62 results on '"Hamou MF"'
Search Results
2. Activation and adhesion molecule expression on lymphoid infiltrates in human glioblastomas
- Author
-
Hamou Mf, de Tribolet N, and Kuppner Mc
- Subjects
Pathology ,medicine.medical_specialty ,CD58 ,CD3 ,Immunology ,Receptors, Lymphocyte Homing ,chemical and pharmacologic phenomena ,Biology ,Antigens, CD ,Cell Movement ,Glioma ,Histocompatibility Antigens ,medicine ,Cell Adhesion ,Immunology and Allergy ,Humans ,Lymphocytes ,Cell adhesion ,Lymphokine-activated killer cell ,Cell adhesion molecule ,Tumor-infiltrating lymphocytes ,Brain Neoplasms ,Integrin beta1 ,CD44 ,hemic and immune systems ,medicine.disease ,Antigens, Differentiation ,Immunohistochemistry ,Lymphocyte Function-Associated Antigen-1 ,Neurology ,Cancer research ,biology.protein ,Leukocyte Common Antigens ,Neurology (clinical) ,Cell Adhesion Molecules - Abstract
Frozen tissue sections obtained from human glioblastomas, brain tumor metastases and normal brain were examined for the expression of molecules known to be involved in lymphocyte activation and/or adhesion and migration. The molecules studied included CD3, CD45R, UCHL-1 (CD45RO), lymphocyte function-associated antigen 1 (LFA-1) (CD11a, CD18), intercellular adhesion molecule 1 (ICAM-1) (CD54), 4B4 (CD29), CD44, CD2, and LFA-3 (CD58). CD3+ lymphocytes infiltrating human glioblastomas and brain tumor metastases expressed LFA-1 alpha and beta. Many cells were also UCHL-1+ whereas only a small percentage were CD45R+. CD2+ lymphocytes were also present. Tumor-infiltrating lymphocytes (TIL) were found to be negative for CD29, which was, however, expressed on intratumoral vessels in addition to vessels found in normal brain. Glioblastoma cells and intratumoral vessels expressed ICAM-1 whereas no ICAM-1 was found on TIL or on normal brain. Glioblastoma cells also expressed high levels of both CD44 and LFA-3 whereas TIL were negative for these antigens. CD44 was also expressed on certain regions of normal brain. Antibodies to LFA-1 alpha and -beta and ICAM-1 could significantly block the binding of lymphokine-activated killer (LAK) cells or TIL to human glioblastoma cells suggesting that these molecules play a role in the binding and subsequent migration of lymphocytes into brain tumor tissue.
- Published
- 1990
3. Stem cell-related "self-renewal" signature and high epidermal growth factor receptor expression associated with resistance to concomitant chemoradiotherapy in glioblastoma.
- Author
-
Murat A, Migliavacca E, Gorlia T, Lambiv WL, Shay T, Hamou MF, de Tribolet N, Regli L, Wick W, Kouwenhoven MC, Hainfellner JA, Heppner FL, Dietrich PY, Zimmer Y, Cairncross JG, Janzer RC, Domany E, Delorenzi M, Stupp R, and Hegi ME
- Published
- 2008
- Full Text
- View/download PDF
4. Identification of the putative brain tumor antigen BF7/GE2 as the (De)toxifying enzyme microsomal epoxide hydrolase
- Author
-
Kessler, R., Hamou, Mf, Albertoni, M., Tribolet, N., Michael Arand, Meir, Eg, University of Zurich, and Van Meir, E G
- Subjects
Epoxide Hydrolases ,Antibodies, Neoplasm ,Brain Neoplasms ,Antibodies, Monoclonal ,10050 Institute of Pharmacology and Toxicology ,610 Medicine & health ,Glioma ,Transfection ,Rats ,Mice ,Antibody Specificity ,Antigens, Neoplasm ,Tumor Cells, Cultured ,Animals ,Humans ,570 Life sciences ,biology ,2730 Oncology ,1306 Cancer Research - Abstract
Malignant gliomas are the main cause of death from primary brain tumors. Despite surgery, radiation, and chemotherapy, patients have a median survival of less than a few years; therefore, it is clearly imperative to investigate new ways of treatment. The development of new therapeutic strategies for brain tumors is dependent on a better understanding of the differences between normal and tumoral brain cells. Our group had described previously a Mr 48,000 antigen defined by reactivity with two monoclonal antibodies (GE2 and BF7) obtained by immunization of mice with human glioblastoma cells. Here, we describe the identification of the GE2/BF7 antigen as microsomal epoxide hydrolase (mEH), a drug-metabolizing enzyme that is involved both in toxification and detoxification of carcinogens. We initially used immunoaffinity purification using GE2 and BF7 and analyzed the purified proteins by microsequencing. Edman degradation identified 15 amino acids of the NH2-terminal sequence that were 100% identical to mEH. To further confirm the identity of the BF7/GE2 antigen as mEH, we showed that the protein immunopurified with GE2 and BF7 was recognized by an anti-mEH antibody and that in vitro and in vivo synthesized human mEH is recognized by BF7 and GE2 antibodies. Furthermore, anti-mEH antibody recognizes an antigen expressed both in gliomas and reactive astrocytes, as do BF7 and GE2. Finally, we demonstrate that in contrast to what has been reported in rat embryo fibroblasts, p53 does not regulate mEH mRNA expression in glioma cells.
