transforming-growth-factor-beta has been researched along with Precursor-Cell-Lymphoblastic-Leukemia-Lymphoma* in 27 studies
4 review(s) available for transforming-growth-factor-beta and Precursor-Cell-Lymphoblastic-Leukemia-Lymphoma
Article | Year |
---|---|
CD4
Regulatory T-cells (Tregs) are a very important subtype of lymphocytes when it comes to self-control in the human immunological system. Tregs are decisive not only in the protection against destruction of own tissues by autoimmune immunocompetent cells but also in the immunological answer to developing cancers. On the other hand, Tregs could be responsible for the progression of acute and chronic leukemias. In our study, we review publications available in the PUMED database concerning acute leukemia, with a particular emphasis on child's leukemias. The percentage of regulatory T-lymphocytes in peripheral blood and bone marrow was elevated compared to those in healthy individuals and correlated with progressive disease. Regulatory T-cells taken from children diagnosed with leukemia showed a higher suppressive capability, which was confirmed by detecting elevated levels of secreted IL-10 and TGF-beta. The possibility of pharmacological intervention in the self-control of the immunological system is now under extensive investigation in many human cancers. Presumably, Treg cells could be a vital part of targeted therapies. Routine Treg determination could be used to assess the severity of disease and prognosis in children with acute lymphoblastic leukemia. This proposition results from the fact that in some studies, higher percentage of Treg cells in peripheral blood was demonstrated. However, observations confirming these facts are scarce; thus, extrapolating them to the population of children with hematological malignancies needs to be verified in additional studies. Topics: Disease Progression; Flow Cytometry; Forkhead Transcription Factors; Humans; Immunophenotyping; Interleukin-10; Leukemia, Myeloid, Acute; Precursor Cell Lymphoblastic Leukemia-Lymphoma; T-Lymphocytes, Regulatory; Transforming Growth Factor beta | 2019 |
[Acute lymphoblastic leukemia in a pediatric patient with Marfan's syndrome].
We report a rare case of acute lymphoblastic leukemia (ALL) in a 7-year-old boy with Marfan's syndrome. He was diagnosed as having Marfan's syndrome by clinical findings at the age of 2 years, and the diagnosis was confirmed by the detection of gene mutation in FBN1. He was referred to our hospital because of the swelling of cervical lymph nodes at the age of 7 years. Findings on bone marrow examination demonstrated T lymphoblastic ALL. He obtained complete remission after induction therapy, and had no serious side effects including cardiotoxicity during chemotherapy. He has remained in continuous complete remission for 34 months following diagnosis. To our knowledge, only three cases of leukemia in patients with Marfan's syndrome were reported previously. We speculate that increased activity of TGF-β, which is known as a tumor suppressor factor, in patients with Marfan's syndrome may diminish the risk of developing leukemia, although such a thesis was not proven in this case. Topics: Antineoplastic Combined Chemotherapy Protocols; Child; Fibrillin-1; Fibrillins; Humans; Male; Marfan Syndrome; Microfilament Proteins; Mutation; Precursor Cell Lymphoblastic Leukemia-Lymphoma; Remission Induction; Transforming Growth Factor beta | 2011 |
[The molecular mechanism of Notch-induced transformation and the therapeutic potential of its inhibitor].
Topics: Alanine; Amyloid Precursor Protein Secretases; Azepines; Cytoplasm; gamma-Aminobutyric Acid; Humans; Immunoglobulin J Recombination Signal Sequence-Binding Protein; Precursor Cell Lymphoblastic Leukemia-Lymphoma; Receptors, Notch; Signal Transduction; Transforming Growth Factor beta; Triglycerides | 2006 |
P15INK4b gene methylation and myelodysplastic syndromes.
Myelodysplastic syndromes (MDS) are clonal disorders, which frequently undergo leukemic transformation. It was recently shown that the promoter of the p15INK4b but not the p16INK4a gene is frequently and selectively hypermethylated in MDS. The p15INK4b gene is a cyclin dependent kinase inhibitor gene, which is actively transcribed after TGFbeta exposure. Methylation of the p15INK4b gene is significantly correlated with blastic bone marrow involvement, and sequential analyses have shown that methylation increases with disease evolution toward AML. These data strongly suggest that p15INK4b gene methylation is a mechanism allowing leukemic cells to escape to inhibitory signals from the bone marrow environment, however the exact role of p15INK4b gene methylation in disruption of the signal mediated by TGFbeta remains to be investigated. Topics: Acute Disease; Animals; Antimetabolites, Antineoplastic; Azacitidine; Bone Marrow; Carrier Proteins; Cell Cycle; Cell Cycle Proteins; Cell Differentiation; Cell Transformation, Neoplastic; Chromosomes, Human, Pair 9; Clinical Trials, Phase II as Topic; Cyclin-Dependent Kinase Inhibitor p15; Cyclin-Dependent Kinase Inhibitor p16; Cyclin-Dependent Kinases; Decitabine; Disease Progression; DNA Methylation; Genes, p16; Genes, Tumor Suppressor; Hematopoiesis; Humans; Leukemia, Myeloid; Mice; Myelodysplastic Syndromes; Neoplasm Proteins; Precancerous Conditions; Precursor Cell Lymphoblastic Leukemia-Lymphoma; Transforming Growth Factor beta; Tumor Suppressor Proteins | 1999 |
23 other study(ies) available for transforming-growth-factor-beta and Precursor-Cell-Lymphoblastic-Leukemia-Lymphoma
Article | Year |
---|---|
Photobiomodulation Therapy Alleviates Tissue Fibroses Associated with Chronic Graft-Versus-Host Disease: Two Case Reports and Putative Anti-Fibrotic Roles of TGF-β.
