transforming-growth-factor-beta and Astrocytoma

transforming-growth-factor-beta has been researched along with Astrocytoma* in 38 studies

Reviews

2 review(s) available for transforming-growth-factor-beta and Astrocytoma

ArticleYear
Cytokine gene expression within the central nervous system.
    Cellular and molecular neurobiology, 1998, Volume: 18, Issue:6

    1. The identification of cytokine genes expressed in the central nervous system is critical to understanding the immune network in various diseases of brain, such as infection, degeneration, and malignancy. 2. Expression of cytokine genes in human astrocytoma cell lines and in fresh brain specimens was studied by the reverse-transcribed/polymerase chain reaction method. 3. The correlation between clinical malignancy and cytokine gene expression within malignant glioma was examined, especially regarding the relevancy of inhibitory cytokines, such as transforming growth factor-beta and interleukin-10.

    Topics: Animals; Astrocytoma; Brain; Brain Diseases; Brain Neoplasms; Cytokines; Humans; Interleukin-10; Neurodegenerative Diseases; Transforming Growth Factor beta; Tumor Cells, Cultured

1998
Modulation of the immune response by transforming growth factor beta.
    International archives of allergy and immunology, 1992, Volume: 99, Issue:1

    For the past several years immunologists have been fascinated by a series of experiments showing that transforming growth factor beta (TGF beta) suppresses T- and B-lymphocyte growth as well as IgM and IgG production by B cells. Moreover, while exerting chemotactic activity on monocytes and inducing expression of interleukin-1 and interleukin-6 by these cells, TGF beta interferes with bacterially induced tumor necrosis factor alpha production, oxygen radical formation and the adhesiveness of granulocytes to endothelial cells. These mechanisms may provide the basis for the effect of TGF beta to prevent the microvascular changes associated with brain edema formation in bacterial meningitis. Given the potential of lymphocytes as well as macrophages to produce TGF beta 1, this cytokine may exert negative feedback signals on the immune response, provided the cytokine is processed from its latent form to the bioactive homodimer. Potent effects of TGF beta have been observed in experimental animals including the inhibition of the generation of virus-specific cytotoxic T cells and antiviral antibodies as well as the diminution of cellular infiltrates with decreased major histocompatibility complex class-II expression and CD8+ T cells in the tissue of virally infected animals. TGF beta may also be of importance in tumor immunology. By the production of bioactive TGF beta as detected in glioblastoma and acute T-cell leukemia, tumor cells may induce an immunodeficiency state and escape immune surveillance. In inflammation, monitoring of TGF beta in the tissue will bring light on the immune regulation in acute and chronic inflammatory diseases.

    Topics: Animals; Astrocytoma; Autoimmunity; B-Lymphocytes; Brain Neoplasms; Cytokines; Humans; Immunoglobulins; Immunosuppression Therapy; Meningitis, Pneumococcal; T-Lymphocytes; Transforming Growth Factor beta

1992

Other Studies

36 other study(ies) available for transforming-growth-factor-beta and Astrocytoma

ArticleYear
Pre-radiation lymphocyte harvesting and post-radiation reinfusion in patients with newly diagnosed high grade gliomas.
    Journal of neuro-oncology, 2015, Volume: 124, Issue:2

    Radiation (RT), temozolomide (TMZ), and dexamethasone in newly diagnosed high grade gliomas (HGG) produces severe treatment-related lymphopenia (TRL) that is associated with early cancer-related deaths. This TRL may result from inadvertent radiation to circulating lymphocytes. This study reinfused lymphocytes, harvested before chemo-radiation, and assessed safety, feasibility, and trends in lymphocyte counts. Patients with newly diagnosed HGG and total lymphocyte counts (TLC) ≥ 1000 cells/mm(3) underwent apheresis. Cryopreserved autologous lymphocytes were reinfused once radiation was completed. Safety, feasibility, and trends in TLC, T cell subsets and cytokines were studied. Serial TLC were also compared with an unreinfused matched control group. Ten patients were harvested (median values: age 56 years, dexamethasone 3 mg/day, TLC/CD4 1980/772 cells/mm(3)). After 6 weeks of RT/TMZ, TLC fell 69 % (p < 0.0001) with similar reductions in CD4, CD8 and NK cells but not Tregs. Eight patients received lymphocyte reinfusions (median = 7.0 × 10(7) lymphocytes/kg) without adverse events. A post-reinfusion TLC rise of ≥300 cells/mm(3) was noted in 3/8 patients at 4 weeks and 7/8 at 14 weeks which was similar to 23 matched controls. The reduced CD4/CD8 ratio was not restored by lymphocyte reinfusion. Severe lymphopenia was not accompanied by elevated serum interleukin-7 (IL-7) levels. This study confirms that severe TRL is common in HGG and is not associated with high plasma IL-7 levels. Although lymphocyte harvesting/reinfusion is feasible and safe, serial lymphocyte counts are similar to unreinfused matched controls. Studies administering higher lymphocyte doses and/or IL-7 should be considered to restore severe treatment-related lymphopenia in HGG.

    Topics: Adult; Astrocytoma; Blood Transfusion, Autologous; Chemoradiotherapy; Feasibility Studies; Female; Glioblastoma; Humans; Interleukin-7; Lymphocyte Count; Lymphocyte Transfusion; Male; Middle Aged; Prospective Studies; Transforming Growth Factor beta; Treatment Outcome

2015
Human U87 astrocytoma cell invasion induced by interaction of βig-h3 with integrin α5β1 involves calpain-2.
    PloS one, 2012, Volume: 7, Issue:5

    It is known that βig-h3 is involved in the invasive process of many types of tumors, but its mechanism in glioma cells has not been fully clarified. Using immunofluorescent double-staining and confocal imaging analysis, and co-immunoprecipitation assays, we found that βig-h3 co-localized with integrin α5β1 in U87 cells. We sought to elucidate the function of this interaction by performing cell invasion assays and gelatin zymography experiments. We found that siRNA knockdowns of βig-h3 and calpain-2 impaired cell invasion and MMP secretion. Moreover, βig-h3, integrins and calpain-2 are known to be regulated by Ca(2+), and they are also involved in tumor cell invasion. Therefore, we further investigated if calpain-2 was relevant to βig-h3-integrin α5β1 interaction to affect U87 cell invasion. Our data showed that βig-h3 co-localized with integrin α5β1 to enhance the invasion of U87 cells, and that calpain-2, is involved in this process, acting as a downstream molecule.

    Topics: Astrocytes; Astrocytoma; Calpain; Cell Adhesion; Cell Line, Tumor; Cell Movement; Extracellular Matrix Proteins; Humans; Integrin alpha5beta1; RNA, Small Interfering; Transforming Growth Factor beta

2012
Modulation of TGF-beta activity by latent TGF-beta-binding protein 1 in human malignant glioma cells.
    International journal of cancer, 2009, Aug-01, Volume: 125, Issue:3

    High biological activity of the transforming growth factor (TGF)-beta-Smad pathway characterizes the malignant phenotype of malignant gliomas and confers poor prognosis to glioma patients. Accordingly, TGF-beta has become a novel target for the experimental treatment of these tumors. TGF-beta is processed by furin-like proteases (FLP) and secreted from cells in a latent complex with its processed propeptide, the latency-associated peptide (LAP). Latent TGF-beta-binding protein 1 (LTBP-1) covalently binds to this small latent TGF-beta complex (SLC) and regulates its function, presumably via interaction with the extracellular matrix (ECM). We report here that the levels of LTBP-1 protein in vivo increase with the grade of malignancy in gliomas. LTBP-1 is associated with the ECM as well as secreted into the medium in cultured malignant glioma cells. The release of LTBP-1 into the medium is decreased by the inhibition of FLP activity. Gene-transfer mediated overexpression of LTBP-1 in glioma cell lines results in an increase inTGF-beta activity. Accordingly, Smad2 phosphorylation as an intracellular marker of TGF-beta activity is enhanced. Conversely, LTBP-1 gene silencing reduces TGF-beta activity and Smad2 phosphorylation without affecting TGF-beta protein levels. Collectively, we identify LTBP-1 as an important modulator of TGF-beta activation in glioma cells, which may contribute to the malignant phenotype of these tumors.

    Topics: Astrocytoma; Brain Neoplasms; Cell Line, Tumor; Cell Proliferation; Extracellular Matrix; Gene Expression Regulation, Neoplastic; Glioblastoma; Glioma; Humans; Immunoblotting; Immunohistochemistry; Latent TGF-beta Binding Proteins; Oligonucleotide Array Sequence Analysis; Phenotype; Phosphorylation; Polymerase Chain Reaction; Signal Transduction; Smad2 Protein; Transforming Growth Factor beta; Up-Regulation

2009
High TGFbeta-Smad activity confers poor prognosis in glioma patients and promotes cell proliferation depending on the methylation of the PDGF-B gene.
    Cancer cell, 2007, Volume: 11, Issue:2

    TGFbeta acts as a tumor suppressor in normal epithelial cells and early-stage tumors and becomes an oncogenic factor in advanced tumors. The molecular mechanisms involved in the malignant function of TGFbeta are not fully elucidated. We demonstrate that high TGFbeta-Smad activity is present in aggressive, highly proliferative gliomas and confers poor prognosis in patients with glioma. We discern the mechanisms and molecular determinants of the TGFbeta oncogenic response with a transcriptomic approach and by analyzing primary cultured patient-derived gliomas and human glioma biopsies. The TGFbeta-Smad pathway promotes proliferation through the induction of PDGF-B in gliomas with an unmethylated PDGF-B gene. The epigenetic regulation of the PDGF-B gene dictates whether TGFbeta acts as an oncogenic factor inducing PDGF-B and proliferation in human glioma.