5. Metabolic and transcriptomic profiles of glioblastoma invasion revealed by comparisons between patients and corresponding orthotopic xenografts in mice.
- Author
-
Cudalbu C, Bady P, Lai M, Xin L, Gusyatiner O, Hamou MF, Lepore M, Brouland JP, Daniel RT, Hottinger AF, and Hegi ME
- Subjects
- Adult, Aged, Animals, Brain Neoplasms diagnostic imaging, Brain Neoplasms metabolism, Brain Neoplasms pathology, Female, Glioblastoma diagnostic imaging, Glioblastoma metabolism, Glioblastoma pathology, Humans, Magnetic Resonance Imaging, Male, Metabolome, Mice, Middle Aged, Neoplasm Invasiveness, Neoplasm Transplantation, Proton Magnetic Resonance Spectroscopy, Transcriptome, Brain Neoplasms genetics, Glioblastoma genetics
- Abstract
The invasive behavior of glioblastoma, the most aggressive primary brain tumor, is considered highly relevant for tumor recurrence. However, the invasion zone is difficult to visualize by Magnetic Resonance Imaging (MRI) and is protected by the blood brain barrier, posing a particular challenge for treatment. We report biological features of invasive growth accompanying tumor progression and invasion based on associated metabolic and transcriptomic changes observed in patient derived orthotopic xenografts (PDOX) in the mouse and the corresponding patients' tumors. The evolution of metabolic changes, followed in vivo longitudinally by
1 H Magnetic Resonance Spectroscopy (1 H MRS) at ultra-high field, reflected growth and the invasive properties of the human glioblastoma transplanted into the brains of mice (PDOX). Comparison of MRS derived metabolite signatures, reflecting temporal changes of tumor development and invasion in PDOX, revealed high similarity to spatial metabolite signatures of combined multi-voxel MRS analyses sampled across different areas of the patients' tumors. Pathway analyses of the transcriptome associated with the metabolite profiles of the PDOX, identified molecular signatures of invasion, comprising extracellular matrix degradation and reorganization, growth factor binding, and vascular remodeling. Specific analysis of expression signatures from the invaded mouse brain, revealed extent of invasion dependent induction of immune response, recapitulating respective signatures observed in glioblastoma. Integrating metabolic profiles and gene expression of highly invasive PDOX provided insights into progression and invasion associated mechanisms of extracellular matrix remodeling that is essential for cell-cell communication and regulation of cellular processes. Structural changes and biochemical properties of the extracellular matrix are of importance for the biological behavior of tumors and may be druggable. Ultra-high field MRS reveals to be suitable for in vivo monitoring of progression in the non-enhancing infiltration zone of glioblastoma., (© 2021. The Author(s).)- Published
- 2021
- Full Text
- View/download PDF
6. Hyperpolarized 13 C-glucose magnetic resonance highlights reduced aerobic glycolysis in vivo in infiltrative glioblastoma.