Patients who receive allogeneic hematopoietic stem cell transplantation may experience oral complications due to chronic graft-versus-host disease (cGVHD). The manifestations may include progressive sclerosis-like changes that may involve various body sites, including the oropharynx.. We present two cGVHD cases of oropharyngeal fibrotic changes that affected functions that were treated with photobiomodulation (PBM) therapy. These case reports suggest that PBM therapy represents an additional, innovative approach affecting discrete phases in cGVHD-associated fibrotic changes.. We discuss these observations in the context of currently understood molecular mechanisms, especially induction of transforming growth factor beta and NFκB that appear to be counter-intuitive to their known roles in matrix synthesis and inflammation that contribute to tissue fibroses. The clinical benefit noted in the two cases presented clearly indicates that there are distinct mechanistic and biological insights in the regulation of these molecular pathways in determining therapeutic efficacy with PBM therapy. Topics: Adult; Child; Chronic Disease; Female; Fibrosis; Follow-Up Studies; Graft vs Host Disease; Hematopoietic Stem Cell Transplantation; Humans; Leukemia, Myeloid, Acute; Low-Level Light Therapy; Male; Precursor Cell Lymphoblastic Leukemia-Lymphoma; Radiotherapy Dosage; Sampling Studies; Time Factors; Transforming Growth Factor beta; Treatment Outcome | 2018 |
ChiPPI: a novel method for mapping chimeric protein-protein interactions uncovers selection principles of protein fusion events in cancer.
Fusion proteins, comprising peptides deriving from the translation of two parental genes, are produced in cancer by chromosomal aberrations. The expressed fusion protein incorporates domains of both parental proteins. Using a methodology that treats discrete protein domains as binding sites for specific domains of interacting proteins, we have cataloged the protein interaction networks for 11 528 cancer fusions (ChiTaRS-3.1). Here, we present our novel method, chimeric protein-protein interactions (ChiPPI) that uses the domain-domain co-occurrence scores in order to identify preserved interactors of chimeric proteins. Mapping the influence of fusion proteins on cell metabolism and pathways reveals that ChiPPI networks often lose tumor suppressor proteins and gain oncoproteins. Furthermore, fusions often induce novel connections between non-interactors skewing interaction networks and signaling pathways. We compared fusion protein PPI networks in leukemia/lymphoma, sarcoma and solid tumors finding distinct enrichment patterns for each disease type. While certain pathways are enriched in all three diseases (Wnt, Notch and TGF β), there are distinct patterns for leukemia (EGFR signaling, DNA replication and CCKR signaling), for sarcoma (p53 pathway and CCKR signaling) and solid tumors (FGFR and EGFR signaling). Thus, the ChiPPI method represents a comprehensive tool for studying the anomaly of skewed cellular networks produced by fusion proteins in cancer. Topics: Gene Expression Regulation, Neoplastic; Humans; Metabolic Networks and Pathways; Oncogene Proteins, Fusion; Precursor Cell Lymphoblastic Leukemia-Lymphoma; Protein Interaction Domains and Motifs; Protein Interaction Mapping; Protein Interaction Maps; Receptors, Notch; Sarcoma; Signal Transduction; Software; Transforming Growth Factor beta; Wnt Proteins | 2017 |
TGFβ and IL10 have an impact on risk group and prognosis in childhood ALL.
Cytokines and their genes have been described to have an influence on incidence and prognosis in malignant, infectious and autoimmune disease. We previously described the impact of cytokine production on prognosis in paediatric standard-risk acute lymphoblastic leukaemia (ALL).. In this study, we investigated the influence of cytokine gene polymorphisms (TNFα, TGFβ, IL10 and IFNγ) on frequency, risk group and prognosis in 95 paediatric ALL-patients. We further report on intracellular production of these cytokines in T-cells.. IL10 high-producer-haplotypes were reduced in ALL-patients compared with healthy controls and resulted in a reduced relapse rate compared with low-producer haplotypes. TGFβ high-producer-haplotypes were correlated with a high initial blast-count (codon 25: G/G) and were elevated in high-risk ALL-patients (codon 10: T/T). IL10 was positively and IFNγ-production was negatively correlated with initial blast-count. At diagnosis the expression of TNFα and IFNγ was reduced in patients compared with healthy controls. This was more pronounced in high-risk and in T-ALL-patients.. We conclude that gene-polymorphisms of the regulatory/anti-inflammatory cytokines, TGFβ and IL10, but not of the pro-inflammatory cytokines, IFNγ and TNFα, have an impact on prognosis and risk-group of ALL. However, the reduced capacity to produce pro-inflammatory cytokines at diagnosis may serve as another important, functional risk factor. These data may help in further risk stratification and adaptation of therapy-intensity in paediatric patients with ALL. Topics: Case-Control Studies; Child; Female; Flow Cytometry; Follow-Up Studies; Genotype; Haplotypes; Humans; Interferon-gamma; Interleukin-10; Male; Neoplasm Recurrence, Local; Neoplasm Staging; Polymerase Chain Reaction; Polymorphism, Single Nucleotide; Precursor Cell Lymphoblastic Leukemia-Lymphoma; Prognosis; Survival Rate; Transforming Growth Factor beta; Tumor Necrosis Factor-alpha | 2015 |
Binding of a Smad4/Ets-1 complex to a novel intragenic regulatory element in exon12 of FPGS underlies decreased gene expression and antifolate resistance in leukemia.
Polyglutamylation of antifolates catalyzed by folylpoly-γ-glutamate synthetase (FPGS) is essential for their intracellular retention and cytotoxic activity. Hence, loss of FPGS expression and/or function results in lack of antifolate polyglutamylation and drug resistance. Members of the TGF-β/Smad signaling pathway are negative regulators of hematopoiesis and deregulation of this pathway is considered a major contributor to leukemogenesis. Here we show that FPGS gene expression is inversely correlated with the binding of a Smad4/Ets-1 complex to exon12 of FPGS in both acute lymphoblastic leukemia cells and acute myeloid leukemia blast specimens. We demonstrate that antifolate resistant leukemia cells harbor a heterozygous point mutation in exon12 of FPGS which disrupts FPGS activity by abolishing ATP binding, and alters the binding pattern of transcription factors to the genomic region of exon12. This in turn results in the near complete silencing of the wild type allele leading to a 97% loss of FPGS activity. We show that exon12 is a novel intragenic transcriptional regulator, endowed with the ability to drive transcription in vitro, and is occupied by transcription factors and chromatin remodeling agents (e.g. Smad4/Ets-1, HP-1 and Brg1) in vivo. These findings bear important implications for the rational overcoming of antifolate resistance in leukemia. Topics: Adult; Aged; Base Sequence; Binding Sites; Cell Line, Tumor; Chromobox Protein Homolog 5; Chromosomal Proteins, Non-Histone; DNA Helicases; DNA Methylation; Drug Resistance, Neoplasm; Female; Folic Acid Antagonists; Gene Silencing; Humans; Leukemia, Myeloid, Acute; Male; Middle Aged; Nuclear Proteins; Peptide Synthases; Polymorphism, Single Nucleotide; Precursor Cell Lymphoblastic Leukemia-Lymphoma; Protein Binding; Proto-Oncogene Protein c-ets-1; Sequence Analysis, DNA; Smad4 Protein; Transcription Factors; Transcription, Genetic; Transforming Growth Factor beta; Young Adult | 2014 |
[Expression level of Th22 cells and its cytokines in patients with acute lymphoblastic leukemia and its significance].
