Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
Brain Pathol ; 11(3): 296-305, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11414472

ABSTRACT

The p53 gene is normally wild type in meningiomas. Since all three members of the p53 gene family recognize the same DNA sequence, tumors containing wild type p53 could decrease transactivation of p53 target genes by mutating either p63 or p73. In meningiomas the most likely target is p73, because loss of heterozygosity of the chromosomal band containing p73 is the commonest genetic lesion in these tumors. To screen p73 for mutations we have developed a functional assay which tests the ability of p73 to activate transcription from a p53-responsive promoter in yeast. The assay correctly identified p73 mutants with mutations equivalent to hotspot mutations in p53, demonstrating that the assay can detect transcriptionally inactive p73. No mutations in p73 were identified in meningiomas. p73 RNA level was higher in more advanced tumors, but there was no correlation between the expression level of p73 and p21, a known p53 target gene. The yeast assay was also used to measure the intrinsic sensitivity of the p73 protein to mutagenesis. Like p53, p73 is exceptionally easy to inactivate as a transcription factor by point mutation. Taken together, these results indicate that p53 and p73 serve very different functions in tumors.


Subject(s)
DNA-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic , Meningeal Neoplasms/genetics , Meningioma/genetics , Neoplasm Proteins/genetics , Nuclear Proteins/genetics , Aged , Codon/genetics , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/biosynthesis , Cyclins/genetics , DNA Mutational Analysis , DNA, Neoplasm/genetics , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/physiology , Disease Progression , Female , Genes, Tumor Suppressor , Humans , Male , Meningeal Neoplasms/metabolism , Meningeal Neoplasms/pathology , Meningioma/metabolism , Meningioma/pathology , Middle Aged , Neoplasm Proteins/biosynthesis , Nuclear Proteins/biosynthesis , Nuclear Proteins/physiology , RNA Splicing , RNA, Messenger/biosynthesis , RNA, Neoplasm/biosynthesis , Recombinant Fusion Proteins/physiology , Reverse Transcriptase Polymerase Chain Reaction , Saccharomyces cerevisiae/genetics , Transcriptional Activation , Tumor Protein p73 , Tumor Suppressor Protein p53/physiology , Tumor Suppressor Proteins
2.
Oncogene ; 19(47): 5329-37, 2000 Nov 09.
Article in English | MEDLINE | ID: mdl-11103934

ABSTRACT

p53-germline mutations located in the core DNA-binding domain have been associated with a more dominant tumor penetrance especially for breast cancer and brain tumors. We previously reported an unusual accumulation of CNS tumors associated with a unique p53 germline mutation, Y236delta (deletion of codon 236). To test whether this tissue-specific tumor predisposition reflects a gain-of-function activity of Y236delta, we generated transgenic mice expressing Y236delta in astrocytes using the regulatory elements of the glial fibrillary acidic protein (GFAP) gene. After transplacental exposure to N-ethyl-N-nitrosourea (25 mg/kg BW) brain tumors developed in 18% (7/39) of GFAP-Y236delta transgenic p53-/- mice, while in p53+/- mice the incidence was 28% (11/40) (P>0.3). However, the mean tumor latency for GFAP-Y236delta/p53+/- mice was significantly shorter than for p53+/- mice, with 19.9 weeks vs 31.6 weeks (P=0.039), respectively. Taken together, cell specific expression of Y236delta results in an acceleration of tumor progression but does not confer a higher tumor penetrance. Conceivably, the transdominant effect of Y236delta provided a growth advantage early in the progression of neoplastic cells, since the endogenous p53 wild-type allele was lost in all brain tumors independent of the genotype. This reflects well observations from human astrocytic neoplasms with p53 mutations.


Subject(s)
Astrocytes/metabolism , Brain Neoplasms/pathology , Glioma/pathology , Tumor Suppressor Protein p53/physiology , Animals , Astrocytoma/classification , Astrocytoma/metabolism , Astrocytoma/pathology , Brain Neoplasms/classification , Brain Neoplasms/metabolism , Female , Gene Expression , Germ-Line Mutation , Glioblastoma/classification , Glioblastoma/metabolism , Glioblastoma/pathology , Glioma/classification , Glioma/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Transgenic , Microsatellite Repeats , Neoplasm Invasiveness , Telencephalon , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
3.
Lab Invest ; 80(11): 1681-9, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11092528