- Author
-
Mishkovsky M, Gusyatiner O, Lanz B, Cudalbu C, Vassallo I, Hamou MF, Bloch J, Comment A, Gruetter R, and Hegi ME
- Subjects
- Aerobiosis, Animals, Aspartic Acid analogs & derivatives, Aspartic Acid metabolism, Cell Line, Tumor, Humans, Lactic Acid metabolism, Metabolomics, Mice, SCID, Pyruvic Acid metabolism, Mice, Brain Neoplasms diagnostic imaging, Brain Neoplasms metabolism, Carbon Isotopes chemistry, Glioblastoma diagnostic imaging, Glioblastoma metabolism, Glucose metabolism, Glycolysis, Magnetic Resonance Imaging
- Abstract
Glioblastoma (GBM) is the most aggressive brain tumor type in adults. GBM is heterogeneous, with a compact core lesion surrounded by an invasive tumor front. This front is highly relevant for tumor recurrence but is generally non-detectable using standard imaging techniques. Recent studies demonstrated distinct metabolic profiles of the invasive phenotype in GBM. Magnetic resonance (MR) of hyperpolarized
13 C-labeled probes is a rapidly advancing field that provides real-time metabolic information. Here, we applied hyperpolarized13 C-glucose MR to mouse GBM models. Compared to controls, the amount of lactate produced from hyperpolarized glucose was higher in the compact GBM model, consistent with the accepted "Warburg effect". However, the opposite response was observed in models reflecting the invasive zone, with less lactate produced than in controls, implying a reduction in aerobic glycolysis. These striking differences could be used to map the metabolic heterogeneity in GBM and to visualize the infiltrative front of GBM.- Published
- 2021
- Full Text
- View/download PDF
7. In vivo characterization of brain metabolism by 1 H MRS, 13 C MRS and 18 FDG PET reveals significant glucose oxidation of invasively growing glioma cells.
- Author
-
Lai M, Vassallo I, Lanz B, Poitry-Yamate C, Hamou MF, Cudalbu C, Gruetter R, and Hegi ME
- Subjects
- Adaptor Proteins, Signal Transducing metabolism, Animals, Aspartic Acid analogs & derivatives, Aspartic Acid metabolism, Brain diagnostic imaging, Brain Neoplasms genetics, Brain Neoplasms metabolism, Cell Line, Tumor, Glioma genetics, Glioma metabolism, Glutamic Acid metabolism, Humans, Male, Mice, Neoplasm Transplantation, Oxidation-Reduction, Positron-Emission Tomography methods, Proton Magnetic Resonance Spectroscopy methods, Repressor Proteins metabolism, gamma-Aminobutyric Acid metabolism, Adaptor Proteins, Signal Transducing genetics, Brain metabolism, Brain Neoplasms diagnostic imaging, Fluorodeoxyglucose F18 metabolism, Glioma diagnostic imaging, Glucose metabolism, Repressor Proteins genetics
- Abstract
Glioblastoma are notorious for their highly invasive growth, diffusely infiltrating adjacent brain structures that precludes complete resection, and is a major obstacle for cure. To characterize this "invisible" tumor part, we designed a high resolution multimodal imaging approach assessing in vivo the metabolism of invasively growing glioma xenografts in the mouse brain. Animals were subjected longitudinally to magnetic resonance imaging (MRI) and
1 H spectroscopy (MRS) at ultra high field (14.1 Tesla) that allowed the measurement of 16 metabolic biomarkers to characterize the metabolic profiles. As expected, the neuronal functionality was progressively compromised as indicated by decreasing N-acetyl aspartate, glutamate and gamma-aminobutyric acid and reduced neuronal TCA cycle (-58%) and neurotransmission (-50%). The dynamic metabolic changes observed, captured differences in invasive growth that was modulated by re-expression of the tumor suppressor gene WNT inhibitory factor 1 (WIF1) in the orthotopic xenografts that attenuates invasion. At late stage mice were subjected to13 C MRS with infusion of [1,6-13 C]glucose and18 FDG positron emission tomography (PET) to quantify cell-specific metabolic fluxes involved in glucose metabolism. Most interestingly, this provided the first in vivo evidence for significant glucose oxidation in glioma cells. This suggests that the infiltrative front of glioma does not undergo the glycolytic switch per se, but that environmental triggers may induce metabolic reprograming of tumor cells., (© 2018 UICC.)- Published