This study was purposed to analyze the expression level of Th22 cells and their cytokines in patients with acute lymphoblastic leukemia (ALL) and evaluate its significance. Forty-eight patients with ALL were selected. According to the treatment, all patients were divided into the newly diagnosed group (n = 26) and complete remission (CR) group (n = 22). The proportion of Th22 cells in peripheral blood was detected by flow cytometry (FCM). The expression levels of cytokines IL-22, IL-6, TNF-α and TGF-β in peripheral blood were measured by ELISA. The expression level of IL-22 mRNA in peripheral blood mononuclear cells was examined by semi-quantitative-reverse transcription PCR (RT-PCR). Meanwhile, 30 healthy individuals were selected as a control group. The parameters of the 3 groups were compared. The results showed that the percentage of Th22 cells and the expression levels of IL-22, IL-6, TNF-α and IL-22 mRNA in newly diagnosed group and the CR group were significantly lower than that in control group, the expression level of TGF-β in above mentioned two group was obviously higher than that in control group (P < 0.05). The percentage of Th22 cells and the expression levels of IL-22, IL-6, TNF-α and IL-22 mRNA in newly diagnosed group were evidently lower than that in CR group (P < 0.05), but the expression level of TGF-β in newly diagnosed group obviously higher than that in CR group. The expression level of IL-22 in newly diagnosed group was positively related with expression level of IL-6 and TNF-α, but it was negatively related with expression level of TGF-β. It is concluded that the decreasing of Th22 cells and down-regulation of IL-22 expression level may be related with pathogenesis of ALL, the decreasing of Th22 cells is risk factor for ALL. Topics: Adult; Aged; Case-Control Studies; Female; Humans; Interleukin-22; Interleukin-6; Interleukins; Middle Aged; Precursor Cell Lymphoblastic Leukemia-Lymphoma; Remission Induction; RNA, Messenger; T-Lymphocytes, Helper-Inducer; Transforming Growth Factor beta; Tumor Necrosis Factor-alpha; Young Adult | 2013 |
A case-control study of childhood acute lymphoblastic leukaemia and polymorphisms in the TGF-beta and receptor genes.
Inherited genetic variants in critical genes can putatively modulate susceptibility to childhood acute lymphoblastic leukemia (ALL).. We used allelic discrimination method to genotype 19 polymorphisms in the transforming growth factor-beta1 (TGF-beta1), transforming growth factor-beta receptor 1 (TGF-betaR1) and transforming growth factor-beta receptor 2 (TGF-betaR2) genes in 460 cases of childhood acute ALL and 552 ethnically matched controls. The genotyped polymorphisms included functional and tagging variants to cover the three genes in entirety. We used multidimensionality reduction (MDR) method to test effect of multiple genes on disease susceptibility. In order to increase statistical power and detect susceptibility variants not directly genotyped in this study, we used imputation using HapMap data.. None of the genotyped polymorphisms or the consequent haplotypes showed any association with risk modulation. The results, however, did show a marginal association (odds ratio OR 0.76, 95% confidence interval CI 0.59-0.97) of the variant allele for the rs10417924 polymorphism located at 3'untranslated region of the TGF-beta1 gene with the B-cell lineage ALL. No other polymorphism showed any association with childhood ALL susceptibility. A signal of marginal significance for the rs10417924 polymorphism in the TGF-beta1 gene in B-cell lineage ALL showed up with both MDR and imputation techniques.. These data rule out the role of polymorphisms in the TGF-beta1, TGF-betaR1 and TGF-betaR2 genes in susceptibility to childhood ALL. However, for B-lineage ALL, the role of the rs10417924 polymorphism in TGF-beta1 gene needs further investigation. Topics: Adult; B-Lymphocytes; Case-Control Studies; Child; Female; Haplotypes; Humans; Male; Polymorphism, Single Nucleotide; Precursor Cell Lymphoblastic Leukemia-Lymphoma; Prognosis; Protein Serine-Threonine Kinases; Receptor, Transforming Growth Factor-beta Type I; Receptor, Transforming Growth Factor-beta Type II; Receptors, Transforming Growth Factor beta; Transforming Growth Factor beta | 2009 |
The TEL-AML1 leukemia fusion gene dysregulates the TGF-beta pathway in early B lineage progenitor cells.