ABSTRACT

Endothelin-1 (ET-1) is a powerful mitogenic and/or anti-apoptotic peptide produced by many cancer cells. To evaluate the potential role of the endothelin system in glioblastoma we first determined the cellular distribution of the mRNA and proteins of the components of the endothelin system, preproendothelin-1 (PPET-1), endothelin-converting enzyme-1 (ECE-1), and ET(A) and ET(B) receptors in human glioblastoma tissue and glioblastoma cell lines. PPET-1, ECE-1, and ET(A) receptor were highly expressed in glioblastoma vessels and in some scattered glioblastoma areas whereas ET(B) receptor was mainly found in cancer cells. This suggests that glioblastoma vessels constitute an important source of ET-1 that acts on cancer cells via the ET(B) receptor. Four human glioblastoma cell lines expressed mRNA for all of the components of the ET-1 pathway. Bosentan, a mixed ET(A) and ET(B) receptor antagonist, induced apoptosis in these cell lines in a dose-dependent manner. Apoptosis was potentiated by Fas Ligand (APO-1L, CD95L), a pro-apoptotic peptide, only in LNZ308 cells, corresponding to the known functional Fas expression in these cell lines. LNZ308 cells also expressed the long and short forms of the cellular FLICE/caspase-8 inhibitory protein (FLIP). Bosentan and a protein kinase C inhibitor down-regulated short FLIP in these cells. ET-1 induced transient phosphorylation of extracellular signal-regulated kinase but did not induce long-term thymidine incorporation in LNZ308 glioblastoma cells. These results suggest that, in glioblastoma cells, ET-1, mainly acting via the ET(B) receptor, is a survival/antiapoptotic factor produced by tumor vasculature, but not a proliferation factor, involving protein kinase C and extracellular signal-regulated kinase pathways, and stabilization of the short form of FLIP.


Subject(s)
Brain Neoplasms/physiopathology , Endothelin-1/physiology , Glioblastoma/physiopathology , Base Sequence , DNA Primers , Endothelin-1/genetics , Fas Ligand Protein , Humans , Immunohistochemistry , Membrane Glycoproteins/physiology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , fas Receptor/physiology
4.
J Exp Med ; 191(10): 1789-98, 2000 May 15.
Article in English | MEDLINE | ID: mdl-10811871

ABSTRACT

Angiogenesis, the sprouting of new capillaries from preexisting blood vessels, results from a disruption of the balance between stimulatory and inhibitory factors. Here, we show that anoxia reduces expression of thrombospondin-1 (TSP-1), a natural inhibitor of angiogenesis, in glioblastoma cells. This suggests that reduced oxygen tension can promote angiogenesis not only by stimulating the production of inducers, such as vascular endothelial growth factor, but also by reducing the production of inhibitors. This downregulation may significantly contribute to glioblastoma development, since we show that an increase in TSP-1 expression is sufficient to strongly suppress glioblastoma cell tumorigenicity in vivo.


Subject(s)
Glioblastoma/genetics , Hypoxia/genetics , Thrombospondin 1/genetics , Animals , Base Sequence , DNA Primers/genetics , Down-Regulation , Genes, p53 , Glioblastoma/blood supply , Glioblastoma/metabolism , Humans , Hypoxia/physiopathology , Male , Mice , Mice, Nude , Neoplasm Transplantation , Neovascularization, Pathologic/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Thrombospondin 1/biosynthesis , Transplantation, Heterologous , Tumor Cells, Cultured
5.
Brain Pathol ; 9(3): 469-79, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10416987

ABSTRACT

In this study we established the simultaneous status of TP53, p16, p14ARF and PTEN tumor suppressor genes in 34 randomly chosen human glioma cell lines. Nine cell lines (26.4%) harbored mutations or deletions in all four tumor suppressor genes and 22 cell lines (64%) had alterations in at least three. Mutations/deletions were found at the following frequencies: TP53 (76.5%), p14ARF (64.7%), p16 (64.7%), PTEN (73.5%). Thus, there was a high incidence of alterations in the cellular pathways involving the p53 transcription factor (94.1%), the retinoblastoma protein (64.7%) and the PTEN phosphatase (73.5%) and 91% of cell lines carried mutations in two or more pathways. This provides the first clear genetic evidence that these tumor suppressors participate in biological pathways which are functioning separately/independently in glioma cells. The status of the gene alterations did not correlate with tumorigenicity in immunocompromized mice or any clinical parameters. Although the mutation rate was higher in glioma cell lines than that reported for glioma tissues, the alterations were molecularly representative of those found in adult de novo glioblastoma. This study highlights the importance of developing therapeutic approaches applicable to tumors with a broad range of genetic alterations and also provides an invaluable panel of glioma cell lines to make this possible.