    Topics: Adolescent; Adult; Aged; Astrocytoma; Brain Neoplasms; Cell Proliferation; Child; Child, Preschool; DNA Methylation; Gene Expression Profiling; Gene Expression Regulation, Neoplastic; Glioblastoma; Glioma; Humans; Infant; Middle Aged; Oligonucleotide Array Sequence Analysis; Phosphorylation; Prognosis; Proto-Oncogene Proteins c-sis; Receptors, Transforming Growth Factor beta; Signal Transduction; Smad2 Protein; Smad7 Protein; Survival Rate; Transforming Growth Factor beta; Tumor Cells, Cultured

2007
Hepatocyte growth factor production is stimulated by gangliosides and TGF-beta isoforms in human glioma cells.
    Journal of neuro-oncology, 2007, Volume: 85, Issue:1

    Hepatocyte growth factor (HGF) is a pleiotrophic cytokine that stimulates motility and invasion of several cancer cell types and induces angiogenesis, which is known to be expressed in several malignancies including glioma. The effect of transforming growth factor-beta (TGF-beta) isoforrns as well as gangliosides on HGF production was investigated in human glioma cell lines. TGF-beta isoforms and gangliosides were found to differentially stimulate HGF production by these cells. The ganglioside GD3 enhanced this release to the greatest extent and the stimulation was more marked in a glioblastoma cell line than in the two other anaplastic astrocytoma cell lines. These results suggest that both TGF-betas and gangliosides may act as indirect angiogenic factors by stimulating HGF secretion.

    Topics: Astrocytoma; Brain Neoplasms; Cell Line, Tumor; Culture Media; Culture Media, Serum-Free; Enzyme-Linked Immunosorbent Assay; Gangliosides; Glioma; Hepatocyte Growth Factor; Humans; Isomerism; Neovascularization, Pathologic; Stimulation, Chemical; Transforming Growth Factor beta

2007
Tenascin-C protein is induced by transforming growth factor-beta1 but does not correlate with time to tumor progression in high-grade gliomas.
    Journal of neuro-oncology, 2006, Volume: 77, Issue:1

    Tenascin-C is an extracellular matrix protein known to correlate with prognosis in patients with glioblastoma, probably by stimulation of invasion and neoangiogenesis. Transforming Growth Factor-beta1 (TGF-beta1) plays an important role in the biology of high-grade gliomas, partly by regulating invasion of these tumors into parenchyma. This study was designed to evaluate if TGF-beta1 induces the expression and deposition of Tenascin-C in the extracellular matrix of high-grade gliomas which may be pivotal for the invasion of these tumors into healthy parenchyma.. A series of 20 high-grade gliomas was stained immunohistochemically with Tenascin-C- and TGF-beta1- specific antibodies. Expression levels of both proteins were evaluated and correlated with each other, time to progression and molecular and morphological markers of invasion. A quantitative PCR assay was performed evaluating the induction of Tenascin-C mRNA by treatment with TGF-beta1 in vitro.. Tenascin-C was expressed in 18 of 19 (95%) evaluable tumors, whereas 14 of 20 tumors (70%) expressed TGF-beta1 in a significant percentage of cells. Treatment with TGF-beta1 did induce the expression of Tenascin-C at the mRNA and protein level in vitro. The expression of Tenascin-C and TGF-beta1 did neighter statistically correlate with each other nor with time to progression.. In our series, Tenascin-C and TGF-beta1 were expressed in the vast majority of high-grade gliomas. We could not detect a correlation of one of the proteins with time to progression. Nevertheless, we describe induction of Tenascin-C by TGF-beta1, possibly providing a mechanism for the invasion of high-grade gliomas into healthy parenchyma.

    Topics: Adult; Aged; Astrocytoma; Biomarkers, Tumor; Brain Neoplasms; Child; Disease Progression; Extracellular Matrix Proteins; Gene Expression Regulation, Neoplastic; Glioblastoma; Glioma; Gliosarcoma; Humans; Immunohistochemistry; Middle Aged; Neoplasm Invasiveness; RNA, Messenger; Tenascin; Time Factors; Transforming Growth Factor beta; Transforming Growth Factor beta1; Tumor Cells, Cultured

2006
Transforming growth factor beta receptor family ligands inhibit hepatocyte growth factor synthesis and secretion from astrocytoma cells.
    Brain research. Molecular brain research, 2004, Feb-05, Volume: 121, Issue:1-2

    Transforming growth factor beta (TGFbeta) and hepatocyte growth factor (HGF) promote glioma progression. Using U87human astrocytoma cells, which express TGFbeta receptors (TbetaRs), we show (1) mRNA expression of Smads (2, 3, 4), bone morphogenetic protein (BMP)- and activin-A receptors; (2) TGFbeta1 inhibits and HGF induces proliferation; (3) TGFbeta1 and activin-A equipotently inhibit HGF secretion more than BMP-2, but none alters c-Met expression. Because interfering with TbetaR signaling might nullify the beneficial inhibition of HGF secretion, activin-A should instead be considered for combination glioma therapy.

    Topics: Activin Receptors; Activins; Analysis of Variance; Astrocytoma; Blotting, Northern; Bone Morphogenetic Protein 2; Bone Morphogenetic Proteins; Bromodeoxyuridine; Cell Division; Cell Line, Tumor; Culture Media, Conditioned; DNA-Binding Proteins; Dose-Response Relationship, Drug; Enzyme-Linked Immunosorbent Assay; Gene Expression Regulation; Hepatocyte Growth Factor; Humans; Inhibin-beta Subunits; Ligands; Receptors, Transforming Growth Factor beta; Reverse Transcriptase Polymerase Chain Reaction; RNA, Messenger; Smad Proteins; Starvation; Trans-Activators; Transforming Growth Factor beta; Transforming Growth Factor beta1

2004
Transforming growth factor-beta-inducible gene-h3 (beta(ig)-h3) promotes cell adhesion of human astrocytoma cells in vitro: implication of alpha6beta4 integrin.
    Neuroscience letters, 2003, Jan-16, Volume: 336, Issue:2

    Beta(ig)-h3 is a secretory protein that is induced by transforming growth factor (TGF)-beta. We have recently found that beta(ig)-h3 expression is induced in cultured astrocytes by TGF-beta1 and in rat cerebral cortex by stab wound. The purpose of this study was to examine the effect of the secreted beta(ig)-h3 on cell adhesion of astrocytes and the underlying mechanisms. When U87 human astrocytoma cells were seeded on dishes coated with recombinant beta(ig)-h3, cell adhesion was significantly enhanced. Blocking experiments using various antibodies to the integrin subunit suggested alpha6beta4 integrin could be involved in the beta(ig)-h3-mediated astrocyte cell adhesion. Cell adhesion to beta(ig)-h3 substrate was substantially blocked by preincubation with the inhibitor to the src kinase. When cells were plated on beta(ig)-h3-coated dishes, tyrosine phosphorylation of focal adhesion kinase was prominently increased within 20 min in a beta4 integrin-dependent manner. The results suggest that alpha6beta4 integrin-mediated interactions of astrocytes with beta(ig)-h3 transduce intracellular signals through the focal adhesion proteins, which may regulate certain aspects of astrocyte response to brain injury.

    Topics: Astrocytoma; Cell Adhesion; Cytoskeletal Proteins; Dose-Response Relationship, Drug; Extracellular Matrix Proteins; Humans; Integrin alpha6beta1; Neoplasm Proteins; Reproducibility of Results; Sensitivity and Specificity; src-Family Kinases; Transforming Growth Factor beta; Tumor Cells, Cultured

2003
Interaction of transforming growth factor-beta (TGF-beta) and epidermal growth factor (EGF) in human glioma cells.
    Journal of neuro-oncology, 2003, Volume: 63, Issue:2

    Gliomas are characterized by a deregulation of growth factor production and growth factor receptors expression, e.g. overproduction of the cytokine transforming growth factor-beta (TGF-beta) and overexpression/constitutive activation of receptors for the epidermal growth factor (EGF). Potential interactions of such growth factors and their signaling cascades could enhance the malignancy of these tumors. Therefore, we investigated the effects of TGF-beta and EGF alone and in combination on the proliferation of glioma cells cultivated from eight solid human WHO grade IV gliomas and one glioma cell line, analyzed the expression and intactness of the TGF-beta-signaling molecules Samd-4 and -2, and the phosphorylation of the EGF-signaling kinases ERK 1/2. The effects were divergent and complex: Whereas EGF mostly stimulated glioma cell proliferation, TGF-beta either enhanced, inhibited or had no significant effect on proliferation. In combination, co-stimulation and inhibition of the EGF-induced mitogenic activity could be observed. Smad-4/-2 were expressed in all glioma cells, one point mutation at base 1595 in Smad-4 did not affect its protein sequence. In part of the glioma cells, reduced phosphorylation of ERK 1/2 and expression of cyclin-dependent kinase inhibitor 1 or p21 was observed in co-stimulation experiments. These experiments show that TGF-beta can inhibit EGF-mediated effects only in some gliomas, whereas it enhances it in others. The interaction of both factors is very complex and varies between different gliomas.