Chromosome translocation to generate the TEL-AML1 (also known as ETV6-RUNX1) chimeric fusion gene is a frequent and early or initiating event in childhood acute lymphoblastic leukemia (ALL). Our starting hypothesis was that the TEL-AML1 protein generates and maintains preleukemic clones and that conversion to overt disease requires secondary genetic changes, possibly in the context of abnormal immune responses. Here, we show that a murine B cell progenitor cell line expressing inducible TEL-AML1 proliferates at a slower rate than parent cells but is more resistant to further inhibition of proliferation by TGF-beta. This facilitates the competitive expansion of TEL-AML1-expressing cells in the presence of TGF-beta. Further analysis indicated that TEL-AML1 binds to a principal TGF-beta signaling target, Smad3, and compromises its ability to activate target promoters. In mice expressing a TEL-AML1 transgene, early, pre-pro-B cells were increased in number and also showed reduced sensitivity to TGF-beta-mediated inhibition of proliferation. Moreover, expression of TEL-AML1 in human cord blood progenitor cells led to the expansion of a candidate preleukemic stem cell population that had an early B lineage phenotype (CD34+CD38-CD19+) and a marked growth advantage in the presence of TGF-beta. Collectively, these data suggest a plausible mechanism by which dysregulated immune responses to infection might promote the malignant evolution of TEL-AML1-expressing preleukemic clones. Topics: Animals; Cell Line; Cell Proliferation; Core Binding Factor Alpha 2 Subunit; Cyclin-Dependent Kinase Inhibitor p27; Fetal Blood; Gene Expression; Hematopoiesis; Humans; Mice; Models, Biological; Neoplastic Stem Cells; Oncogene Fusion; Oncogene Proteins, Fusion; Precursor Cell Lymphoblastic Leukemia-Lymphoma; Precursor Cells, B-Lymphoid; Signal Transduction; Smad Proteins; Stem Cells; Transfection; Transforming Growth Factor beta | 2009 |
Bone marrow stroma cells regulate TIEG1 expression in acute lymphoblastic leukemia cells: role of TGFbeta/BMP-6 and TIEG1 in chemotherapy escape.
The bone marrow microenvironment regulates early B lymphopoiesis and protects leukemia cells against chemotherapy treatment, thus the microenvironment may serve as a sanctuary site for these cells. Yet, few factors that contribute to this process are known. We have explored the role of transforming growth factor beta (TGFbeta) and bone morphogenetic protein-6 (BMP-6) and one target gene, TGFbeta inducible early gene 1 (TIEG1), in the communication between stroma cells and acute lymphoblastic leukemia (ALL) cell lines and their escape from chemotherapy. Here, we have demonstrated TIEG1 expression in both normal B progenitor cells and ALL cells, which increased rapidly upon TGFbeta and BMP-6 treatment. Stimulation with TGFbeta or BMP-6, as well as overexpression of TIEG1 inhibited proliferation. Furthermore, interaction with stroma cells induced TIEG1 expression in ALL cells, inhibited their proliferation and protected the cells against chemotherapeutic treatment. Similarly, treatment with TGFbeta or BMP-6, as well as overexpression of TIEG1, protected ALL cells against chemotherapy-induced cell death. These data suggest that TGFbeta and BMP-6 in the bone marrow microenvironment allow leukemia cells to escape therapy. Further, the data indicate that TIEG1 might be involved in mediating this effect from the microenvironment onto the leukemia cells. Topics: Antineoplastic Agents; Blotting, Northern; Blotting, Western; Bone Marrow; Bone Morphogenetic Protein 6; Cell Line, Tumor; Drug Screening Assays, Antitumor; Early Growth Response Transcription Factors; Gene Expression Regulation, Neoplastic; Humans; Kruppel-Like Transcription Factors; Microarray Analysis; Polymerase Chain Reaction; Precursor Cell Lymphoblastic Leukemia-Lymphoma; Transforming Growth Factor beta; Up-Regulation | 2008 |
Immunosuppressive properties of mesenchymal stem cells derived from bone marrow of patient with hematological malignant diseases.
Mesenchymal stem cells (MSCs) have received much attention because of their capabilities of differentiating into multiple mesenchymal lineages and supporting hematopoiesis. Recently, MSCs have gained further interests after the demonstration of an immunosuppressive role. However, it's still unclear whether the immunosuppressive capability of MSCs will be altered with disease state. In this study, our results showed that MSCs derived from patients with lymphoblastic leukemia (ALL), Hodgkin disease (HD), and non-Hodgkin lymphoma (NHL) capable of suppressing the proliferation of T-lymphocyte stimulated in a mixed-lymphocyte reaction (MLR). The immunosuppressive effect of MSCs derived from ALL, HD and NHL on T-cell proliferation was dose-dependent. The supernatants of MSCs derived from ALL, HD and NHL had effect on T-cell proliferation. By using neutralising monoclonal antibodies, we found that transforming growth factor beta1 (TGFbeta1) and hepatocyte growth factor were major mediators of T-cell suppression by MSCs derived from ALL, HD and NHL. Although MSCs derived from patients with myelodysplastics syndromes (MDS) could inhibit T-cell proliferation stimulated with mitogen or in MLR, the inhibitory effect of MDS-MSCs was impaired. However, adherent cells derived from patients with acute myeloid leukemia (AML) showed abnormal immunomodulatory functions. Adherent cells derived from AML failed to suppress the proliferation of T-cell stimulated in MLR. Topics: Bone Marrow Cells; Hematologic Neoplasms; Hepatocyte Growth Factor; Hodgkin Disease; Humans; Immunologic Factors; Immunosuppression Therapy; Lymphocyte Culture Test, Mixed; Lymphoma, Non-Hodgkin; Mesenchymal Stem Cells; Precursor Cell Lymphoblastic Leukemia-Lymphoma; T-Lymphocytes; Transforming Growth Factor beta | 2008 |
Expression of angiogenic factors in childhood B-cell precursor acute lymphoblastic leukemia.
Pathological angiogenesis is increasingly recognized to be an important feature of pathogenesis in solid tumors and also in leukemias. Specific blockers of angiogenesis are now being introduced into early clinical trials with encouraging results. Vascular endothelial growth factor (VEGF) seems to play a central role in tumor angiogenesis and is associated with a poor prognosis in both solid tumors and adult leukemias. In pediatric acute lymphocytic leukemia however, the expression of angiogenic molecules and its relation to prognosis and relapse are unknown. Therefore, we prospectively analyzed 46 pediatric patients with precursor B cell acute lymphocytic leukemia by semi-quantitative RT-PCR for expression of the angiogenic molecules VEGF, VEGF-C, iNOS and TGF-beta and correlated relapse and survival data with the expression of these factors. We found a high mRNA expression of TGF-beta and iNOS, a moderate expression of VEGF but no expression of bFGF and VEGF-C. A significantly higher expression of VEGF mRNA was found in patients with late relapses compared to patients without relapses (p=0.043). A significantly higher mRNA expression of iNOS was found in surviving patients compared with non-surviving patients (p=0.023). Angiogenic factors are expressed in the bone marrow of patients with pediatric B cell precursor ALL and VEGF is a potential candidate for therapeutic intervention as it is significantly higher expressed in children with late relapses. The mRNA expression of iNOS in the surviving children possibly reflects an increased activity of the immune system against the leukemia which leads to a superior survival. Topics: Adolescent; Angiogenic Proteins; Bone Marrow Cells; Burkitt Lymphoma; Child; Child, Preschool; Female; Fibroblast Growth Factor 2; Humans; Infant; Male; Neovascularization, Pathologic; Nitric Oxide Synthase Type II; Precursor Cell Lymphoblastic Leukemia-Lymphoma; Reverse Transcriptase Polymerase Chain Reaction; RNA, Messenger; Transcription, Genetic; Transforming Growth Factor beta; Vascular Endothelial Growth Factor A; Vascular Endothelial Growth Factor C | 2007 |
Unique human CD133+ leukemia cell line and its modulation towards a mesenchymal phenotype by FGF2 and TGFbeta1.