Subject(s)
Brain Neoplasms/genetics , Cyclin-Dependent Kinase Inhibitor p16/genetics , Glioma/genetics , Phosphoric Monoester Hydrolases/genetics , Proteins/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Proteins , Adult , Aged , Animals , Brain Neoplasms/pathology , Child , DNA Mutational Analysis , Female , Genes, Tumor Suppressor/genetics , Glioma/pathology , Humans , Male , Mice , Mice, Nude , Middle Aged , Neoplasm Transplantation , Neoplastic Processes , PTEN Phosphohydrolase , Point Mutation , Sequence Deletion , Sex Factors , Tumor Cells, Cultured , Tumor Suppressor Protein p14ARF
6.
Oncogene ; 18(7): 1447-56, 1999 Feb 18.
Article in English | MEDLINE | ID: mdl-10050881

ABSTRACT

Oxygen deprivation is an important biological feature of tumor growth. We previously showed that in glioma, anoxia increases expression of IL-8, a chemokine and angiogenic factor. Here, we analysed for the first time the biochemical mechanisms inducing the IL-8 gene upon anoxia in glioma cells, and showed that they differ from those inducing the VEGF gene. Both genes are induced in biologically and genetically heterogenous glioblastoma cell lines (LN-229, LN-Z308, U87MG, T98G), whereas, in gliosarcoma cells (D247MG), only the VEGF gene is induced. The kinetics of IL-8 and VEGF mRNA inductions differ in these cells and reoxygenation experiments showed that the induction is due to the anoxic stress per se. Furthermore, in LN-229 and LN-Z308 cell lines actinomycin D, DRB and nuclear run-on experiments showed that anoxia stimulates increased transcription of both genes. Electromobility shift assays show increased protein binding to the AP-1 site on the IL-8 promoter following anoxia treatment. Finally, in situ hybridization on glioblastoma sections shows that the in vivo expression patterns of IL-8 and VEGF genes overlap, but are not identical. Since intratumoral augmentation of IL-8 and VEGF secretion, following microenvironmental decreases in oxygen pressure, may promote angiogenesis, further definition of these pathways is essential to appropriately target them for antitumoral therapy.


Subject(s)
Interleukin-8/genetics , Oxygen/physiology , Animals , Cell Hypoxia , Cobalt/pharmacology , Dactinomycin/pharmacology , Dichlororibofuranosylbenzimidazole/pharmacology , Endothelial Growth Factors/genetics , Gene Expression Regulation , Glioblastoma , Humans , Lymphokines/genetics , Mice , Nucleic Acid Synthesis Inhibitors/pharmacology , Oxidation-Reduction , RNA, Messenger , Response Elements , Transcription Factor AP-1/metabolism , Transcription, Genetic , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
7.
J Exp Med ; 186(8): 1201-12, 1997 Oct 20.
Article in English | MEDLINE | ID: mdl-9334359

ABSTRACT

Leukocyte infiltration and necrosis are two biological phenomena associated with the development of neovascularization during the malignant progression of human astrocytoma. Here, we demonstrate expression of interleukin (IL)-8, a cytokine with chemotactic and angiogenic properties, and of IL-8-binding receptors in astrocytoma. IL-8 expression is first observed in low grade astrocytoma in perivascular tumor areas expressing inflammatory cytokines. In glioblastoma, it further localizes to oxygen-deprived cells surrounding necrosis. Hypoxic/anoxic insults on glioblastoma cells in vitro using anaerobic chamber systems or within spheroids developing central necrosis induced an increase in IL-8 messenger RNA (mRNA) and protein expression. mRNA for IL-8-binding chemokine receptors CXCR1, CXCR2, and the Duffy antigen receptor for chemokines (DARC) were found in all astrocytoma grades by reverse transcription/PCR analysis. In situ hybridization and immunohistochemistry localized DARC expression on normal brain and tumor microvascular cells and CXCR1 and CXCR2 expression to infiltrating leukocytes. These results support a model where IL-8 expression is initiated early in astrocytoma development through induction by inflammatory stimuli and later in tumor progression increases due to reduced microenvironmental oxygen pressure. Augmented IL-8 would directly and/or indirectly promote angiogenesis by binding to DARC and by inducing leukocyte infiltration and activation by binding to CXCR1 and CXCR2.


Subject(s)
Chemotaxis, Leukocyte , Glioblastoma/metabolism , Glioblastoma/pathology , Interleukin-8/metabolism , Neovascularization, Physiologic/immunology , Up-Regulation , Anaerobiosis , Antigens, CD/biosynthesis , Blotting, Northern , Cell Hypoxia/immunology , Disease Progression , Glioblastoma/physiopathology , Humans , In Situ Hybridization , Interleukin-8/biosynthesis , Interleukin-8/genetics , Necrosis , RNA, Messenger/biosynthesis , Receptors, Interleukin/biosynthesis , Receptors, Interleukin-8A , Spheroids, Cellular
8.
Proc Natl Acad Sci U S A ; 92(4): 1008-12, 1995 Feb 14.
Article in English | MEDLINE | ID: mdl-7862624