    Topics: Astrocytoma; Brain Neoplasms; Cell Cycle Proteins; Cell Division; Cyclin-Dependent Kinase Inhibitor p21; Cyclin-Dependent Kinase Inhibitor p27; Cyclins; DNA Primers; Epidermal Growth Factor; ErbB Receptors; Humans; Immunoenzyme Techniques; Ki-67 Antigen; MAP Kinase Signaling System; Mitogen-Activated Protein Kinase 1; Mitogen-Activated Protein Kinase 3; Mitogen-Activated Protein Kinases; Phosphorylation; Polymerase Chain Reaction; Signal Transduction; Transforming Growth Factor beta; Tumor Cells, Cultured; Tumor Suppressor Proteins

2003
cDNA array analysis of SPARC-modulated changes in glioma gene expression.
    Journal of neuro-oncology, 2002, Volume: 60, Issue:3

    We have demonstrated that secreted protein acidic and rich in cysteine (SPARC) is highly expressed in human gliomas and it promotes glioma invasion and delays tumor growth in vitro and in vivo. cDNA array analyses were performed to determine whether SPARC, which interacts at the cell surface, has an impact on intracellular signaling and downstream gene expression changes, which might account for some of its effects on invasion and growth. Using a doxycycline (dox)-controlled gene expression system, two cDNA array analyses were performed using a parental U87T2 clone (-SPARC) transfected with the dox-controlled transactivator and a U87T2 parental-derived SPARC-transfected clone, A2b2 (+SPARC). Array analysis performed between the parental and the SPARC-transfected clone (-dox) identified 13 upregulated genes and 14 downregulated genes. With the exception of PAI-1 and MMP2, the identified genes are novel with respect to SPARC's mechanism of action. Array analysis performed using the SPARC-transfected clone ( +/- dox) identified 2 types of gene regulation; one reversible upon SPARC suppression, the other irreversible. Two of the SPARC-induced genes, BIGH3 (irreversible by dox) and PAI-1 (reversible by dox) were further studied in additional SPARC-transfected clones, human astrocytoma tissues, and human glioma cell lines by RT-PCR and Northern blot analyses. The results indicate that: (1) the array results were validated, (2) the dox regulation was validated, and (3) the differential expression identified by the array analyses was present between normal brain and in human astrocytoma tissues and cell lines. Therefore, we conclude that these cDNA array analyses provide candidate genes involved in SPARC-mediated effects on glioma cell cycle progression, signaling, and migration, and that SPARC may induce reversible and irreversible gene expression changes. Further investigation of these candidates may shed insights into SPARC's role in glioma cell proliferation and invasion, and potential use as a therapeutic target.

    Topics: Anti-Bacterial Agents; Astrocytes; Astrocytoma; Blotting, Northern; Brain; Cell Transformation, Neoplastic; Doxycycline; Extracellular Matrix Proteins; Fetus; Gene Expression Profiling; Gene Expression Regulation, Neoplastic; Glioma; Humans; Neoplasm Invasiveness; Neoplasm Proteins; Oligonucleotide Array Sequence Analysis; Osteonectin; Reverse Transcriptase Polymerase Chain Reaction; RNA-Binding Proteins; Transfection; Transforming Growth Factor beta; Tumor Cells, Cultured

2002
Effects of ethanol and basic fibroblast growth factor on the transforming growth factor beta1 regulated proliferation of cortical astrocytes and C6 astrocytoma cells.
    Alcoholism, clinical and experimental research, 2002, Volume: 26, Issue:5

    The effects of ethanol on transforming growth factor beta1 (TGFbeta1)-regulated proliferation of primary cultured cortical astrocytes and C6 astrocytoma cells were determined.. Cultured cells were treated with TGFbeta1 (0 or 10 ng/ml) and ethanol (0 or 400 mg/dl) for as many as 4 days. The effects of these treatments on cell growth (the change in cell number) and cell proliferation (incorporation of [3H]thymidine) were determined. In addition, the effects of serum and basic fibroblast growth factor (bFGF) on TGFbeta1-affected cell proliferation were examined.. Both TGFbeta1 and ethanol inhibited cell growth and proliferation among the two types of cells; this effect was only evident when the cells were grown in the presence of a mitogen, serum, or bFGF. When added together (either in a serum-supplemented or serum-free medium), however, TGFbeta1 and the mitogen effectively nullified each other. TGFbeta1 blocked the mitogenic effect of bFGF on astrocytes and C6 cells. Ethanol did not affect (i.e., neither added nor nullified) this antagonistic effect.. TGFbeta1, like ethanol, is a potent anti-proliferative agent; either can nullify the mitogenic activity of serum or bFGF. Astrocytes and astrocyte-like cells react to combined TGFbeta1 and ethanol treatment differently than do neurons for which the inhibitory effects of TGFbeta1 and ethanol are additive.

    Topics: Animals; Astrocytes; Astrocytoma; Cell Division; Cells, Cultured; Cerebral Cortex; Culture Media, Serum-Free; Ethanol; Fibroblast Growth Factor 2; Rats; Rats, Sprague-Dawley; Transforming Growth Factor beta; Transforming Growth Factor beta1; Tumor Cells, Cultured

2002
Inhibition of furin-mediated processing results in suppression of astrocytoma cell growth and invasiveness.
    Clinical cancer research : an official journal of the American Association for Cancer Research, 2002, Volume: 8, Issue:6

    Astrocytoma arises in the central nervous system as a tumor of great lethality, in part because of the invasive potential of the neoplastic cells that are able to release extracellular matrix-degrading enzymes. Furin convertase activates several precursor matrix metalloproteases involved in the breakdown of the extracellular matrix. In the present study inhibition of furin was achieved by gene transfer of alpha(1)-antitrypsin Portland (PDX) cDNA.. This furin inhibitor was transfected into two tumorigenic astrocytoma cell lines. The inhibitory effect was evaluated using in vivo tumorigenicity, invasion, and proliferation assays, as well as by investigating impairment of furin substrate processing.. Expression of PDX prevented the s.c. growth of the transfected cells. Invasion assays demonstrated that PDX-transfected cells exhibited a reduced invasive ability in vitro and in vivo. Furthermore, s.c. growth of PDX transfectant xenotransplants showed a significant reduction in size that coincided with a significant decrease of the in vitro doubling time and of the in vivo cell proliferation ability. Additional studies showed that the furin substrates insulin-like growth factor IR, transforming growth factor beta and membrane type 1-matrix metalloprotease were not activated in PDX-expressing astrocytoma cells.. PDX expression in astrocytoma cells demonstrated a direct mechanistic link between furin inhibition, and decreased astrocytoma proliferation and invasive ability. Because furin inhibition inhibits both invasiveness and cell growth in astrocytoma, furin should be considered a promising target for glioblastoma therapy.

    Topics: alpha 1-Antitrypsin; Animals; Astrocytoma; Blotting, Western; Brain Neoplasms; Cell Division; Extracellular Matrix; Furin; Humans; Immunoenzyme Techniques; In Vitro Techniques; Injections, Subcutaneous; Metalloendopeptidases; Mice; Mice, SCID; Neoplasm Invasiveness; Neoplasm Transplantation; Rats; Receptor, IGF Type 1; Subtilisins; Trachea; Transfection; Transforming Growth Factor beta; Tumor Cells, Cultured

2002
TGF-beta1 up-regulates paxillin protein expression in malignant astrocytoma cells: requirement for a fibronectin substrate.
    Oncogene, 2001, Nov-29, Volume: 20, Issue:55

    Cytokines can influence the interactions between members of the integrin cell adhesion receptor family and the extracellular matrix thereby potentially affecting cell function and promoting cell adhesion, growth and migration of malignant astrocytoma tumor cells. As malignant astrocytoma cells synthesize TGF-beta1 in vivo, we analysed the effects of TGF-beta1 on signaling events associated with integrin receptor ligation, focusing on the effects on paxillin, a phosphorylated adaptor protein, that acts as a scaffold for signaling molecules recruited to focal adhesions. TGF-beta1-stimulation of primary astrocytes and serum-starved U-251MG malignant astrocytoma cells attached to fibronectin induced a substantial increase in the levels of paxillin protein (fivefold increase at 2.0 ng/ml) in a dose- and time-dependent manner compared to the levels observed on plating onto fibronectin in the absence of stimulation. In the astrocytoma cells, this resulted in an increase in the pool of tyrosine-phosphorylated paxillin, although it did not appear to alter the extent of phosphorylation of the paxillin molecules. In contrast, in primary astrocytes the protein levels were upregulated in the absence of a parallel increase in phosphorylation. The TGF-beta1-stimulated increase in paxillin levels required ligation of the fibronectin receptor, as it was not induced when the cells were plated onto vitronectin, collagen or laminin. The increase in the pool of paxillin on TGF-beta1 stimulation of the fibronectin-plated astrocytoma cells was associated with an increase in translation, but was not associated with an increase in the steady-state levels of paxillin mRNA. Stimulation with TGF-beta1 on a fibronectin substrate increased subsequent attachment and spreading of U-251MG cells onto fibronectin and, to a lesser extent, vitronectin, but not collagen. Our results indicate that physiologic levels of TGF-beta1 stimulate the expression of paxillin protein at the level of translation through a process that requires engagement of the fibronectin receptor, and promotes attachment and spreading of malignant astrocytoma cells on fibronectin.