Immunological features of GM-490 cells, a new blood cell line from a patient with acute lymphoblastic leukemia, included lack of CD34, CD38, CD45, CD14, HLA-DR, and lymphoid and myeloid markers and expression of CD29, CD36, CD44, CD54, CD71, CD105, and CD133. Molecular analysis indicated CD45 gene expression was absent but CD34 mRNA was present. GM-490 cells constitutively produced fibronectin (FN), type III and traces of type I collagen, collagenases, glycosaminoglycans (GAG) and biglycan and betaglycan proteoglycans (PG) as well as FGF2 and TGFbeta1. When FGF2 and/or TGFbeta1 were added to cells in vitro, they stimulated cell proliferation and differently modulated matrix production and growth factor receptor expression. Reverse transcription-polymerase chain reaction (RT-PCR) detection of transcripts encoding for osteocalcin and RUNX2 suggests GM-490 cells differentiate towards the osteoblast pathway. GM-490 cells expressed the low affinity nerve growth factor receptor (p75LNGFR), a somatic stem cell marker that is not detected in hematopoietic cells, leading to the hypothesis that GM-490 has mesenchymal stem cell properties. The reciprocal modulating effects of FGF2 and TGFbeta1 on each other's receptors make the GM-490 cell line a new model for investigating the relationship between these growth factors and their receptors in autocrine loops which are believed to sustain the malignant clone in hematological diseases. Topics: AC133 Antigen; Animals; Antigens, CD; Bone Marrow Cells; Cell Differentiation; Cell Line, Tumor; Cell Proliferation; Collagen; Collagenases; Colony-Forming Units Assay; Core Binding Factor Alpha 1 Subunit; Fibroblast Growth Factor 2; Fibronectins; Glycoproteins; Glycosaminoglycans; Humans; Mesenchymal Stem Cells; Osteocalcin; Peptides; Precursor Cell Lymphoblastic Leukemia-Lymphoma; Receptor, Fibroblast Growth Factor, Type 2; Receptor, Nerve Growth Factor; RNA, Messenger; Transforming Growth Factor beta | 2006 |
[Identification of the isoform in type II receptor of transforming growth factor-beta in patients with acute leukemia and its clinical significance].
Recent research indicates that TGF-beta and type II receptor (TbetaR-II) play an important role in the pathogenesis of tumor. A high frequency of abnormalities in TbetaR-II has been demonstrated in various cancers. To identify the mutation of TbetaR-II in patients with acute leukemia, the bone marrow samples from 6 patients with acute leukemia and 11 normal individuals as control were detected by long-range RT-PCR. To detect a deletion in sequence of the TbetaR-IIgene, the PCR products were cloned to T vector and then sequenced. The results showed that there was existance of the isoform of TbetaR-II in 2 cases out of 6 patients with acute leukemia. These two patients had more poor prognosis than others. In conclusion, there was the isoform of TbetaR-II in partial patients with acute leukemia, and the isoform may be related with prognosis. Topics: Adult; Base Sequence; Female; Humans; Leukemia, Myeloid, Acute; Male; Middle Aged; Molecular Sequence Data; Precursor Cell Lymphoblastic Leukemia-Lymphoma; Prognosis; Protein Isoforms; Protein Serine-Threonine Kinases; Receptor, Transforming Growth Factor-beta Type II; Receptors, Transforming Growth Factor beta; Signal Transduction; Transforming Growth Factor beta | 2006 |
[Expression of Smad4 in leukemia cells].
Loss of transforming growth factor (TGF)-beta signaling has been implicated in malignant transformation of various tissues. Smad4 plays a central role in the signal transduction of TGF-beta. Deletion or mutation of Smad4 has been described in a number of cancers. This study was aimed to investigate a potential role of Smad4 in leukemia including its expression and location in blast cells. The mononuclear cells were separated from bone marrow of leukemia patients. The samples, blast cells of which were more than 90% in mononuclear cells, were selected. The expression and location of Smad4 protein were analyzed by immunohistochemistry methods. The results showed that the Smad4 protein located mainly in nucleus, part of this protein located in cytoplasma, the expressions of Smad4 were not detected in 6 out of 9 ALL patients, in 7 out of 24 AML patients and in 1 out of 2 CML patients; these leukemia patients, in whose cells the expression of Smad4 was not detected, included one L1 and one L3, four L2, one M0, one M1, two M2a, one M3a, one M4b, one M6 and one CML. In conclusion, the Smad4 protein was mainly in nucleus, the deletion or functional change of Smad4 may related with the pathogenesis of human AML. Topics: Humans; Leukemia, Myeloid, Acute; Precursor Cell Lymphoblastic Leukemia-Lymphoma; Signal Transduction; Smad4 Protein; Transforming Growth Factor beta | 2006 |
Bone marrow stroma damage induced by chemotherapy for acute lymphoblastic leukemia in children.