ABSTRACT

Mutation of the p53 tumor suppressor gene is one of the earliest identified genetic lesions during malignant progression of human astrocytomas. To assess the functional significance of these mutations, wild-type (WT) p53 genes were introduced into glioblastoma cell lines having mutant, WT, or null endogenous p53 alleles. Populations of cells with mutant or null endogenous p53 alleles and exogenous WT p53 were spontaneously selected in culture for cells expressing only mutant p53 or no p53, which then displayed a growth and tumorigenic phenotype identical to the parental cells. To determine the phenotypic consequences of WT p53 expression before the occurrence of mutations, we developed a single cell assay to monitor WT p53-dependent transcription activity. Transfer and expression of exogenous WT p53 genes to cells with endogenous mutant or deleted, but not WT, p53 alleles caused growth arrest and morphological changes, including increased cell size and acquisition of multiple nuclei. This supports the hypothesis that genetic lesions of the p53 gene play an important role in the genesis of astrocytomas. Furthermore, the high sensitivity of the episomal single cell reporter strategy developed here has potential clinical applications in the rapid screening of patients for germ-line mutations of the p53 gene or any other gene with known targets for transcriptional transactivation.


Subject(s)
Alleles , Brain Neoplasms/genetics , Gene Transfer Techniques , Genes, p53 , Glioblastoma/genetics , Brain Neoplasms/pathology , Cell Division/genetics , Glioblastoma/pathology , Humans , Transcriptional Activation , Tumor Cells, Cultured
9.
Int J Cancer ; 58(2): 240-7, 1994 Jul 15.
Article in English | MEDLINE | ID: mdl-7517920

ABSTRACT

Expression of the monocyte chemoattractant protein-1 (MCP-1) was examined in human central nervous system tumours (glioblastomas and astrocytomas) and normal human brain. Northern blot analysis demonstrated constitutive expression of MCP-1 mRNA in 6 of 12 glioblastoma cell lines. Expression could be stimulated by interleukin (IL)-1 beta and tumour necrosis factor (TNF)-alpha in all cell lines tested. Immunoprecipitation demonstrated secretion of both isoforms, MCP-1 alpha and -beta, of the MCP-1 protein. Reverse-transcription polymerase chain reaction and Northern blot analysis on tissues demonstrated MCP-1 mRNA expression in 17 of 17 glioblastomas, 3 of 6 anaplastic astrocytomas and 6 of 6 low-grade astrocytomas, as well as in fetal brain but not in normal adult brain. In situ hybridization on 2 glioblastomas and 1 low-grade astrocytoma indicates that neoplastic astrocytes and endothelial cells express MCP-1 mRNA in vivo. Moreover, tumour cyst fluids of glioblastomas and astrocytomas were able to induce monocyte chemoattraction in an in vitro assay. This chemotactic activity was specifically neutralized by anti-MCP-1 antibodies in 9 of 10 samples, further demonstrating the production of bioactive MCP-1 in vivo and supporting an important role for this factor in the infiltration of monocytes/macrophages into tumour tissue.


Subject(s)
Astrocytoma/metabolism , Brain Neoplasms/metabolism , Chemotactic Factors/biosynthesis , Glioblastoma/metabolism , Astrocytoma/genetics , Base Sequence , Blotting, Northern , Brain Neoplasms/genetics , Chemokine CCL2 , Chemotactic Factors/genetics , Chemotactic Factors/metabolism , Cytokines/biosynthesis , Cytokines/genetics , Cytokines/metabolism , Gene Expression , Glioblastoma/genetics , Humans , Molecular Sequence Data , Polymerase Chain Reaction , RNA, Messenger/genetics , RNA-Directed DNA Polymerase , Tumor Cells, Cultured
10.
J Neurosurg ; 80(6): 1063-73, 1994 Jun.
Article in English | MEDLINE | ID: mdl-7514661

ABSTRACT

To elucidate which cytokine receptors may be expressed by human glioblastoma and normal astrocytic cells, the presence of messenger ribonucleic acid (RNA) for a number of cytokine receptors was examined in 16 glioblastoma cell lines and adult and fetal astrocytes. A complementary deoxyribonucleic acid copy of total RNA was synthesized and amplified with specific primers using the polymerase chain reaction method. The receptors studied were interleukin (IL)-1 receptor type I (IL-1RI) and type II (IL-1RII), p75 and p55 tumor necrosis factor (TNF) receptors (p75TNFR and p55TNFR), interferon (IFN)-alpha/beta and -gamma receptors (IFN-alpha/beta R and IFN-gamma R), granulocyte-macrophage (GM) colony-stimulating factors receptor alpha subunit (GM-CSFR), G-CSF receptor (G-CSFR), M-CSF receptor (c-fms, M-CSFR), stem cell factor receptor (c-kit, SCFR), IL-6 receptor (IL-6R), and IL-8 receptor (IL-8R). Transcripts for IL-1RI, p55TNFR, IFN-alpha/beta R, and IFN-gamma R were present in all cell lines. The presence of IL-1RII, p75TNFR, GM-CSFR, M-CSFR, SCFR, IL-6R, and IL-8R was identified in 13, eight, seven, eight, 14, three, and one cell lines, respectively. Normal astrocytes were positive for IL-1RI, p75TNFR, p55TNFR, IFN-alpha/beta R, IFN-gamma R, M-CSFR, and SCFR, showing a similarity to glioblastoma cells. Expression of IL-1RII was observed in adult astrocytes but not in fetal astrocytes. Furthermore, gene expression was assessed in normal brain tissue and 11 glioblastoma tissue specimens. The normal brain tissue expressed IL-1RI, IL-1RII, IFN-alpha/beta R, M-CSFR, and SCFR. Of the 11 glioblastoma tissue specimens, IL-1RI was positive in 11, IL-1RII in 10, p75TNFR in nine, p55TNFR in nine, IFN-alpha/beta R in 10, IFN-gamma R in 10, GM-CSFR in two, G-CSFR in three, IL-8R in eight, and M-CSFR and SCFR in 11. These expressions were consistent with those in the cell lines, except for IL-8R. It is concluded that glioblastoma cells and normal astrocytes express a similar set of cytokine receptor genes in vitro and in vivo. Possible autocrine loops are suggested for IL-1 alpha/IL-1RI, TNF-alpha/p55TNFR, IFN-beta/IFN-alpha/beta R, M-CSF/M-CSFR, and SCF/SCFR in glioblastomas.