    Topics: Animals; Astrocytoma; Blotting, Western; Cell Adhesion; Cell Size; Central Nervous System Neoplasms; Cytoskeletal Proteins; Dose-Response Relationship, Drug; Extracellular Matrix; Fibronectins; Half-Life; Humans; Microscopy, Fluorescence; Paxillin; Phosphoproteins; Phosphorylation; Protein Biosynthesis; Pseudopodia; Rats; Receptors, Fibronectin; RNA, Messenger; Substrate Specificity; Time Factors; Transforming Growth Factor beta; Transforming Growth Factor beta1; Tumor Cells, Cultured; Up-Regulation

2001
Expression of TGF-beta isoforms, TGF-beta receptors, and SMAD molecules at different stages of human glioma.
    International journal of cancer, 2000, May-20, Volume: 89, Issue:3

    Human gliomas express TGF-beta but, so far the expression of downstream mediators has been investigated in only a few cell lines. We have examined tissue specimens of 23 gliomas: 3 astrocytomas grade II (AST), 8 anaplastic astrocytomas grade III (AAST), and 12 glioblastoma multiforme grade IV (GBM). We analyzed the mRNA expression of TGF-beta1, TGF-beta2, TGF-beta3, the TGF-beta receptors type I (TbetaR-I) and type II (TbetaR-II), Smad2, Smad3, and Smad4. mRNA expression of IL-10 and CD95 (FAS/APO-1) were also studied. We detected increased mRNA levels of the 3 TGF-beta isoforms, correlating with the degree of malignancy. TGF-beta3 mRNA was increased, particularly in AST and AAST, while TGF-beta1 and TGF-beta2 mRNAs were strongly expressed in GBM. TGF-beta normally up-regulates the TGF-beta receptors, and TbetaR-I and TbetaR-II showed stronger expression in all gliomas when compared to normal tissues. However, the mRNA expression of Smad2, Smad3, and Smad4 was decreased in GBM. IL-10 mRNA expression was detected in glioma tissues but not in glioma cell lines. No marked increase in the expression of soluble CD95 splicing variants was found in the gliomas compared with normal tissue. However, total CD95 mRNA was elevated among GBM tissues.

    Topics: Activin Receptors, Type I; Adolescent; Adult; Astrocytoma; Brain; Brain Neoplasms; DNA-Binding Proteins; DNA, Complementary; Enzyme-Linked Immunosorbent Assay; fas Receptor; Female; Glioblastoma; Glioma; Humans; Interleukin-10; Male; Middle Aged; Protein Isoforms; Protein Serine-Threonine Kinases; Receptor, Transforming Growth Factor-beta Type I; Receptor, Transforming Growth Factor-beta Type II; Receptors, Transforming Growth Factor beta; Reverse Transcriptase Polymerase Chain Reaction; RNA, Messenger; Signal Transduction; Smad2 Protein; Smad3 Protein; Smad4 Protein; Trans-Activators; Transforming Growth Factor beta; Tumor Cells, Cultured

2000
Oncostatin M regulation of interleukin-6 expression in astrocytes: biphasic regulation involving the mitogen-activated protein kinases ERK1/2 and p38.
    Journal of neurochemistry, 2000, Volume: 75, Issue:2

    Oncostatin M (OSM) is a member of the interleukin (IL)-6 family of cytokines and has both pro- and anti-inflammatory properties. Of interest, OSM has functional effects within the CNS. We have shown recently that OSM can modulate expression of the cytokine IL-6 in astrocytes. Herein we characterize the molecular mechanisms and signaling cascades involved in this response. OSM induces IL-6 protein expression in a dose- and time-dependent manner in astrocytes. In addition, OSM can synergize with the cytokines tumor necrosis factor-alpha, IL-1beta, and transforming growth factor-beta for enhanced IL-6 expression. Using neutralizing antibodies to gp 130, the OSM receptor (OSMR), and the leukemia inhibitory factor receptor (LIFR), we document that OSM exclusively uses the OSMR/gp 130 heterodimer in signaling events, rather than the LIFR/gp 130 heterodimer. Kinetic analysis of OSM-induced IL-6 mRNA reveals two up-regulatory events. The first, peaking at 1 h, is transient, does not require protein synthesis, and is regulated at the transcriptional level. The second, peaking between 6 and 8 h, is prolonged and sensitive to puromycin, suggesting a requirement for de novo protein synthesis, and also is transcriptionally regulated. OSM-induced IL-6 mRNA and protein expression is inhibited by the mitogen-activated protein kinase (MAPK) inhibitors U0126 and SB202190, suggesting a requirement for the MAPKs ERK1/2 and p38 in this response. Finally, we show that the MAPKs ERK1/2 and p38 are activated by OSM in astrocytes and that this activation is reduced by the MAPK inhibitors. These data demonstrate that OSM induces IL-6 expression in astrocytes and that the MAPKs ERK1/2 and p38 participate in this response.

    Topics: Adult; Astrocytes; Astrocytoma; Cytokines; Gene Expression Regulation; Growth Inhibitors; Humans; Interleukin-1; Interleukin-6; Kinetics; Leukemia Inhibitory Factor; Lymphokines; Mitogen-Activated Protein Kinase 1; Mitogen-Activated Protein Kinase 3; Mitogen-Activated Protein Kinases; Oncostatin M; p38 Mitogen-Activated Protein Kinases; Peptides; Receptors, Cytokine; Receptors, Oncostatin M; Recombinant Proteins; Transcription, Genetic; Transforming Growth Factor beta; Tumor Cells, Cultured; Tumor Necrosis Factor-alpha

2000
Costimulatory effects of interferon-gamma and interleukin-1beta or tumor necrosis factor alpha on the synthesis of Abeta1-40 and Abeta1-42 by human astrocytes.
    Neurobiology of disease, 2000, Volume: 7, Issue:6 Pt B

    Chronic inflammation and astrocytosis are characteristic histopathological features of Alzheimer's Disease (AD). Astrocytes are one of the predominant cell types in the brain. In AD they are activated and produce inflammatory components such as complement components, acute phase proteins, and cytokines. In this study we analyzed the effect of cytokines on the production of amyloid beta (Abeta) in the astrocytoma cell line U373 and in primary human astrocytes isolated postmortem from healthy aged persons as well as from patients with AD. Astrocytes did not produce Abeta in the absence of stimuli or following stimulation with IL-1beta, TNFalpha, IL-6, and TGF-beta1. Neither did combinations of TNFalpha and IL-1beta, IL-6 or TGF-beta1, or the coadministration of IFNgamma and IL-6 or TGF-beta1 induce Abeta production. In contrast, pronounced production of Abeta1-40 and Abeta1-42 was observed when primary astrocytes or astrocytoma cells were stimulated with combinations of IFNgamma and TNFalpha or IFNgamma and IL-1beta. Induction of Abeta production was accompanied by decreased glycosylation of APP as well as by increased secretion of APPsbeta. Our results suggest that astrocytes may be an important source of Abeta in the presence of certain combinations of inflammatory cytokines. IFNgamma in combination with TNFalpha or IL-1beta seems to trigger Abeta production by supporting beta-secretase cleavage of the immature APP molecule.

    Topics: Aged; Aged, 80 and over; Alzheimer Disease; Amyloid beta-Peptides; Amyloid beta-Protein Precursor; Astrocytes; Astrocytoma; Cells, Cultured; Cytokines; Drug Synergism; Female; Glycosylation; Humans; Interferon-gamma; Interleukin-1; Interleukin-6; Male; Middle Aged; Peptide Fragments; Protein Processing, Post-Translational; Transforming Growth Factor beta; Transforming Growth Factor beta1; Tumor Necrosis Factor-alpha

2000
Correlation of thrombospondin-1 and transforming growth factor-beta expression with malignancy of glioma.
    Neuropathology : official journal of the Japanese Society of Neuropathology, 2000, Volume: 20, Issue:3

    The expression of thrombospondin-1 (TSP-1) and its role in gliomas have not been well examined. In the present study TSP-1 expression in a panel of malignant glioma cell lines and the expression of TSP-1 and transforming growth factor (TGF-beta) proteins in low-grade and malignant glioma tissues were investigated. Reverse transcription-polymerase chain reaction analysis showed that nine of nine malignant glioma cell lines expressed TSP-1 mRNA, and seven of nine glioma lines expressed TSP-2 mRNA. Production and secretion of TSP-1 were examined in the T98G glioblastoma cell line by western blot analysis. Total TSP-1 protein content in the supernatant was 10 times higher than that in the cell lysate. Secretion of TSP-1 was examined in these glioma cell lines by western blot analysis. All glioma lines secreted significant levels of TSP-1. Bioassay showed that all tumor lines had the capacity to activate latent TGF-beta. Localization of TSP-1, TGF-beta1, -beta2, and -beta3 was examined immunohistochemically in surgically resected glioma tissues, including 11 glioblastomas, six anaplastic astrocytomas, and eight astrocytomas. Most glioblastomas expressed high levels of both TSP-1 and TGF-beta. Anaplastic astrocytomas expressed moderate levels of TSP-1 and TGF-beta. Most malignant gliomas expressed various levels of TGF-beta1, -beta2, and -beta3. The expression of both proteins, however, was weak in low-grade gliomas. Normal brain tissues around the tumors were negatively or very weakly positively stained for TSP-1 and TGF-beta. These results indicate that most malignant glioma cells express TSP-1 in vitro and in vivo, and the expression of TSP-1 and TGF-beta in vivo correlates with the histologic malignancy of glioma. Overexpression of both TSP-1 and TGF-beta may increase the biologic malignancy of malignant gliomas, through generating the active form of TGF-beta in tumor tissues.

