001). Thymosin beta4 overexpression was associated with a mean 2.3-fold increase (95% CI = 1.9- to 2.7-fold increase; P<.001) in B16-F10 cell migration and a mean 4.4-fold increase (95% CI = 3.3- to 5.5-fold increase; P<.001) in the number of blood vessels in solid tumors derived from injected B16-F10 cells but had no effect on cell invasion, proliferation, or matrix metalloproteinase activity. This induction of angiogenesis by thymosin beta4 was associated with induction of vascular endothelial growth factor expression.. Thymosin beta4 may stimulate tumor metastasis by activating cell migration and angiogenesis.

    Topics: Adenoviridae; Animals; Cell Line, Tumor; Cell Movement; Fluorescent Antibody Technique; Gene Expression Regulation, Neoplastic; Lung Neoplasms; Melanoma, Experimental; Mice; Neoplasm Metastasis; Neovascularization, Pathologic; Recombinant Proteins; Thymosin; Up-Regulation

2003
Molecular switches in metastasis.
    Nature, 2000, Aug-03, Volume: 406, Issue:6795

    Topics: Animals; Fibronectins; Gene Expression Regulation, Neoplastic; Humans; Melanoma; Mice; Neoplasm Metastasis; Oligonucleotide Array Sequence Analysis; ras Proteins; rho GTP-Binding Proteins; rhoC GTP-Binding Protein; Thymosin; Tumor Cells, Cultured; Up-Regulation

2000
Genomic analysis of metastasis reveals an essential role for RhoC.
    Nature, 2000, Aug-03, Volume: 406, Issue:6795

    The most damaging change during cancer progression is the switch from a locally growing tumour to a metastatic killer. This switch is believed to involve numerous alterations that allow tumour cells to complete the complex series of events needed for metastasis. Relatively few genes have been implicated in these events. Here we use an in vivo selection scheme to select highly metastatic melanoma cells. By analysing these cells on DNA arrays, we define a pattern of gene expression that correlates with progression to a metastatic phenotype. In particular, we show enhanced expression of several genes involved in extracellular matrix assembly and of a second set of genes that regulate, either directly or indirectly, the actin-based cytoskeleton. One of these, the small GTPase RhoC, enhances metastasis when overexpressed, whereas a dominant-negative Rho inhibits metastasis. Analysis of the phenotype of cells expressing dominant-negative Rho or RhoC indicates that RhoC is important in tumour cell invasion. The genomic approach allows us to identify families of genes involved in a process, not just single genes, and can indicate which molecular and cellular events might be important in complex biological processes such as metastasis.

    Topics: Animals; Fibronectins; Gene Expression Profiling; Gene Expression Regulation, Neoplastic; Gene Transfer Techniques; Humans; Lung Neoplasms; Melanoma; Mice; Mice, Inbred C57BL; Mice, Nude; Mutation; Neoplasm Metastasis; Neoplasm Transplantation; Oligonucleotide Array Sequence Analysis; ras Proteins; rho GTP-Binding Proteins; rhoA GTP-Binding Protein; rhoC GTP-Binding Protein; Thymosin; Tumor Cells, Cultured

2000