Subject(s)
Astrocytes/chemistry , Brain Neoplasms/chemistry , Glioblastoma/chemistry , RNA, Messenger/analysis , Receptors, Cytokine/genetics , Adult , Base Sequence , Brain Neoplasms/genetics , Gene Expression , Glioblastoma/genetics , Humans , Molecular Sequence Data , Polymerase Chain Reaction , RNA-Directed DNA Polymerase , Transcription, Genetic
11.
J Neuroimmunol ; 50(2): 187-94, 1994 Mar.
Article in English | MEDLINE | ID: mdl-8120140

ABSTRACT

We investigated the expression and production of the interleukin-1 receptor antagonist (IL-1ra) in three human glioblastoma cell lines (LN443, LN444, LN859). Reverse transcription-polymerase chain reaction (RT-PCR) demonstrated the expression of IL-1ra mRNA transcripts in the three cell lines. These three cell lines also expressed mRNA for IL-1 alpha, IL-1 beta, as well as IL-1 receptor type I and type II, suggesting the presence of an IL-1 autocrine loop in these cell lines. Northern blot analysis demonstrated that the IL-1ra mRNA expression increased with IL-1 beta or tumor necrosis factor (TNF)-alpha but not with GM-CSF stimulation in both LN443 and LN444 cell lines. PMA stimulation increased the mRNA expression in LN444 but not in LN443 cells. Immunocytochemical staining showed IL-1ra immunoreactivity in these three cell lines. ELISA on culture supernatants demonstrated that the IL-1ra was secreted from the cell lines in agreement with the mRNA expression. RT-PCR with isoform-specific primers showed that both intracellular and secreted forms of IL-1ra were expressed by the three cell lines, with a predominance of the intracellular form. In vivo study with RT-PCR and Northern blot analysis demonstrated IL-1ra mRNA in six out of 12 human glioblastoma and two out of five anaplastic astrocytoma tissues, although the expression level was not high in some cases. Immunohistochemistry demonstrated the presence of IL-1ra within the cytoplasm of tumor cells in six out of 10 glioblastomas in vivo. These results suggest a potential role of IL-1ra in regulation of the IL-1 autocrine loop in glioblastomas.


Subject(s)
Glioblastoma/metabolism , Interleukin-1/metabolism , Receptors, Interleukin/antagonists & inhibitors , Base Sequence , Cytokines/physiology , Glioblastoma/pathology , Humans , Immunohistochemistry , Intracellular Membranes/metabolism , Molecular Sequence Data , Oligonucleotide Probes/genetics , Polymerase Chain Reaction , RNA, Messenger/metabolism , Tumor Cells, Cultured
12.
No Shinkei Geka ; 22(2): 125-9, 1994 Feb.
Article in Japanese | MEDLINE | ID: mdl-8115006

ABSTRACT

We analyzed the levels of p53 protein in 10 frozen astrocytoma specimens by using an ELISA kit. p53 protein was detected in seven of ten specimens, and ranged from 0.4 to 18.0ng/ml. To confirm that the p53 protein detected in this study actually represented mutations occurring within the p53 gene, we determined the nucleotide sequence in five of six positive cases, and in three of four negative cases. We amplified exons 5 to 9 by the PCR and sequence analysis was carried out by the dideoxy chain termination method. Point mutations resulting in amino acid substitutions were found in four of five positive cases but not in negative cases. Then we analyzed the presence of p53 protein in sera of 42 patients with malignant astrocytoma and 34 healthy donors, by the same method. p53 protein was detected in sera of 7 of 42 patients and 6 of 34 healthy donors, suggesting that this result may be due to non-specific responses. In conclusion, using the ELISA kit, overexpression of p53 protein, resulting from p53 mutations, in frozen astrocytoma specimens can be detected. This may be a good screening method for the detection of overexpression of p53 protein.