    Topics: Astrocytoma; Biomarkers, Tumor; Diagnosis, Differential; Glioblastoma; Glioma; Humans; RNA, Messenger; Thrombospondin 1; Thrombospondins; Transforming Growth Factor beta; Tumor Cells, Cultured

2000
Expression of cytokines by human astrocytomas following stimulation by C3a and C5a anaphylatoxins: specific increase in interleukin-6 mRNA expression.
    Journal of neurochemistry, 1999, Volume: 72, Issue:6

    C3a and C5a anaphylatoxins are two proinflammatory peptides generated during complement activation that act through distinct Gi protein-coupled receptors named C3aR and C5aR, respectively. We have demonstrated previously that human astrocytes expressed C3aR and C5aR constitutively and were able to produce a functional complement. In this study, we examined the effect of an anaphylatoxin stimulation on cytokine expression by human astrocyte cell lines. Interleukin (IL)-1beta, IL-6, tumor necrosis factor-alpha, and transforming growth factor-beta mRNA expression was studied by quantitative RT-PCR. Whereas IL-1beta, tumor necrosis factor-alpha, and transforming growth factor-beta mRNA levels remained unchanged, stimulation of astrocytoma cells (T98G, CB193, U118MG) by C3a, C5a, and peptidic C3aR and C5aR agonists induced an increase in the IL-6 mRNA level. The amount of IL-6 was markedly increased at 3 and 6 h and returned to the basal level at 9 h of stimulation. This response was specific, because pretreatment of cells with pertussis toxin or with polyclonal anti-C3aR or anti-C5aR antibodies completely blocked the IL-6 mRNA increase. The IL-6 response was also investigated at the protein level, but IL-6 protein was detected neither in cell lysates nor in supernatants of stimulated cells. The anaphylatoxin-mediated transcriptional activation of IL-6 gene suggests that C3a and C5a could play a role in priming glial cells during the inflammatory process in the brain.

    Topics: Anaphylatoxins; Antibodies; Antigens, CD; Astrocytoma; Complement C3a; Complement C5a; Cytokines; Gene Expression Regulation, Neoplastic; Humans; Interleukin-1; Interleukin-6; Kinetics; Membrane Proteins; Pertussis Toxin; Receptor, Anaphylatoxin C5a; Receptors, Complement; Reverse Transcriptase Polymerase Chain Reaction; RNA, Messenger; Time Factors; Transcription, Genetic; Transforming Growth Factor beta; Tumor Cells, Cultured; Virulence Factors, Bordetella

1999
Differential effects of transforming growth factor-beta(s) and glial cell line-derived neurotrophic factor on gene expression of presenilin-1 in human post-mitotic neurons and astrocytes.
    Neuroscience, 1999, Volume: 93, Issue:3

    Mutations in the presenilin-1 gene are linked to the majority of early-onset familial Alzheimer's disease cases. We have previously shown that the expression of transforming growth factor-beta is altered in Alzheimer's patients, compared to controls. Here we examine presenilin- expression in human post-mitotic neurons (hNT cells), normal human astrocytes, and human brain tumor cell lines following treatment with three isoforms of transforming growth factor-beta, or glial cell line-derived neurotrophic factor, a member of the transforming growth factor-beta superfamily. As the NT2/D1 teratocarcinoma cell line is treated with retinoic acid to induce differentiation to hNT cells, presenilin-1 messenger RNA expression is dramatically increased. Furthermore, there is a 2-3-fold increase in presenilin-1 messenger RNA expression following treatment of hNT cells with growth factors and similar results are found by Western blotting and with immunohistochemical staining for presenilin-1 protein. However, treatment of normal human astrocytes with cytokines results in minimal changes in presenilin-1 messenger RNA and protein. Interestingly, the expression of presenilin-1 in human U87 MG astrocytoma and human SK-N-SH neuroblastoma cells is only increased when cells are treated with glial cell line-derived neurotrophic factor or transforming growth factor-beta3. These findings suggest that endogenous presenilin-1 gene expression in human neurons can be induced by growth factors present in normal and diseased brain tissue. Cytokines may play a major role in regulating expression of presenilin-1 which may affect its biological actions in physiological and pathological conditions.

    Topics: Astrocytes; Astrocytoma; Blotting, Western; Brain Neoplasms; Gene Expression Regulation; Glial Cell Line-Derived Neurotrophic Factor; Glioblastoma; Humans; Membrane Proteins; Neoplasm Proteins; Nerve Growth Factors; Nerve Tissue Proteins; Neuroblastoma; Neurons; Presenilin-1; Protein Isoforms; Reverse Transcriptase Polymerase Chain Reaction; RNA, Messenger; RNA, Neoplasm; Teratocarcinoma; Transforming Growth Factor beta; Tretinoin; Tumor Cells, Cultured

1999
Human astrocytomas co-expressing Fas and Fas ligand also produce TGFbeta2 and Bcl-2.
    Journal of neuro-oncology, 1999, Volume: 44, Issue:3

    Human astrocytomas frequently co-express Fas (APO-1/CD95) and Fas ligand (FasL), yet do not appear to be overly susceptible to suicidal, fratricidal and immune-mediated elimination. This suggests that these gliomas have acquired mechanisms to prevent Fas-mediated apoptosis from occurring. Candidates for such a role include transforming growth factor-beta2 (TGFbeta2) and B-cell lymphoma/leukemia-2 (Bcl-2). TGFbeta2 effectively functions by hiding tumor cells from the immune system. This may potentially prevent the delivery of FasL from cytolytic T cells to Fas bearing astrocytomas. Bcl-2 works by rendering gliomas resistant to Fas-mediated apoptosis. Using immunohistochemistry, we analyzed seventy-six human astrocytomas (11 World Health Organization (WHO) grade I, 17 grade II, 17 grade III, and 31 grade IV) for the expression of Fas, FasL, TGFbeta2 and Bcl-2 in vivo. Positive immunoreactivity was found to significantly increase with increasing tumor grade for Fas (p < 0.0002), FasL (p < 0.0001), TGFbeta2 (p < 0.001) and Bcl-2 (p < 0.01). In addition, Fas/FasL co-expression, a counter-intuitive combination of factors in regards to glioma survival, also increased with WHO grade. Forty-five of 76 (59%) astrocytomas co-expressed Fas and FasL. Of those co-expressing Fas and FasL, 44 of 45 (98%) produced TGFbeta2, and 26 of 45 (58%) expressed Bcl-2. We found a significant positive correlation between Fas/FasL co-expression and TGFbeta2 (p < 0.002) and Bcl-2 (p < 0.005) production. We conclude that Fas and FasL are frequently co-expressed in human astrocytomas and these tumors are likely to produce other immunosuppressive and antiapoptotic factors such as TGFbeta2 and Bcl-2.

    Topics: Astrocytoma; Fas Ligand Protein; fas Receptor; Glioblastoma; Humans; Immunohistochemistry; Membrane Glycoproteins; Proto-Oncogene Proteins c-bcl-2; Transforming Growth Factor beta

1999
Interleukin-1 alpha (IL-1 alpha), IL-1 beta, IL-1 receptor type I, IL-1 receptor antagonist, and TGF-beta 1 mRNAs in pediatric astrocytomas, ependymomas, and primitive neuroectodermal tumors.
    Molecular and chemical neuropathology, 1998, Volume: 33, Issue:2

    Interleukin-1 alpha (IL-1 alpha), IL-1 beta, interleukin-1 receptor type I (IL-1RI, signaling receptor), and IL-1 receptor antagonist (IL-1Ra, endogenous inhibitor) are pivotal components of the IL-1 system. IL-1 and other cytokines induced by IL-1, such as TGF-beta 1, may participate in the growth of various tumor cells. In children, primary nervous system tumors represent the most common solid malignancy. We investigated the levels of IL-1 alpha, IL-1 beta, IL-1RI, IL-1Ra, and TGF-beta 1 mRNAs in pediatric astrocytomas (n = 19), ependymomas (n = 13), and primitive neuroectodermal tumors (n = 22) using sensitive and specific RNase protection assays. The data show a significant distinct cytokine mRNA profile among brain tumor types. Pilocytic, nonpilocytic, and anaplastic astrocytomas have significant increased levels of IL-1 beta, IL-1RI, and TGF-beta 1 mRNAs, but low levels of IL-1Ra mRNA; this may have implications for an IL-1 beta feedback system and IL-1 beta<-->TGF-beta 1 interactions in astrocytomas. Ependymomas show increased levels of IL-1 alpha and IL-1 beta mRNAs associated with low levels of IL-1Ra mRNA; primitive neuroectodermal tumors do not exhibit increased levels of any cytokine component examined. The data also suggest that a dysregulation of the balance between stimulatory and inhibitory cytokines may be involved in the growth and development of brain tumors via autocrine/paracrine mechanisms.

    Topics: Adolescent; Astrocytoma; Brain Neoplasms; Child; Child, Preschool; Ependymoma; Female; Humans; Interleukin 1 Receptor Antagonist Protein; Interleukin-1; Male; Neuroectodermal Tumors, Primitive; Receptors, Interleukin-1; RNA, Messenger; Sialoglycoproteins; Transforming Growth Factor beta

1998
Neural cell adhesion molecule L1 in gliomas: correlation with TGF-beta and p53.
    Journal of clinical pathology, 1998, Volume: 51, Issue:1

    To assess immunohistochemically whether the neural cell adhesion molecule L1, which is a member of the immunoglobulin superfamily and has been shown recently to be a stimulating factor for glioma migration, is expressed in glioma tissues, and to investigate factors that can regulate this expression.. Twenty seven glioma tissue specimens including 13 glioblastomas, seven anaplastic astrocytomas, and seven astrocytomas were examined. Immunohistochemical analyses of L1, p53, and transforming growth cell factor beta (TGF-beta) were performed on each tumour using both polyclonal and monoclonal antibodies.. Nine (33%) specimens (six glioblastomas and three anaplastic astrocytomas) had L1 positive immunostaining. p53 positive staining was detected in 10 (43%) of 23 glioma specimens (seven glioblastomas and three anaplastic astrocytomas). TGF-beta positive immunostaining was observed in 12 (52%) of the 23 glioma specimens (six glioblastomas, four anaplastic astrocytomas, and two astrocytomas). There was a statistical correlation between both p53 and L1 expression and TGF-beta and L1 expression. No such correlation was found between p53 and TGF-beta expression.. These results suggest that mutation of the p53 gene or expression of TGF-beta may upregulate the expression of the L1 gene, thus resulting in high grade migration of glioma cells.