- 2018
- Full Text
- View/download PDF
8. Early detection of human glioma sphere xenografts in mouse brain using diffusion MRI at 14.1 T.
- Author
-
Porcari P, Hegi ME, Lei H, Hamou MF, Vassallo I, Capuani S, Gruetter R, and Mlynarik V
- Subjects
- Algorithms, Animals, Cell Line, Tumor, Female, Humans, Image Enhancement methods, Image Interpretation, Computer-Assisted methods, Mice, Mice, Nude, Neoplasm Invasiveness, Reproducibility of Results, Sensitivity and Specificity, Spheroids, Cellular pathology, Brain Neoplasms diagnostic imaging, Brain Neoplasms pathology, Diffusion Magnetic Resonance Imaging methods, Early Detection of Cancer methods, Glioma diagnostic imaging, Glioma pathology
- Abstract
Glioma models have provided important insights into human brain cancers. Among the investigative tools, MRI has allowed their characterization and diagnosis. In this study, we investigated whether diffusion MRI might be a useful technique for early detection and characterization of slow-growing and diffuse infiltrative gliomas, such as the proposed new models, LN-2669GS and LN-2540GS glioma sphere xenografts. Tumours grown in these models are not visible in conventional T
2 -weighted or contrast-enhanced T1 -weighted MRI at 14.1 T. Diffusion-weighted imaging and diffusion tensor imaging protocols were optimized for contrast by exploring long diffusion times sensitive for probing the microstructural alterations induced in the normal brain by the slow infiltration of glioma sphere cells. Compared with T2 -weighted images, tumours were properly identified in their early stage of growth using diffusion MRI, and confirmed by localized proton MR spectroscopy as well as immunohistochemistry. The first evidence of tumour presence was revealed for both glioma sphere xenograft models three months after tumour implantation, while no necrosis, oedema or haemorrhage were detected either by MRI or by histology. Moreover, different values of diffusion indices, such as mean diffusivity and fractional anisotropy, were obtained in tumours grown from LN-2669GS and LN-2540GS glioma sphere lines. These observations highlighted diverse tumour microstructures for both xenograft models, which were reflected in histology. This study demonstrates the ability of diffusion MRI techniques to identify and investigate early stages of slow-growing, invasive tumours in the mouse brain, thus providing a potential imaging biomarker for early detection of tumours in humans., (Copyright © 2016 John Wiley & Sons, Ltd.)- Published
- 2016
- Full Text
- View/download PDF
9. Phase II Study of Radiotherapy and Temsirolimus versus Radiochemotherapy with Temozolomide in Patients with Newly Diagnosed Glioblastoma without MGMT Promoter Hypermethylation (EORTC 26082).
- Author
-
Wick W, Gorlia T, Bady P, Platten M, van den Bent MJ, Taphoorn MJ, Steuve J, Brandes AA, Hamou MF, Wick A, Kosch M, Weller M, Stupp R, Roth P, Golfinopoulos V, Frenel JS, Campone M, Ricard D, Marosi C, Villa S, Weyerbrock A, Hopkins K, Homicsko K, Lhermitte B, Pesce G, and Hegi ME
- Subjects
- Adult, Aged, Brain Neoplasms mortality, Brain Neoplasms radiotherapy, DNA Methylation, DNA Modification Methylases genetics, DNA Repair Enzymes genetics, Dacarbazine administration & dosage, Dacarbazine analogs & derivatives, Female, Glioblastoma mortality, Glioblastoma radiotherapy, Humans, Kaplan-Meier Estimate, Male, Middle Aged, Promoter Regions, Genetic genetics, Proportional Hazards Models, Sirolimus administration & dosage, Temozolomide, Tumor Suppressor Proteins genetics, Young Adult, Brain Neoplasms drug therapy, Chemoradiotherapy methods, Glioblastoma drug therapy, Sirolimus analogs & derivatives
- Abstract
Purpose: EORTC 26082 assessed the activity of temsirolimus in patients with newly diagnosed glioblastoma harboring an unmethylated O6 methylguanine-DNA-methyltransferase (MGMT) promoter., Experimental Design: Patients (n = 257) fulfilling eligibility criteria underwent central MGMT testing. Patients with MGMT unmethylated glioblastoma (n = 111) were randomized 1:1 between standard chemo-radiotherapy with temozolomide or radiotherapy plus weekly temsirolimus (25 mg). Primary endpoint was overall survival at 12 months (OS12). A positive signal was considered >38 patients alive at 12 months in the per protocol population. A noncomparative reference arm of 54 patients evaluated the assumptions on OS12 in a standard-treated cohort of patients. Prespecified post hoc analyses of markers reflecting target activation were performed., Results: Both therapies were administered per protocol with a median of 13 cycles of maintenance temsirolimus. Median age was 55 and 58 years in the temsirolimus and standard arms, the WHO performance status 0 or 1 for most patients (95.5%). In the per protocol population, 38 of 54 patients treated with temsirolimus reached OS12. The actuarial 1-year survival was 72.2% [95% confidence interval (CI), 58.2-82.2] in the temozolomide arm and 69.6% (95% CI, 55.8-79.9) in the temsirolimus arm [hazard ratio (HR) 1.16; 95% CI, 0.77-1.76; P = 0.47]. In multivariable prognostic analyses of clinical and molecular factors, phosphorylation of mTORSer2448 in tumor tissue (HR 0.13; 95% CI, 0.04-0.47; P = 0.002), detected in 37.6%, was associated with benefit from temsirolimus., Conclusions: Temsirolimus was not superior to temozolomide in patients with an unmethylated MGMT promoter. Phosphorylation of mTORSer2448 in the pretreatment tumor tissue may define a subgroup benefitting from mTOR inhibition. Clin Cancer Res; 22(19); 4797-806. ©2016 AACR., (©2016 American Association for Cancer Research.)