Subject(s)
Astrocytoma/metabolism , Brain Neoplasms/metabolism , Genes, p53 , Tumor Suppressor Protein p53/analysis , Astrocytoma/genetics , Brain Neoplasms/genetics , DNA, Neoplasm/analysis , Enzyme-Linked Immunosorbent Assay , Humans , Mutation , Polymerase Chain Reaction
13.
Cancer Res ; 54(3): 649-52, 1994 Feb 01.
Article in English | MEDLINE | ID: mdl-8306326

ABSTRACT

Chromosome 17p has been shown to be an early and frequent target for loss of heterozygosity through mitotic recombination in astrocytomas. These losses are frequently accompanied by point mutations in the p53 gene of the remaining allele, resulting in loss of wild type p53 function. However, a fraction of astrocytomas retain constitutional heterozygosity and do not have p53 mutations; some of these lose wild type p53 activity through binding to the protein product of amplified mdm2 genes. To test whether loss of wild type p53 biological function is a necessary step in astrocytoma progression we analyzed p53 expression and biological function in 13 glioma cell lines. All the cell lines expressed a 2.8-kilobase p53 transcript and showed various amounts of p53 protein by immunoprecipitation, except for cell line LN-Z308 which had only a small truncated p53 mRNA and no protein expression. To test whether the p53 expressed in these cell lines was functionally wild type or mutant we transfected them with a plasmid construct harboring a chloramphenicol acetyltransferase (CAT) reporter gene under the control of transcriptional elements that are induced by wild type but not mutant p53. Four lines were shown to retain wild type p53 function. Sequencing of the p53 gene in two of these cell lines confirmed the wild type genotype. These results show that inactivation of the p53 gene is not an obligatory step in glioblastoma genesis. This suggests either that two pathways (p53 inactivation dependent or independent) may lead to a tumor group classified histologically as glioblastoma or that in some cases p53 mutations are bypassed due to the presence of mutations in downstream effector genes.


Subject(s)
Brain Neoplasms/genetics , Gene Expression/genetics , Genes, p53/genetics , Glioblastoma/genetics , Animals , Base Sequence , Blotting, Northern , Brain Neoplasms/pathology , Female , Glioblastoma/pathology , Humans , Mice , Mice, Nude , Neoplasm Transplantation , Promoter Regions, Genetic/genetics , RNA, Neoplasm/genetics , Transcriptional Activation/genetics , Transfection , Tumor Cells, Cultured
14.
Cancer Res ; 53(22): 5345-9, 1993 Nov 15.
Article in English | MEDLINE | ID: mdl-7693337

ABSTRACT

Although human glioblastomas are highly invasive tumors intracerebrally, only rarely do they metastasize outside the central nervous system. In contrast, the brain is a major target for metastatic spread of many systemic tumors. Recently, it was demonstrated that expression of splice variants of CD44 (CD44v), but not standard CD44 (CD44s), was sufficient to confer metastatic potential to low- or nonmetastatic rat tumor cells. Because CD44 is expressed in brain tumors, we examined whether differential expression of CD44 isoforms was correlated with the metastatic behavior of these tumors. We compared CD44s and CD44v expression in 17 human glioblastomas, 18 glioma cell lines, and metastases of 15 other tumors to the brain by reverse transcription/polymerase chain reaction, Northern blotting, and immunocytochemistry. These experiments showed that 0 of 17 glioblastomas and 0 of 18 glioma cell lines expressed CD44v as compared to 12 of 15 brain metastases. These data show a correlation between CD44v expression and the metastatic ability of the tumors analyzed (P < 0.01). This suggests (a) that the biological significance of the lack of CD44v expression in human glioblastomas warrants further examination with regard to their inability to metastasize extraneurally and (b) that CD44v expression may play a role in the intracerebral spread of about 80% [corrected] of the brain metastases. Therefore, CD44v expression should be further considered as a potential marker for differential diagnosis and prognosis of patients with brain metastases.