    Topics: Antigens, Neoplasm; Astrocytoma; Glioblastoma; Glioma; Humans; Immunoenzyme Techniques; Leukocyte L1 Antigen Complex; Neoplasm Proteins; Neural Cell Adhesion Molecules; Transforming Growth Factor beta; Tumor Suppressor Protein p53

1998
Constitutive and cytokine-inducible expression of prion protein gene in human neural cell lines.
    Journal of neuropathology and experimental neurology, 1998, Volume: 57, Issue:2

    Prion diseases are a group of neurodegenerative disorders characterized by intracerebral accumulation of a protease-resistant prion protein (PrP(Sc)) that causes extensive neuronal degeneration and astrogliosis. The regulation of prion protein (PrP) gene expression by a panel of glial and neuronal cytokines (TNF-alpha, IFN-gamma, IL-1beta, IL-10, and TGF-beta1) was investigated in human neural cell lines by reverse transcription-polymerase chain reaction and Northern blot analysis. The constitutive expression of PrP mRNA was identified in all human neural cell lines and tissues examined including Y79 retinoblastoma, IMR-32 neuroblastoma, SK-N-SH neuroblastoma, U-373MG astrocytoma, KG-1-C glioma, NTera2 teratocarcinoma, NTera2-derived differentiated neurons (NTera2-N), peripheral nerve, and cerebral and cerebellar tissues. In SK-N-SH cells, a 48 hour (h) treatment with 100 ng/ml IL-1beta, 100 ng/ml TNF-alpha, or 100 nM phorbol 12-myristate 13-acetate induced a 2.7- to 4.2-fold increase in the level of PrP mRNA, while the exposure to 100 ng/ml IFN-gamma resulted in a 50% decrease. By contrast, none of these cytokines significantly altered the levels of PrP mRNA in IMR-32, NTera2-N, or U-373MG cells. These results indicate that the PrP gene expression is constitutive in a wide range of human neural cell lines and tissues where it is controlled by cell type-specific regulatory mechanisms.

    Topics: Astrocytoma; Brain; Brain Neoplasms; Cell Line; Cerebellum; Cytokines; Eye Neoplasms; Gene Expression Regulation; Humans; Interferon-gamma; Interleukin-1; Interleukin-10; Neuroblastoma; Neuroglia; Neurons; Polymerase Chain Reaction; Prions; Retinoblastoma; RNA, Messenger; Teratocarcinoma; Tetradecanoylphorbol Acetate; Transcription, Genetic; Transforming Growth Factor beta; Tumor Cells, Cultured; Tumor Necrosis Factor-alpha

1998
Receptor expression, cytogenetic, and molecular analysis of six continuous human glioma cell lines.
    In vitro cellular & developmental biology. Animal, 1998, Volume: 34, Issue:6

    Six human glioma cell lines were established from tissues obtained from five patients diagnosed with Kernohan grade IV glioblastoma multiforme and one from a patient with a grade II astrocytoma. One line was from a recurrent patient who had received prior therapy; the other lines were derived from patients at initial diagnosis and/or before cytoreductive therapies other than surgery were given. Considerable variability in phenotypic, karyotypic, and cell surface marker expression was displayed between the six human glioma cell lines. The karyotypes ranged from apparently normal (grade II astrocytoma) to those with complex rearrangements. Trisomy of chromosome 7 was the most common abnormality. The extensive cytogenetic and molecular characterization of these lines may facilitate their utilization in cellular and molecular biologic studies.

    Topics: Adult; Aged; Animals; Astrocytoma; Biomarkers, Tumor; Brain Neoplasms; DNA, Neoplasm; Female; Glioblastoma; Humans; Immunoenzyme Techniques; In Situ Hybridization, Fluorescence; Karyotyping; Male; Middle Aged; Polymerase Chain Reaction; Receptors, Platelet-Derived Growth Factor; Transforming Growth Factor beta; Tumor Cells, Cultured

1998
TGF-beta suppresses IFN-gamma induction of class II MHC gene expression by inhibiting class II transactivator messenger RNA expression.
    Journal of immunology (Baltimore, Md. : 1950), 1997, Mar-01, Volume: 158, Issue:5

    Recently, a non-DNA binding protein, class II transactivator (CIITA), has been shown to be required for constitutive and IFN-gamma-inducible class II MHC transcription. The cytokine TGF-beta inhibits IFN-gamma-induced class II MHC expression at the transcriptional level. In this study, we provide evidence that TGF-beta blocks IFN-gamma-induced CIITA mRNA accumulation. TGF-beta down-regulates class II MHC and CIITA mRNA accumulation in human astroglioma and fibrosarcoma cell lines, but TGF-beta does not destabilize the CIITA message, suggesting an effect at the transcriptional level. In cells that stably overexpressed CIITA, leading to a constitutive class II MHC-positive phenotype, the inhibitory effect of TGF-beta on class II MHC was abrogated, but the cells remained responsive for expression of TGF-beta-inducible genes. Cell lines that possessed defects in TGF-beta signaling also became refractory to inhibition of IFN-gamma-induced CIITA and class II MHC expression. Our data indicate that TGF-beta suppresses IFN-gamma-induced class II MHC expression by inhibiting accumulation of CIITA mRNA.

    Topics: Astrocytoma; Fibrosarcoma; Gene Expression Regulation, Neoplastic; Gene Transfer Techniques; Genes, MHC Class II; Humans; Interferon-gamma; Nuclear Proteins; RNA, Messenger; Signal Transduction; Tetracycline; Trans-Activators; Transforming Growth Factor beta; Tumor Cells, Cultured

1997
Characterization of a spontaneous murine astrocytoma and abrogation of its tumorigenicity by cytokine secretion.
    Neurosurgery, 1997, Volume: 41, Issue:6

    The promise of immunotherapies developed against brain tumors in animal models has not been realized in human clinical trials. This may be because of the routine use of rodent tumors artificially induced by chemicals or viruses that do not accurately portray the intrinsic qualities of spontaneously arising human tumors and that often fail to incorporate the role of immunosuppressants, such as transforming growth factor-beta, that are secreted by human gliomas. From an astrocytoma that arose spontaneously in inbred VM/Dk mice, we have characterized a highly tumorigenic spontaneous murine astrocytoma cell line (SMA-560) that retains features of glial differentiation and naturally produces high levels of biologically active transforming growth factor-beta. We have used this model to determine whether cytokine production by tumor cells will inhibit intracerebral astrocytoma growth.. Packaging cell lines producing replication-incompetent retroviral vectors were used to transfect the SMA-560 cell line in vitro with the genes encoding the murine cytokines interleukin (IL)-2, IL-3, IL-4, IL-6, tumor necrosis factor-alpha, gamma-interferon, or granulocyte-macrophage colony-stimulating factor or the costimulatory molecule B7.1 (CD80).. Mice challenged intracerebrally with 5000 untransfected SMA-560 cells all succumbed to tumor within 30 days, with a median survival of 25 days. In contrast, mice challenged with SMA-560 cells producing IL-2, IL-4, or tumor necrosis factor-alpha each had a more than 400% increase in median survival (P < 0.0001). In these groups, 78.3% (18 of 23 mice), 66.7% (10 of 15 mice), and 60% (6 of 10 mice) of the mice, respectively, remained alive without evidence of tumor for longer than 100 days after the initial tumor challenge. All other cytokines tested and the expression of B7.1 failed to result in an increase in median survival.. Using a spontaneous astrocytoma model in an inbred mouse strain, we have shown that cytokine production by glial tumors can abrogate their tumorigenicity in vivo despite production of transforming growth factor-beta. These results predict that approaches directed at cytokine production within intracerebral astrocytomas may be efficacious in human trials and that the "immunological privilege" of the brain may not be absolute under such conditions.

    Topics: Animals; Astrocytoma; Brain Neoplasms; Carcinogenicity Tests; Cytokines; Female; Genetic Vectors; Histocompatibility Antigens Class I; Immunohistochemistry; Mice; Mice, Inbred Strains; Retroviridae; Transfection; Transforming Growth Factor beta; Tumor Cells, Cultured

1997
Stimulus-specific inhibition of intracellular adhesion molecule-1 gene expression by TGF-beta.
    Journal of immunology (Baltimore, Md. : 1950), 1996, Jul-15, Volume: 157, Issue:2

    Astrocytes and microglia, the two major glial cells within the central nervous system, can function as immune effector cells upon activation. Intercellular adhesion molecule-1 (ICAM-1), a cell surface glycoprotein involved in extravasation into inflamed tissue and Ag-specific activation of T lymphocytes, can be induced in astrocytes and microglia by numerous stimuli. In this study, we investigated the role of TGF-beta, an immunosuppressive cytokine, in regulating ICAM-1 expression in glial cells. We previously demonstrated that TNF-alpha, IL-1 beta, IFN-gamma, or IFN-gamma plus LPS (IFN-gamma/LPS) can enhance ICAM-1 expression in astrocytes, while microglia express ICAM-1 only in response to IFN-gamma or IFN-gamma/LPS. TGF-beta alone has a minimal effect on constitutive ICAM-1 expression in either astrocytes or microglia, but inhibits, in a time-dependent manner, TNF-alpha- or IL-1 beta-induced ICAM-1 mRNA and protein expression in astrocytes. Interestingly, TGF-beta has no effect on IFN-gamma- or IFN-gamma/LPS-induced ICAM-1 expression in astrocytes or microglia. Inhibition of TNF-alpha- or IL-1 beta-induced ICAM-1 mRNA levels by TGF-beta in astrocytes was not due to degradation of ICAM-1 message, rather, inhibition was mediated at the transcriptional level. Similar results were observed in two human astroglioma cell lines, CRT and STT; TGF-beta inhibited TNF-alpha- and IL-1 beta-induced ICAM-1 expression, but IFN-gamma induction of ICAM-1 was unaffected. These results indicate that TGF-beta suppresses ICAM-1 expression in glial cells in a stimulus-specific manner.