- Published
- 2016
- Full Text
- View/download PDF
10. WIF1 re-expression in glioblastoma inhibits migration through attenuation of non-canonical WNT signaling by downregulating the lncRNA MALAT1.
- Author
-
Vassallo I, Zinn P, Lai M, Rajakannu P, Hamou MF, and Hegi ME
- Subjects
- Adaptor Proteins, Signal Transducing genetics, Animals, Brain Neoplasms genetics, Brain Neoplasms pathology, Cell Line, Tumor, Cell Proliferation physiology, Down-Regulation, Gene Expression Regulation, Neoplastic, Glioblastoma genetics, Glioblastoma pathology, Heterografts, Humans, Mice, Mice, Nude, Proto-Oncogene Proteins genetics, RNA, Long Noncoding genetics, Repressor Proteins genetics, Signal Transduction, Wnt Proteins genetics, Wnt Signaling Pathway, Wnt-5a Protein, Adaptor Proteins, Signal Transducing biosynthesis, Brain Neoplasms metabolism, Cell Movement physiology, Glioblastoma metabolism, Proto-Oncogene Proteins metabolism, RNA, Long Noncoding metabolism, Repressor Proteins biosynthesis, Wnt Proteins metabolism
- Abstract
Glioblastoma is the most aggressive primary brain tumor in adults and due to the invasive nature cannot be completely removed. The WNT inhibitory factor 1 (WIF1), a secreted inhibitor of WNTs, is systematically downregulated in glioblastoma and acts as strong tumor suppressor. The aim of this study was the dissection of WIF1-associated tumor-suppressing effects mediated by canonical and non-canonical WNT signaling. We found that WIF1 besides inhibiting the canonical WNT pathway selectively downregulates the WNT/calcium pathway associated with significant reduction of p38-MAPK (p38-mitogen-activated protein kinase) phosphorylation. Knockdown of WNT5A, the only WNT ligand overexpressed in glioblastoma, phenocopied this inhibitory effect. WIF1 expression inhibited cell migration in vitro and in an orthotopic brain tumor model, in accordance with the known regulatory function of the WNT/Ca(2+) pathway on migration and invasion. In search of a mediator for this function differential gene expression profiles of WIF1-expressing cells were performed. Metastasis-associated lung adenocarcinoma transcript 1 (MALAT1), a long non-coding RNA and key positive regulator of invasion, emerged as the top downregulated gene. Indeed, knockdown of MALAT1 reduced migration in glioblastoma cells, without effect on proliferation. Hence, loss of WIF1 enhances the migratory potential of glioblastoma through WNT5A that activates the WNT/Ca(2+) pathway and MALAT1. These data suggest the involvement of canonical and non-canonical WNT pathways in glioblastoma promoting key features associated with this deadly disease, proliferation on one hand and invasion on the other. Successful targeting will require a dual strategy affecting both canonical and non-canonical WNT pathways.
- Published
- 2016
- Full Text
- View/download PDF
11. Presence of an oligodendroglioma-like component in newly diagnosed glioblastoma identifies a pathogenetically heterogeneous subgroup and lacks prognostic value: central pathology review of the EORTC_26981/NCIC_CE.3 trial.