Several studies have suggested a role of bone marrow stroma injury in long-term chemotherapy-induced hematopoietic failure. To evaluate whether bone marrow microenvironment is altered by chemotherapy for acute lymphoblastic leukemia (ALL) and to determine its contribution to postchemotherapy anemia, we investigated the ability of stroma from children receiving maintenance chemotherapy for ALL to support hematopoiesis. Long-term bone marrow cultures (LTBMC) were established with bone marrow cells either from ALL children under therapy (n = 24) or from control subjects (n = 19). Nonadherent cells and colony forming units-granulocytic monocytic (CFU-GM) output in LTBMC did not differ between patients and controls. In contrast, burst forming unit-erythroid (BFU-E) numbers were lower in patient LTBMC (p = 0.013). Co-cultures of normal CD34+ cells and preformed patient or control stromas showed significantly reduced hematopoietic supportive capabilities of patient stromas: both CFU-GM and BFU-E were reduced (p = 0.002 and 0.046, respectively). In addition, supernatants (SN) of patients' LTBMC inhibited normal BFU-E growth compared with SN of normal LTBMC. Transforming growth factor (TGF)-beta1 levels were increased in patient cultures (p = 0.0039) and inversely correlated with BFU-E produced in LTBMC (r = -0.36, p = 0.04). Neutralization of TGF-beta1 significantly increased the BFU-E output of patient LTBMC (p = 0.0078). In contrast, macrophage inflammatory peptide (MIP)-1alpha levels were lower in SN of patients compared with controls (p = 0.015). Thus, chemotherapy for ALL induces functional deregulation within bone marrow stromal cells with an increase in the growth-inhibiting factor TGF-beta1, together with a decrease in MIP-1alpha, which might contribute to hematopoietic toxicity. Topics: Antibodies; Antigens, CD34; Antimetabolites, Antineoplastic; Antineoplastic Combined Chemotherapy Protocols; Bone Marrow Cells; Cells, Cultured; Chemokine CCL3; Chemokine CCL4; Child; Chronic Disease; Coculture Techniques; Cytokines; Erythroid Precursor Cells; Humans; Macrophage Inflammatory Proteins; Mercaptopurine; Methotrexate; Myeloid Progenitor Cells; Precursor Cell Lymphoblastic Leukemia-Lymphoma; Stromal Cells; Transforming Growth Factor beta; Transforming Growth Factor beta1 | 2004 |
TGF-beta signaling, tumor suppression, and acute lymphoblastic leukemia.
Topics: DNA-Binding Proteins; Gene Expression; Genes, myc; Genes, Tumor Suppressor; Precursor Cell Lymphoblastic Leukemia-Lymphoma; Protein Serine-Threonine Kinases; Receptor, Transforming Growth Factor-beta Type II; Receptors, Transforming Growth Factor beta; Signal Transduction; Smad Proteins; Smad3 Protein; T-Lymphocytes; Trans-Activators; Transcription Factors; Transforming Growth Factor beta; Transforming Growth Factor beta1 | 2004 |
Loss of Smad3 in acute T-cell lymphoblastic leukemia.
The receptors for transforming growth factor beta (TGF-beta) and their signaling intermediates make up an important tumor-suppressor pathway. The role of one of these intermediates--Smad3--in the pathogenesis of lymphoid neoplasia is unknown.. We measured Smad3 messenger RNA (mRNA) and protein in leukemia cells obtained at diagnosis from 19 children with acute leukemia, including 10 with T-cell acute lymphoblastic leukemia (ALL), 7 with pre-B-cell ALL, and 2 with acute nonlymphoblastic leukemia (ANLL). All nine exons of the SMAD3 gene (MADH3) were sequenced. Mice in which one or both alleles of Smad3 were inactivated were used to evaluate the role of Smad3 in the response of normal T cells to TGF-beta and in the susceptibility to spontaneous leukemogenesis in mice in which both alleles of the tumor suppressor p27Kip1 were deleted.. Smad3 protein was absent in T-cell ALL but present in pre-B-cell ALL and ANLL. No mutations were found in the MADH3 gene in T-cell ALL, and Smad3 mRNA was present in T-cell ALL and normal T cells at similar levels. In mice, the loss of one allele for Smad3 impairs the inhibitory effect of TGF-beta on the proliferation of normal T cells and works in tandem with the homozygous inactivation of p27Kip1 to promote T-cell leukemogenesis.. Loss of Smad3 protein is a specific feature of pediatric T-cell ALL. A reduction in Smad3 expression and the loss of p27Kip1 work synergistically to promote T-cell leukemogenesis in mice. Topics: Adult; Animals; Cell Cycle Proteins; Child; Cyclin-Dependent Kinase Inhibitor p27; DNA-Binding Proteins; Exons; Gene Deletion; Gene Expression; Humans; Interleukin-2; Leukemia-Lymphoma, Adult T-Cell; Leukemia, Myeloid, Acute; Leukemia, T-Cell; Mice; Mice, Knockout; Precursor Cell Lymphoblastic Leukemia-Lymphoma; Receptors, Transforming Growth Factor beta; RNA, Messenger; Sequence Analysis, DNA; Signal Transduction; Smad3 Protein; T-Lymphocytes; Trans-Activators; Transforming Growth Factor beta; Tumor Suppressor Proteins | 2004 |
mRNA for chosen pro- and anti-inflammatory cytokines in T-lymphocytes in paediatric leukemias and lymphomas--a preliminary report.
We assessed mRNA for chosen pro- and anti-inflammatory cytokines in T-lymphocytes of peripheral blood in paediatric patients with leukemias and lymphomas. Levels of four different cytokine mRNAs (IFN-gamma, IL-10, IL-4, TGF-beta) were determined by the real-time PCR technique. In the whole examined group, at the time of diagnosis, we noted lower amounts of mRNA for TGF-beta1, comparing to respective values in the control patients. In the ALL group, we observed the following: 1) at the time of diagnosis: lower amounts of mRNA for IL-4 and for TGF-beta1, comparing to respective values in the control group; 2) lower amounts of mRNA for IL-10 after remission induction, comparing to the time of diagnosis. In our opinion, "immunedysregulation" in lymphoproliferative diseases in children is not caused by IFN-gamma deficiency. The deficit of anti-inflammatory cytokines, i.e., IL-4, TGF-beta, with higher amounts of IL-10, suggests their role in cancer development. Topics: CD4-Positive T-Lymphocytes; CD8-Positive T-Lymphocytes; Cytokines; Hodgkin Disease; Humans; Interleukin-10; Lymphoma; Lymphoma, Non-Hodgkin; Polymerase Chain Reaction; Precursor Cell Lymphoblastic Leukemia-Lymphoma; Remission Induction; RNA, Messenger; T-Lymphocytes; Transforming Growth Factor beta | 2004 |
[An analysis of sclerodermatous graft-versus-host-disease after allogeneic bone marrow transplantation: CD8+CD57+T-cell proliferation and increased production of TGF-beta].