Subject(s)
Biomarkers, Tumor/analysis , Brain Neoplasms/immunology , Brain Neoplasms/secondary , Glioblastoma/immunology , RNA, Neoplasm/analysis , Receptors, Lymphocyte Homing/analysis , Base Sequence , Blotting, Northern , Humans , Hyaluronan Receptors , Molecular Sequence Data , RNA, Neoplasm/chemistry , Tumor Cells, Cultured
15.
Cancer Res ; 50(20): 6683-8, 1990 Oct 15.
Article in English | MEDLINE | ID: mdl-2208133

ABSTRACT

This study demonstrates interleukin 6 (IL-6) production and release by human glioblastomas. Twenty glioblastoma cell lines were tested for IL-6 bioactivity using an IL-6-dependent cell line (7TD1). All of the lines tested with one exception (LN-229) constitutively released IL-6. A significant induction of IL-6 production and secretion was observed when LN-229 cells were treated with interleukin 1 beta (IL-1 beta) or tumor necrosis factor alpha. Various amounts of IL-6 mRNA were found in five of six cell lines tested. IL-6 mRNA was detected in line LN-229 only when the cells were treated with IL-1 beta or tumor necrosis factor alpha, confirming the bioassay data. Glioblastoma cells also produce IL-6 in vivo. (a) IL-6 activity was detected in 11 of 13 cerebrospinal fluids and five of five tumor cyst fluids. (b) IL-6 mRNA was found in four of four tumors. (c) Immunohistochemical analysis showed IL-6 within the tumor cells in 15 of 20 glioblastoma sections. In conclusion, biologically active IL-6 is released by almost all glioblastomas both in vitro and in vivo. The elevated levels of serum acute phase proteins and immune complexes found in glioblastoma patients may be the result of this secretion.


Subject(s)
Glioma/metabolism , Interleukin-6/metabolism , Glial Fibrillary Acidic Protein/analysis , Humans , Interleukin-1/pharmacology , Interleukin-6/analysis , Interleukin-6/genetics , RNA, Messenger/analysis , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/pharmacology
16.
J Neurooncol ; 9(2): 125-30, 1990 Oct.
Article in English | MEDLINE | ID: mdl-2175768

ABSTRACT

Glioblastoma cells constitutively produce various amounts of PGE2 (prostaglandin E2) in vitro. The amounts of PGE2 found in the conditioned medium of the glioblastoma cultures (less than 5 ng/ml) were not enough to inhibit the IL-2 (interleukin-2) activation of peripheral blood lymphocytes. However the amount of PGE2 produced by approximately 1 x 10(7) of the glioblastoma cells can be assumed to suppress the generation of IL-2-induced killing activity against glioblastoma cells.


Subject(s)
Brain Neoplasms/metabolism , Dinoprostone/biosynthesis , Glioblastoma/metabolism , Interleukin-2/antagonists & inhibitors , Killer Cells, Lymphokine-Activated/physiology , Culture Media , Dinoprostone/physiology , Humans , Radioimmunoassay , Tumor Cells, Cultured
17.
Int J Cancer ; 44(5): 948-51, 1989 Nov 15.
Article in English | MEDLINE | ID: mdl-2531122

ABSTRACT

Several established human glioma cell lines have been previously shown to express the common acute lymphoblastic leukemia antigen (cALLa, CD 10), an important marker in the diagnosis of human acute lymphocytic leukemia (ALL). The amino acid sequence of cALLa is identical to that of neutral endopeptidase (NEP,E.C.3.4.24.11), and cALLa expressed on leukemia and melanoma cell lines is enzymatically active NEP. In the present study, we investigated whether cALLa expressed on glioma cell lines is active NEP. We detected cALLa on 10 out of 13 glioma cell lines using 2 different anti-cALLa MAbs (A12-G4 and FAH99). NEP antigen, as detected by immunostaining with an anti-NEP MAb (135A3), was expressed on the same 10 lines. cALLa-positive, but not cALLa-negative cell lines displayed an endopeptidase activity. This activity could be blocked by phosphoramidon, a specific inhibitor of NEP. Furthermore, mRNAs hybridizing to an NEP-specific probe were present in cALLa-positive glioma cells but not in cALLa-negative cells. Taken together, the results provide strong evidence that cALLa-positive glioma cell lines express endopeptidase activity on the cell surface.


Subject(s)
Antigens, Differentiation/immunology , Antigens, Neoplasm/immunology , Glioma/enzymology , Glioma/immunology , Neprilysin/metabolism , Antigens, CD/analysis , Cell Membrane/enzymology , Humans , Immunologic Techniques , Neprilysin/genetics , Neprilysin/immunology , RNA, Messenger/genetics , RNA, Neoplasm/genetics , Tumor Cells, Cultured
18.
Int J Cancer ; 42(5): 780-6, 1988 Nov 15.
Article in English | MEDLINE | ID: mdl-3141300

ABSTRACT

Human malignant glioma cell lines and clones were incubated with various concentrations of recombinant human TNF-alpha, either alone or in combination with recombinant human IFN-gamma. The surface expression of HLA-ABC (class I) antigens and beta 2-microglobulin, was significantly enhanced by TNF-alpha alone on every cell line and clone tested. After incubation with both TNF-alpha and IFN-gamma, the surface expression of HLA-ABC antigens was only slightly higher than that observed with each cytokine alone. In contrast to IFN-gamma, TNF-alpha had no effect on the surface expression of HLA-DR (class II) antigens. Moreover, the surface expression of HLA-DR induced by IFN-gamma was unaffected by TNF-alpha. The increased expression of HLA-ABC antigens after treatment with TNF-alpha or IFN-gamma correlated with increased levels of HLA-ABC-specific mRNA. In addition, TNF-alpha, like IFN-gamma, selectively enhanced the surface expression of a tumor-associated antigen, Me14-D12, while it had no effect on the expression of various other surface antigens. In the absence of actinomycin D, TNF-alpha exhibited no direct cytotoxic/cytostatic effect on the glioma cell lines tested. These results indicate that TNF-alpha can enhance the surface expression of HLA-ABC antigens on human glioma cells in the absence of a direct cytotoxic/cytostatic effect.