    Topics: Astrocytoma; Cells, Cultured; Cytokines; Drug Stability; Gene Expression Regulation; Humans; Intercellular Adhesion Molecule-1; Microglia; RNA, Messenger; Transcription, Genetic; Transforming Growth Factor beta; Tumor Cells, Cultured; Tumor Necrosis Factor-alpha

1996
The IE2 regulatory protein of human cytomegalovirus induces expression of the human transforming growth factor beta1 gene through an Egr-1 binding site.
    Journal of virology, 1996, Volume: 70, Issue:10

    Increases in transforming growth factor beta1 (TGF-beta1) mRNA and biological activity in the early phase of human cytomegalovirus (CMV) infection in fibroblasts are paralleled by increased TGF-beta1-chloramphenicol acetyltransferase (CAT) reporter gene activity. To determine how CMV infection transactivates the TGF-beta1 promoter, we examined the effects of the cotransfected IE2 regulatory protein of human CMV on 5'-deleted TGF-beta1 promoter-CAT reporter genes in transient DNA transfection assays. Two upstream TGF-beta1 promoter regions each containing an Egr-1 consensus site were shown to be important for IE2-induced transactivation in a cell type that displayed greatly reduced nonspecific activity. Furthermore, transfer of an Egr-l site from between positions -125 and -98, but not point mutant versions of this site, to a heterologous promoter also conveyed IE2 responsiveness. Addition of an IE2 expression vector or use of the U373 A45 astrocytoma cell line expressing IE2 also produced synergistic stimulation of GAL4-Egr-l-mediated activation of a target promoter containing GAL4 binding sites. The 80-kDa IE2 protein present in A45 cells proved to selectively bind to glutathione S-transferase (GST)-Egr-1 beads. The results of in vitro protein binding assays also revealed that an intact in vitro-translated IE2 protein bound directly to the GST-Egr-1 fusion protein through the zinc finger domain of the Egr-1 protein and that this binding activity was abolished by deletion of parts of the zinc finger DNA-binding domain. Similarly, the Egr-1 protein was found to associate preferentially with a small region within the C-terminal half of the IE2 protein adjacent to the DNA-binding and dimerization domains that are important for both transactivation and downregulation. We conclude from these observations that IE2 may regulate transcription of the TGF-beta1 gene as well as other potential cellular targets by virtue of its ability to interact with the Egr-1 DNA-binding protein.

    Topics: Astrocytoma; Cytomegalovirus; DNA-Binding Proteins; Early Growth Response Protein 1; Gene Expression Regulation, Viral; Humans; Immediate-Early Proteins; Transcription Factors; Transcriptional Activation; Transforming Growth Factor beta; Tumor Cells, Cultured; Viral Proteins

1996
Secretion of transforming growth factor-beta 1 and -beta 2 by malignant glioma cells.
    Neurologia medico-chirurgica, 1995, Volume: 35, Issue:7

    The secretion of transforming growth factor-beta (TGF-beta), a growth inhibitory factor with immunosuppressive properties, was investigated in one glioblastoma cell line and seven surgically resected malignant glioma cells. Cultured cells from surgically resected tumors were examined immunohistochemically for glial fibrillary acidic protein (GFAP) and S-100 protein. The levels of TGF-beta 1 and TGF-beta 2 in culture supernatants from malignant glioma cells were determined by a specific bioassay using anti-TGF-beta 1 and anti-TGF-beta 2 antibodies. Two glioblastoma cell lines were cultured in the presence of TGF-beta 1 or TGF-beta 2 to assess the effect of TGF-beta on the growth of glioblastoma cells. Cultured cells from surgically resected tumors were positive for both GFAP and S-100 protein. Both active and latent forms of TGF-beta 1 and TGF-beta 2 were detected in the culture supernatants from malignant gliomas, except in one patient with anaplastic astrocytoma which secreted only latent forms of TGF-beta 1 and TGF-beta 2. There was no statistical difference in the levels of TGF-beta 1 and TGF-beta 2 in glioblastomas and anaplastic astrocytomas. Neither TGF-beta 1 nor TGF-beta 2 affected the growth of glioblastoma cells. These findings suggest that most malignant glioma cells secrete both TGF-beta 1 and TGF-beta 2, can convert TGF-beta from a latent to active form, and may suppress cytokine secretion by activated lymphocytes in vivo as well as in vitro.

    Topics: Astrocytoma; Brain Neoplasms; Cell Division; Cell Line; Glioblastoma; Humans; Immune Tolerance; Peptides; Transforming Growth Factor beta; Tumor Cells, Cultured

1995
Inhibition of proliferation and induction of differentiation in medulloblastoma- and astrocytoma-derived cell lines with phenylacetate.
    Journal of neurosurgery, 1995, Volume: 83, Issue:4

    The authors investigated the effects of a nontoxic differentiation inducer, phenylacetate (PA), on neuroectodermal tumor-derived cell lines. Treatment of medulloblastoma (Daoy and D283 MED) and glioma (U-251MG, C6, and RG2) cell lines resulted in a dose-dependent decline in DNA synthesis and cell proliferation, associated with accumulation in the G0/G1 phase of the cell cycle. Phenylacetate decreased transforming growth factor (TGF)-beta 2 production by medulloblastoma Daoy cells. Neutralizing antibodies against either TGF beta 2 or TGF beta 1 failed to block the growth arrest observed. This suggests that, unlike other differentiation agents, such as retinoic acid, the effect of PA on medulloblastoma proliferation is not mediated by a TGF beta pathway. In addition to cytostasis, PA induced marked morphological changes in U-251MG and C6 glioma cells associated with increased abundance of glial fibrillary acidic protein-positive processes. Although the morphology of PA-treated medulloblastoma cells was not significantly altered, the D283 MED cells exhibited increased expression of neurofilament proteins and Hu antigen, indicative of differentiation along a neuronal pathway. The effects of PA on the medulloblastoma cell lines were compared to its effects on the human neuroblastoma cell line BE(2)C, which is capable of a bidirectional differentiation toward a neuronal or a glial/schwann cell pathway. In BE(2)C cells, PA induced differentiation toward a schwann/glial cell-like phenotype, suggesting that the choice of differentiation pathway is cell type and agent specific. These in vitro antiproliferative and differentiation inducing effects of PA suggest that this agent warrants further evaluation as a potential therapeutic modality for the treatment of medulloblastoma and malignant glioma in humans.

    Topics: Animals; Astrocytoma; Cell Differentiation; Cell Division; Cell Line; DNA, Neoplasm; Dose-Response Relationship, Drug; ELAV Proteins; G1 Phase; Gene Expression Regulation, Neoplastic; Glial Fibrillary Acidic Protein; Humans; Medulloblastoma; Mice; Nerve Tissue Proteins; Neuroblastoma; Neurofilament Proteins; Neuroglia; Neurons; Phenotype; Phenylacetates; Rats; Resting Phase, Cell Cycle; RNA-Binding Proteins; Schwann Cells; Transforming Growth Factor beta

1995
Transforming growth factor beta-1 (TGF-beta 1) potentiates IL1 alpha-induced IL6 mRNA and cytokine protein production in a human astrocytoma cell line.
    Oncology research, 1993, Volume: 5, Issue:10-11

    We analyzed the response of human astrocytoma cell line U373-MG to various cytokines by measuring the production of interleukin-6 (IL6) mRNA and cytokine protein. Interferon gamma (IFN gamma), transforming growth factor beta 1 (TGF-beta 1), granulocyte-macrophage colony-stimulating factor (GM-CSF) and granulocyte-colony-stimulating factor (G-CSF) did not induce IL6 mRNA production; however, IL6 mRNA expression and protein production was strongly induced by IL1 alpha and to a lesser extent by IFN alpha. The IL6 mRNA expression induced by IL1 alpha was potentiated by TGF-beta 1 and IFN alpha and slightly decreased by IFN gamma. The potentiation of cytokine mRNA accumulation by TGF-beta 1 was both time- and concentration-dependent. Induction of IL6 mRNA by IL1 alpha was optimally potentiated either if U373-MG cells were pretreated with TGF-beta 1 or if TGF-beta 1 was added within 30 min after stimulation with IL1 alpha. The potentiation of IL6 mRNA by TGF-beta 1 required de novo synthesis of an intermediate protein since treatment with cycloheximide abrogated the amount of mRNA enhanced by TGF-beta 1 without affecting IL1 alpha-driven mRNA production. Nuclear run-on analyses demonstrated increased transcriptional activity of the IL6 gene when stimulated with IL1 alpha in the presence of TGF-beta 1. However, actinomycin-D pulse chase experiments showed that TGF-beta 1 did not increase the stability of IL6 mRNA. Thus, in concert, the results demonstrate that TGF-beta 1 potentiates IL6 production in astrocytoma cells by promoting the transcriptional activity of the IL6 gene and requires coexpression of new proteins. Since cytokines can provide potent mitogenic signals to tumor cells, the results presented here further suggest that the antitumor effect of combination cytokine therapy might partly depend on heterotypic interactions between tumor cells and cytokines.