A 19-year-old woman with acute lymphoblastic leukemia received an allogeneic bone marrow transplantation (BMT) from an HLA-identical sibling during the second remission, on September 28, 1993. The conditioning regimen consisted of total body irradiation and cyclophosphamide. Short term methotrexate and cyclosporin A were given for prophylaxis of graft-versus-host disease (GVHD). On day 771 after BMT, she complained of bilateral forearm pain, and developed sclerotic lesions on the skin of the abdominal wall, forearms and legs. The diagnosis of sclerodermatous GVHD was established by skin biopsy on day 834. The values of CRP and IgG were elevated, and both antinuclear antibody and anti-DNA antibody became positive. Flow cytometric analysis showed a significant increase in the number of CD57+ cells after appearance of sclerotic change. In addition, 65% of CD8+ cells were positive for CD57. Circulating level of transforming growth factor (TGF)-beta 1 was high. These results suggest that overproduction of CD8+ CD57+ T cells and high level of circulating TGF-beta are related to the development of sclerodermatous GVHD. Topics: Adult; Bone Marrow Transplantation; CD57 Antigens; CD8-Positive T-Lymphocytes; Cell Division; Female; Graft vs Host Disease; Humans; Precursor Cell Lymphoblastic Leukemia-Lymphoma; Scleroderma, Systemic; Transforming Growth Factor beta; Transplantation, Homologous | 1998 |
Microsatellite instability and frameshift mutations in BAX and transforming growth factor-beta RII genes are very uncommon in acute lymphoblastic leukemia in vivo but not in cell lines.
Mutations in the DNA mismatch repair (MMR) system lead to an instability of simple repetitive DNA sequences involved in several cancer types. This instability is reflected in a high mutation rate of microsatellites, and recent studies in colon cancer indicate that defects in MMR result in frequent frameshift mutations in mononucleotide repeats located in the coding regions of BAX and transforming growth factor-beta (TGF-beta) receptor genes. Circumstantial evidence suggests that the MMR defect may be involved in some lymphoid malignancies, although several allelotype analyses have concluded on the low level of microsatellite instability in acute lymphoblastic leukemias. To further evaluate the implication of MMR defects in leukemogenesis, we have studied a series of 98 children with acute lymphoblastic leukemia and 14 leukemic cell lines using several indicators of MMR defects. Microsatellite markers were compared between blast and normal DNA from the same patients and mutations were sought in mononucleotide repeat sequences of BAX and TGF-beta receptor II (TGF-beta RII). The absence of microsatellite instability (MI) and the absence of mutations in the genes examined from patient's leukemic cells contrasted with the observation that half of the cell lines displayed a high degree of MI and that three of seven of these mutator cell lines harbored mutations in BAX and/or TGF-beta RII. From these results we conclude that MMR defects are very uncommon in freshly isolated blasts but are likely to be selected for during the establishment of cell lines. Topics: bcl-2-Associated X Protein; Biomarkers, Tumor; DNA, Satellite; Frameshift Mutation; Humans; Precursor Cell Lymphoblastic Leukemia-Lymphoma; Proto-Oncogene Proteins; Proto-Oncogene Proteins c-bcl-2; Transforming Growth Factor beta; Tumor Cells, Cultured | 1998 |
TGF-beta and its receptor complex in leukemic B-cell precursors.
Transforming growth factor beta (TGF-beta) is a highly conserved peptide with growth-inhibitory activity in multiple normal and transformed cell types. Signal transduction is mediated through the receptor complex, consisting of two active seronine or threonine kinases (TGF-beta-receptor I and II) and the receptor-associated proteins betaglycan (TGF-beta-receptor III) and endoglin. In this study, we assessed the analysis of the role of TGF-beta and the transcription of the genes for TGF-beta and its receptor in highly purified leukemic B-cell precursors (BCPs) of patients with common acute lymphoblastic leukemia (cALL). Leukemic BCPs were positive for gene transcription of TGF-beta (9/9), the TGF-beta-receptor I (9/9), the TGF-beta-receptor II (6/6), betaglycan (5/6), and endoglin (6/6). Incubation with TGF-beta significantly reduced the cell viability of leukemic BCPs by a mean of 45% (p = 0.0009). The reduction of cell viability was associated with the induction of apoptosis by a mean of 31%. TGF-beta caused significant suppression of the S phase (p = 0.002) and accumulation in the G0/G1 phase (p = 0.0005). It also reduced expression of the adhesion surface receptor CD18 and the Fas antigen CD95 from 58% to 40% and from 48% to 27%, respectively. The data indicate that TGF-beta is a negative growth signal in leukemic BCPs and point to an additional role of TGF-beta as an immunomodulatory cytokine, suggesting a complex role of TGF-beta in the leukemogenesis of cALL. Topics: Adjuvants, Immunologic; Adolescent; Adult; Aged; Antibodies, Blocking; Antibodies, Monoclonal; Antigens, CD; Apoptosis; B-Lymphocytes; Cell Division; Cell Survival; Child; Child, Preschool; Humans; Infant; Middle Aged; Precursor B-Cell Lymphoblastic Leukemia-Lymphoma; Precursor Cell Lymphoblastic Leukemia-Lymphoma; Receptors, Transforming Growth Factor beta; RNA; Transcription, Genetic; Transforming Growth Factor beta; Tumor Cells, Cultured | 1998 |
[Study on gene expression of TGF beta 1 and its receptor in leukemia cells and the serum TGF beta 1 level in the patients with acute leukemia].