Subject(s)
Glioma/pathology , Tumor Necrosis Factor-alpha/pharmacology , Animals , Cell Line , Cell Survival , Clone Cells , Dactinomycin/pharmacology , HLA-DR Antigens/analysis , Humans , Interferon-gamma/pharmacology , Mice , Phenotype , Rabbits , Recombinant Proteins/pharmacology
19.
J Natl Cancer Inst ; 76(2): 223-8, 1986 Feb.
Article in English | MEDLINE | ID: mdl-3080635

ABSTRACT

The modulation of HLA-DR and HLA-A, -B, and -C by human recombinant immune interferon (IFN-gamma) was studied on 10 malignant glioma cell lines established in our laboratory, on 8 clones or subclones derived from these lines, and on a fetal astrocyte cell line. Comparative studies were performed with recombinant leukocyte interferon (IFN-alpha). The results not only confirmed the selective activity of IFN-gamma on the modulation of HLA-DR expression, as opposed to that of IFN-alpha, but also demonstrated a marked heterogeneity in the response of glioma cell lines and their clones to the two types of IFN tested. For example, all 3 clones of an inducible cell line could be modulated to express HLA-DR, whereas only 2 of 5 clones derived from a noninducible line were modulated. This heterogeneity did not seem to be due to the absence of the receptor for IFN-gamma on the surface of these cells, since almost all of the cell lines or clones tested (17 of 19) responded to IFN-gamma by the induction or enhancement of the expression for either HLA-DR or HLA-A, -B, and -C (or both). The heterogeneity of induction was also demonstrated between clones derived from a glioma line that did not express HLA-DR after IFN-gamma treatment. The production of HLA-DR by one of the clones was abundant enough to be confirmed by immunoprecipitation and sodium dodecyl sulfate-polyacrylamide gel electrophoresis analysis.


Subject(s)
Glioma/immunology , Histocompatibility Antigens Class II/immunology , Interferon Type I/pharmacology , Interferon-gamma/pharmacology , Antibodies, Monoclonal , Astrocytes/drug effects , Astrocytes/immunology , Cell Line , Clone Cells , Electrophoresis, Polyacrylamide Gel , Fetus , Fluorescent Antibody Technique , Glial Fibrillary Acidic Protein/analysis , HLA-DR Antigens , Humans , Precipitin Tests , Radioimmunoassay
20.
Eur J Cancer Clin Oncol ; 21(2): 207-16, 1985 Feb.
Article in English | MEDLINE | ID: mdl-2985398

ABSTRACT

The reactivity spectrum of three monoclonal antibodies (Mabs) to human malignant glioma, five Mabs to melanomas and one Mab anti-HLA-DR was investigated by an indirect antibody binding radioimmunoassay on a panel of cells derived from 60 glioma lines, including 47 malignant astrocytomas, 11 low-grade astrocytomas and two malignant ependymomas as well on cells from 12 melanoma, three neuroblastoma, three medulloblastoma, two schwannoma, two retinoblastoma, two choroïd plexus papilloma, ten meningioma and 12 unrelated tumor lines. The anti-glioma Mabs BF7 and GE2 reacted preferentially with gliomas, while the anti-glioma Mab CG12 reacted with gliomas, melanomas, neuroblastomas and medulloblastomas. The five anti-melanoma Mabs reacted with gliomas, neuroblastomas and medulloblastomas. The anti-HLA-DR Mab D1-12 reacted with gliomas, melanomas and some meningiomas. On the basis of the data presented, we describe three different antigenic systems; the first one is glioma-associated, the second one is related to differentiation antigens expressed on cells derived from the neuroectoderm and the third is represented by HLA-DR antigens which are expressed not only on B-lymphoblastoid cells but also on melanomas and gliomas.


Subject(s)
Antigens, Neoplasm/analysis , Antigens, Surface/analysis , Glioma/immunology , Histocompatibility Antigens Class II/analysis , Melanoma/immunology , Neoplasms, Nerve Tissue/immunology , Antibodies, Monoclonal/immunology , Cell Line , Glial Fibrillary Acidic Protein , HLA-DR Antigens , Humans , Neoplasm Proteins/analysis , Nerve Tissue Proteins/analysis , Phenotype , S100 Proteins/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...