    Topics: Astrocytoma; Cell Line; Cell Nucleus; Cytokines; Drug Synergism; Gene Expression; Humans; Interleukin-1; Interleukin-6; Kinetics; RNA, Messenger; Time Factors; Transcription, Genetic; Transforming Growth Factor beta; Tumor Cells, Cultured

1993
Inhibition of interferon-gamma-induced major histocompatibility complex class II gene transcription by interferon-beta and type beta 1 transforming growth factor in human astrocytoma cells. Definition of cis-element.
    The Journal of biological chemistry, 1993, Sep-05, Volume: 268, Issue:25

    To address mechanisms by which cytokines inhibit interferon-gamma (IFN gamma)-induced gene expression in astrocytic cells, we have been studying effects of type beta 1 transforming growth factor (TGF beta 1) and interferon-beta (IFN beta) on IFN gamma-induced expression of the well-characterized human major histocompatibility complex (MHC) class II gene DRA. This investigation was motivated by the observations that IFN gamma-induced expression of MHC class II antigen expression on astrocytic cells can be blocked in a tissue-specific fashion by several cytokines and neurotransmitters in tissue culture and that astrocyte expression of MHC class II is severely restricted in vivo. We previously showed that IFN beta inhibited IFN gamma-induced DRA expression at the transcriptional level. This inhibition was not global, since IFN gamma-induction of intercellular adhesion molecule-1 was not affected. Here, TGF beta 1-mediated inhibition of DRA is shown to exhibit similar characteristics. To address the mechanism of this inhibition, sequence requirements for IFN beta and TGF beta 1 to suppress IFN gamma-induced transcription of DRA were determined. A 135-base pair DRA sequence element containing the IFN gamma-responsive region and transcriptional start site was sufficient to direct IFN beta- or TGF beta 1-mediated suppression of a reporter gene. These experiments suggested that either IFN beta or TGF beta 1 could repress IFN gamma-induced DRA transcription directly, without requiring a cis-element extrinsic to the IFN gamma-inducible DRA sequences. Consistently similar effects of IFN beta and TGF beta 1 observed in these experiments prompted comparison of other gene regulatory effects of the two cytokines. TGF beta 1, unlike IFN beta, induced gene expression directed by upstream elements of the gene encoding plasminogen activator inhibitor type-1. IFN beta, unlike TGF beta 1, enhanced levels of 2'-5'-oligoadenylate synthetase activity. Additionally, direct assay of TGF beta 1 and monoclonal anti-TGF beta antibody blocking experiments were used to determine that cells treated with IFN beta did not produce increased amounts of active or latent TGF beta. These data argued that IFN beta and TGF beta 1 utilized distinct pathways to mediate the inhibition of IFN gamma-induced DRA transcription.

    Topics: 2',5'-Oligoadenylate Synthetase; Astrocytoma; Cell Adhesion Molecules; Chloramphenicol O-Acetyltransferase; Gene Expression; Genes, MHC Class II; HLA-DR Antigens; Humans; Intercellular Adhesion Molecule-1; Interferon-beta; Interferon-gamma; Transcription, Genetic; Transfection; Transforming Growth Factor beta; Tumor Cells, Cultured

1993
Effect of the expression of transforming growth factor-beta 2 in primary human glioblastomas on immunosuppression and loss of immune surveillance.
    Journal of neurosurgery, 1992, Volume: 76, Issue:5

    Glioblastomas are malignant brain tumors that are attended by an immunosuppressed state. The authors have studied the expression of transforming growth factor-beta 2, which is known to have potent immunosuppressive and angiogenic properties. Transforming growth factor-beta 2 messenger ribonucleic acid and its protein product are both found to be greatly overexpressed in these tumors and are absent from normal brain tissue. The overexpression of this growth factor may contribute to the escape of neoplastic astrocytes from immune surveillance and, furthermore, to the immunosuppressed state that is characteristic of many of these patients.

    Topics: Astrocytoma; Blotting, Northern; Brain Neoplasms; Gene Expression; Glioma; Humans; Immune Tolerance; Immunologic Surveillance; Neovascularization, Pathologic; Nucleic Acid Hybridization; RNA, Messenger; Transforming Growth Factor beta

1992
Distribution of transforming growth factor-beta 1 in human astrocytomas.
    Human pathology, 1992, Volume: 23, Issue:11

    We used immunohistochemical techniques to study the distribution of transforming growth factor-beta 1 (TGF-beta 1) and infiltrating lymphocytes and macrophages in human astrocytomas. Thirteen of 15 grade 4 astrocytomas (glioblastomas) showed staining with anti-TGF-beta 1 antibody, predominantly in proliferating endothelial complexes and surrounding small and medium-sized blood vessels. Brain tissue microscopically free of tumor cells (n = 8) and more differentiated astrocytomas of varying grade (1 to 3; n = 6) devoid of endothelial proliferation did not stain with anti-TGF-beta 1. Normal brain contained only rare lymphoreticular cells. The majority of astrocytomas studied, however, contained T lymphocytes and macrophages with smaller numbers of B lymphocytes. The lymphoreticular infiltrates were concentrated primarily in close proximity to blood vessels. Within an individual tumor perivascular regions staining for TGF-beta 1 never contained more than occasional T lymphocytes. Perivascular regions not staining for TGF-beta 1 frequently contained low to high numbers of T lymphocytes. The inverse relationship in the distribution of TGF-beta 1 and lymphocyte infiltrates is compatible with a functional relationship between this cytokine and an immune effector cell response to glioblastomas.

    Topics: Astrocytoma; Brain Neoplasms; Humans; Immunoenzyme Techniques; Lymphocytes, Tumor-Infiltrating; Macrophages; Transforming Growth Factor beta

1992
Transforming growth factor-beta 1 differentially regulates proliferation and MHC class-II antigen expression in forebrain and brainstem astrocyte primary cultures.
    Brain research, 1992, Jul-10, Volume: 585, Issue:1-2

    To facilitate investigation of cytokine regulation of reactive astrogliosis, primary astrocyte cultures from neonatal murine forebrain and brainstem were established. Forebrain and brainstem astrocytes proliferated at a similar rate under basal culture conditions, and both were growth-inhibited by treatment with recombinant murine interferon-gamma. The growth of cultured brainstem astrocytes was significantly enhanced by exposure to recombinant human transforming growth factor-beta 1. In contrast, proliferation of forebrain astrocytes was not significantly affected by transforming growth factor-beta 1. The disparate responses of brainstem and forebrain astrocytes to transforming growth factor-beta 1 treatment were not limited to effects on cell growth, since transforming growth factor-beta 1 could block interferon-gamma-induced MHC class-II antigen expression on cultured brainstem astrocytes but not on forebrain cells. Results could not be attributed to use of an heterologous cytokine/cellular target system, since similar variability in transforming growth factor-beta 1 modulation of major histocompatibility complex antigen expression could be demonstrated using two human astrocytoma cell lines. This report is the first to document mitogenic response to transforming growth factor-beta 1 for neuroepithelial cells. The role of transforming growth factor-beta 1 in regulating aspects of reactive astrogliosis, particularly in the context of inflammatory demyelination, requires further investigation. Furthermore, these studies may provide insight into regional variability in the sequelae of inflammation within the central nervous system.

    Topics: Animals; Astrocytes; Astrocytoma; Brain Stem; Cell Division; Histocompatibility Antigens Class II; Interferon-gamma; Prosencephalon; Transforming Growth Factor beta; Tumor Cells, Cultured

1992
Expression of platelet-derived growth factor and transforming growth factor and their correlation with cellular morphology in glial tumors.
    Journal of neurosurgery, 1991, Volume: 75, Issue:3

    This study was undertaken to evaluate the role of two sets of growth factors, platelet-derived growth factor (PDGF) and transforming growth factor-beta (TGF-beta), in the induction and maintenance of glial tumors and their phenotypic expression. Explants from eight malignant tumors, five benign tumors, and two nontumor glial cells were analyzed for levels of messenger ribonucleic acid (mRNA) expression of PDGFA, PDGFB, TGF-beta 1, and TGF-beta 2. Results were normalized to the mRNA expression of tubulin, a "housekeeping" gene present in glial cells. Of the 15 explants tested, PDGFB was seen in six, all of which were malignant tumors; PDGFA was seen in all 15 with much higher levels expressed in malignant tumors; and TGF-beta 1 and TGF-beta 2 were seen in all 15 without a clear difference between cell types, although expression tended to be higher in malignant tumors. This project supports the theory that the induction and maintenance of glial tumors is likely to be a multifactorial phenomenon.

    Topics: Astrocytoma; Cells, Cultured; Gene Expression Regulation, Neoplastic; Glioma; Humans; Neoplasms, Germ Cell and Embryonal; Neuroglia; Nucleic Acid Hybridization; Platelet-Derived Growth Factor; RNA, Messenger; RNA, Neoplasm; Transforming Growth Factor beta; Tumor Cells, Cultured

1991