To investigate the mechanism by which TGF beta 1 inhibits the growth of leukemic cells and the significance of serum TGF beta 1 level in acute leukemia(AL) patients.. RT-PCR was used to detect gene expression of cytokines including IL-1, IL-3, IL-6, TGF beta 1 and their receptors (R) in leukemic and normal cells. Leukemic colony assay was carried out to determine the effect of TGF beta 1 on the growth of leukemic cells and the serum TGF beta 1 level in 33 AL patients were measured by ELISA.. TGF beta 1 was found to significantly inhibit the proliferation of leukemia cells of HL-60, K562 and DAMI cell lines. Furthermore, a variety of leukemia cell lines including HEL, HL-60, K562, U937, DAMI, MEG-01, HUT78 and CA were found to express mRNAs for TGF beta 1 and its receptor. In addition, TGF beta 1 was found to be able to evidently down-regulate the expression of mRNAs for IL-6, IL-6R, IL-3, GM-CSFR in DAMI cells and, interestingly, up-regulate TGF beta 1 gene expression in the cells per se before the cells showed DNA fragmentation, a molecular hallmark for cell apoptosis. It was shown that serum TGF beta 1 levels were significantly decreased in leukemic patients, restored to normal in the patients achieved complete remission, and tended to decrease again in the recurrent patients.. 1. TGF beta 1 may act as an inhibitory autocrine factor in the proliferation of leukemia cells; 2. the mechanism by which TGF beta 1 inhibits the growth of leukemia cells involves its down-regulating expression of positive cytokines and receptors and up-regulating TGF beta 1 expression in leukemia cells per se; 3. serum TGF beta 1 is a valuable parameter in monitoring the prognosis of leukemia and weakened control of TGF beta 1 on leukemia cells may play an important role in the pathogenesis of AL. Topics: Adult; Apoptosis; Female; Humans; Leukemia, Myeloid, Acute; Male; Precursor Cell Lymphoblastic Leukemia-Lymphoma; Receptors, Transforming Growth Factor beta; Reverse Transcriptase Polymerase Chain Reaction; Transforming Growth Factor beta; Transforming Growth Factor beta1 | 1998 |
TGF-beta inhibits growth and induces apoptosis in leukemic B cell precursors.
The uncontrolled proliferation of malignant lymphoblasts is the pathobiological hallmark in B cell precursor-ALL (BCP-ALL). Identification of inhibitory growth factors is of great importance for the understanding of growth control of leukemic B cell precursors and the development of novel therapeutic approaches in BCP-ALL. The aim of our study was the analysis of the effect of TGF-beta on cell survival and apoptosis of B cell precursors (BCP) from patients with acute lymphoblastic leukemia in vitro. Experiments were performed in a coculture system with cloned murine fibroblasts, which efficiently block spontaneous ex vivo apoptosis of BCP and thus allows the assessment of cytokine-induced growth inhibition. TGF-beta significantly reduced cell viability of highly purified, FACS isolated CD10+/CD19+ leukemic BCP by a mean of 53% (P = 0.0001). The loss of cell viability was accompanied by a significant increase of apoptosis with a mean of 70% (P = 0.0028). The TGF-beta effect was blocked specifically by a monoclonal anti-TGF-beta antibody. Induction of apoptotic cell death by TGF-beta was not accompanied by reduction of bcl-2 protein expression. TGF-beta transcription was not detected in the leukemic pre-B cell line BLIN-1, but in the murine fibroblasts. The growth inhibitory effect of TGF-beta was not restricted to leukemic BCP. The cytokine also increased apoptosis of normal, highly purified BCP by a mean of 58%. The data identify TGF-beta as a potent growth inhibitory cytokine for leukemic BCP. Topics: Animals; Antibodies, Monoclonal; Apoptosis; B-Lymphocytes; Cell Division; Cell Survival; Cells, Cultured; Hematopoietic Stem Cells; Humans; Interleukin-7; Mice; Precursor B-Cell Lymphoblastic Leukemia-Lymphoma; Precursor Cell Lymphoblastic Leukemia-Lymphoma; Proto-Oncogene Proteins c-bcl-2; Transforming Growth Factor beta | 1997 |
Molecular and biologic characterization of a newly established Philadelphia-positive acute lymphoblastic leukemia cell line (Z-33) with an autocrine response to GM-CSF.
We have recently established a new Philadelphia chromosome (Ph1)-positive acute lymphoblastic leukemia (ALL) cell line, designated Z-33. This line has L2 morphology, ultrastructural characteristics of lymphoblasts and typical B lineage surface markers identical to those observed in the Ph1-positive ALL patient from whom the line was derived. In addition, a rearranged immunoglobulin heavy-chain gene (JH) band was found in Z-33 cells by Southern blot analysis, confirming B cell clonality. Cytogenetic analysis of the cell line revealed t(9;22)(q34;q11.2). Polymerase chain reaction (PCR)-amplified cDNA from Z-33 cells demonstrated an e1-az BCR-ABL junction, and the p190BCR-ABL protein was detected in them by the immune complex kinase assay. Z-33 cells produce interleukin (IL)-1 beta, IL-6, granulocyte colony-stimulating factor (G-CSF), granulocyte-macrophage CSF (GM-CSF), tumor necrosis factor (TNF)-alpha, and transforming growth factor (TGF)-beta, Neither IL-1 beta, G-CSF, TNF-alpha, nor their corresponding antibodies affected the cell line's growth. In contrast, anti-GM-CSF neutralizing antibodies suppressed Z-33 colony formation, and GM-CSF stimulated it in a dose-dependent fashion. In addition, receptor studies with biotinylated GM-CSF demonstrated specific binding to Z-33 cells, indicating that the cells express GM-CSF receptors. Taken together, our data suggest that the Ph1-positive Z-33 ALL cells produce GM-CSF, express GM-CSF receptors, and show an autocrine proliferative response to this cytokine. Topics: Antibodies; Blotting, Northern; Blotting, Southern; Burkitt Lymphoma; Cell Division; Cytokines; DNA, Neoplasm; Female; Fusion Proteins, bcr-abl; Granulocyte Colony-Stimulating Factor; Granulocyte-Macrophage Colony-Stimulating Factor; Humans; Karyotyping; Microscopy, Electron; Middle Aged; Precursor Cell Lymphoblastic Leukemia-Lymphoma; RNA, Messenger; Stimulation, Chemical; Transforming Growth Factor beta; Tumor Cells, Cultured | 1996 |