- Author
-
Hegi ME, Janzer RC, Lambiv WL, Gorlia T, Kouwenhoven MC, Hartmann C, von Deimling A, Martinet D, Besuchet Schmutz N, Diserens AC, Hamou MF, Bady P, Weller M, van den Bent MJ, Mason WP, Mirimanoff RO, Stupp R, Mokhtari K, and Wesseling P
- Subjects
- Adolescent, Adult, Aged, Brain Neoplasms genetics, Brain Neoplasms therapy, Chemoradiotherapy, Clinical Trials, Phase III as Topic, DNA Methylation, Dacarbazine analogs & derivatives, Dacarbazine therapeutic use, ErbB Receptors genetics, ErbB Receptors metabolism, Female, Glioblastoma genetics, Glioblastoma therapy, Humans, Male, Middle Aged, Mutation, Oligodendroglioma genetics, Oligodendroglioma therapy, Prognosis, Survival Analysis, Temozolomide, Treatment Outcome, Young Adult, Brain Neoplasms pathology, Glioblastoma pathology, Oligodendroglioma pathology
- Abstract
Glioblastoma (GBM) is a morphologically heterogeneous tumor type with a median survival of only 15 months in clinical trial populations. However, survival varies greatly among patients. As part of a central pathology review, we addressed the question if patients with GBM displaying distinct morphologic features respond differently to combined chemo-radiotherapy with temozolomide. Morphologic features were systematically recorded for 360 cases with particular focus on the presence of an oligodendroglioma-like component and respective correlations with outcome and relevant molecular markers. GBM with an oligodendroglioma-like component (GBM-O) represented 15% of all confirmed GBM (52/339) and was not associated with a more favorable outcome. GBM-O encompassed a pathogenetically heterogeneous group, significantly enriched for IDH1 mutations (19 vs. 3%, p = 0.003) and EGFR amplifications (71 vs. 48%, p = 0.04) compared with other GBM, while co-deletion of 1p/19q was found in only one case and the MGMT methylation frequency was alike (47 vs. 46%). Expression profiles classified most of the GBM-O into two subtypes, 36% (5/14 evaluable) as proneural and 43% as classical GBM. The detection of pseudo-palisading necrosis (PPN) was associated with benefit from chemotherapy (p = 0.0002), while no such effect was present in the absence of PPN (p = 0.86). In the adjusted interaction model including clinical prognostic factors and MGMT status, PPN was borderline nonsignificant (p = 0.063). Taken together, recognition of an oligodendroglioma-like component in an otherwise classic GBM identifies a pathogenetically mixed group without prognostic significance. However, the presence of PPN may indicate biological features of clinical relevance for further improvement of therapy.
- Published
- 2012
- Full Text
- View/download PDF
12. DNA fingerprinting of glioma cell lines and considerations on similarity measurements.
- Author
-
Bady P, Diserens AC, Castella V, Kalt S, Heinimann K, Hamou MF, Delorenzi M, and Hegi ME
- Subjects
- DNA, Neoplasm genetics, Humans, Polymerase Chain Reaction, Tumor Cells, Cultured, Biomarkers, Tumor genetics, Brain Neoplasms genetics, DNA Fingerprinting, Glioma genetics, Microsatellite Repeats genetics
- Abstract
Glioma cell lines are an important tool for research in basic and translational neuro-oncology. Documentation of their genetic identity has become a requirement for scientific journals and grant applications to exclude cross-contamination and misidentification that lead to misinterpretation of results. Here, we report the standard 16 marker short tandem repeat (STR) DNA fingerprints for a panel of 39 widely used glioma cell lines as reference. Comparison of the fingerprints among themselves and with the large DSMZ database comprising 9 marker STRs for 2278 cell lines uncovered 3 misidentified cell lines and confirmed previously known cross-contaminations. Furthermore, 2 glioma cell lines exhibited identity scores of 0.8, which is proposed as the cutoff for detecting cross-contamination. Additional characteristics, comprising lack of a B-raf mutation in one line and a similarity score of 1 with the original tumor tissue in the other, excluded a cross-contamination. Subsequent simulation procedures suggested that, when using DNA fingerprints comprising only 9 STR markers, the commonly used similarity score of 0.8 is not sufficiently stringent to unambiguously differentiate the origin. DNA fingerprints are confounded by frequent genetic alterations in cancer cell lines, particularly loss of heterozygosity, that reduce the informativeness of STR markers and, thereby, the overall power for distinction. The similarity score depends on the number of markers measured; thus, more markers or additional cell line characteristics, such as information on specific mutations, may be necessary to clarify the origin.
- Published
- 2012
- Full Text
- View/download PDF
13. The Wnt inhibitory factor 1 (WIF1) is targeted in glioblastoma and has a tumor suppressing function potentially by induction of senescence.
- Author
-
Lambiv WL, Vassallo I, Delorenzi M, Shay T, Diserens AC, Misra A, Feuerstein B, Murat A, Migliavacca E, Hamou MF, Sciuscio D, Burger R, Domany E, Stupp R, and Hegi ME
- Subjects
- Adaptor Proteins, Signal Transducing antagonists & inhibitors, Adaptor Proteins, Signal Transducing genetics, Animals, Blotting, Western, Brain Neoplasms genetics, Brain Neoplasms metabolism, Cell Adhesion, Cell Line, Tumor, Cell Proliferation, DNA Methylation, Down-Regulation, Epigenesis, Genetic, Female, Glioblastoma genetics, Glioblastoma metabolism, Humans, Immunoenzyme Techniques, Mice, Mice, Nude, Promoter Regions, Genetic, RNA, Messenger genetics, RNA, Small Interfering genetics, Repressor Proteins antagonists & inhibitors, Repressor Proteins genetics, Reverse Transcriptase Polymerase Chain Reaction, Adaptor Proteins, Signal Transducing metabolism, Aging, Brain Neoplasms prevention & control, Gene Expression Regulation, Neoplastic, Genes, Tumor Suppressor, Glioblastoma prevention & control, Repressor Proteins metabolism
- Abstract
Gene expression-based prediction of genomic copy number aberrations in the chromosomal region 12q13 to 12q15 that is flanked by MDM2 and CDK4 identified Wnt inhibitory factor 1 (WIF1) as a candidate tumor suppressor gene in glioblastoma. WIF1 encodes a secreted Wnt antagonist and was strongly downregulated in most glioblastomas as compared with normal brain, implying deregulation of Wnt signaling, which is associated with cancer. WIF1 silencing was mediated by deletion (7/69, 10%) or epigenetic silencing by promoter hypermethylation (29/110, 26%). Co-amplification of MDM2 and CDK4 that is present in 10% of glioblastomas was associated in most cases with deletion of the whole genomic region enclosed, including the WIF1 locus. This interesting pathogenetic constellation targets the RB and p53 tumor suppressor pathways in tandem, while simultaneously activating oncogenic Wnt signaling. Ectopic expression of WIF1 in glioblastoma cell lines revealed a dose-dependent decrease of Wnt pathway activity. Furthermore, WIF1 expression inhibited cell proliferation in vitro, reduced anchorage-independent growth in soft agar, and completely abolished tumorigenicity in vivo. Interestingly, WIF1 overexpression in glioblastoma cells induced a senescence-like phenotype that was dose dependent. These results provide evidence that WIF1 has tumor suppressing properties. Downregulation of WIF1 in 75% of glioblastomas indicates frequent involvement of aberrant Wnt signaling and, hence, may render glioblastomas sensitive to inhibitors of Wnt signaling, potentially by diverting the tumor cells into a senescence-like state.
- Published
- 2011
- Full Text
- View/download PDF
14. Pathway analysis of glioblastoma tissue after preoperative treatment with the EGFR tyrosine kinase inhibitor gefitinib--a phase II trial.
- Author
-
Hegi ME, Diserens AC, Bady P, Kamoshima Y, Kouwenhoven MC, Delorenzi M, Lambiv WL, Hamou MF, Matter MS, Koch A, Heppner FL, Yonekawa Y, Merlo A, Frei K, Mariani L, and Hofer S
- Subjects
- Adult, Aged, Cell Line, Tumor, ErbB Receptors genetics, ErbB Receptors metabolism, Female, Gefitinib, Glioblastoma enzymology, Humans, Male, Middle Aged, Phosphorylation drug effects, Proto-Oncogene Proteins c-akt metabolism, Signal Transduction drug effects, Young Adult, Antineoplastic Agents therapeutic use, ErbB Receptors antagonists & inhibitors, Glioblastoma drug therapy, Glioblastoma metabolism, Protein Kinase Inhibitors therapeutic use, Quinazolines therapeutic use
- Abstract
Amplification of the epidermal growth factor receptor (EGFR) gene is one of the most common oncogenic alterations in glioblastoma (45%) making it a prime target for therapy. However, small molecule inhibitors of the EGFR tyrosine kinase showed disappointing efficacy in clinical trials for glioblastoma. Here we aimed at investigating the molecular effects of the tyrosine kinase inhibitor gefitinib on the EGFR signaling pathway in human glioblastoma. Twenty-two patients selected for reoperation of recurrent glioblastoma were treated within a phase II trial for 5 days with 500 mg gefitinib before surgery followed by postoperative gefitinib until recurrence. Resected glioblastoma tissues exhibited high concentrations of gefitinib (median, 4.1 μg/g), 20 times higher than respective plasma. EGFR-pathway activity was evaluated with phosphorylation-specific assays. The EGFR was efficiently dephosphorylated in treated patients as compared to a control cohort of 12 patients. However, no significant effect on 12 pathway constituents was detected. In contrast, in vitro treatment of a glioblastoma cell line, BS-153, with endogenous EGFRwt amplification and EGFRvIII expression resulted not only in dephosphorylation of the EGFR, but also of key regulators in the pathway such as AKT. Treating established xenografts of the same cell line as an in vivo model showed dephosphorylation of the EGFR without affecting downstream signal transductors, similar to the human glioblastoma. Taken together, gefitinib reaches high concentrations in the tumor tissue and efficiently dephosphorylates its target. However, regulation of downstream signal transducers in the EGFR pathway seems to be dominated by regulatory circuits independent of EGFR phosphorylation.
- Published
- 2011
- Full Text
- View/download PDF
15. Extent and patterns of MGMT promoter methylation in glioblastoma- and respective glioblastoma-derived spheres.
- Author
-
Sciuscio D, Diserens AC, van Dommelen K, Martinet D, Jones G, Janzer RC, Pollo C, Hamou MF, Kaina B, Stupp R, Levivier M, and Hegi ME
- Subjects
- Adult, Aged, Aged, 80 and over, Chromatin ultrastructure, Female, Gene Frequency, Humans, Male, Middle Aged, Promoter Regions, Genetic, Tumor Cells, Cultured, Brain Neoplasms genetics, DNA Methylation, Glioblastoma genetics, O(6)-Methylguanine-DNA Methyltransferase genetics
- Abstract
Purpose: Quantitative methylation-specific tests suggest that not all cells in a glioblastoma with detectable promoter methylation of the O6-methylguanine DNA methyltransferase (MGMT) gene carry a methylated MGMT allele. This observation may indicate cell subpopulations with distinct MGMT status, raising the question of the clinically relevant cutoff of MGMT methylation therapy. Epigenetic silencing of the MGMT gene by promoter methylation blunts repair of O6-methyl guanine and has been shown to be a predictive factor for benefit from alkylating agent therapy in glioblastoma., Experimental Design: Ten paired samples of glioblastoma and respective glioblastoma-derived spheres (GS), cultured under stem cell conditions, were analyzed for the degree and pattern of MGMT promoter methylation by methylation-specific clone sequencing, MGMT gene dosage, chromatin status, and respective effects on MGMT expression and MGMT activity., Results: In glioblastoma, MGMT-methylated alleles ranged from 10% to 90%. In contrast, methylated alleles were highly enriched (100% of clones) in respective GS, even when 2 MGMT alleles were present, with 1 exception (<50%). The CpG methylation patterns were characteristic for each glioblastoma exhibiting 25% to 90% methylated CpGs of 28 sites interrogated. Furthermore, MGMT promoter methylation was associated with a nonpermissive chromatin status in accordance with very low MGMT transcript levels and undetectable MGMT activity., Conclusions: In MGMT-methylated glioblastoma, MGMT promoter methylation is highly enriched in GS that supposedly comprise glioma-initiating cells. Thus, even a low percentage of MGMT methylation measured in a glioblastoma sample may be relevant and predict benefit from an alkylating agent therapy., (©2010 AACR.)
- Published
- 2011
- Full Text
- View/download PDF
16. Marker-independent identification of glioma-initiating cells.
- Author
-
Clément V, Marino D, Cudalbu C, Hamou MF, Mlynarik V, de Tribolet N, Dietrich PY, Gruetter R, Hegi ME, and Radovanovic I
- Subjects
- AC133 Antigen, Animals, Antigens, CD analysis, Cell Differentiation, Cells, Cultured, Fluorescence, Gene Expression Profiling, Glycoproteins analysis, Humans, Mice, Peptides analysis, Biomarkers, Tumor analysis, Brain Neoplasms pathology, Glioma pathology, Neoplastic Stem Cells pathology
- Abstract
Tumor-initiating cells with stem cell properties are believed to sustain the growth of gliomas, but proposed markers such as CD133 cannot be used to identify these cells with sufficient specificity. We report an alternative isolation method purely based on phenotypic qualities of glioma-initiating cells (GICs), avoiding the use of molecular markers. We exploited intrinsic autofluorescence properties and a distinctive morphology to isolate a subpopulation of cells (FL1(+)) from human glioma or glioma cultures. FL1(+) cells are capable of self-renewal in vitro, tumorigenesis in vivo and preferentially express stem cell genes. The FL1(+) phenotype did not correlate with the expression of proposed GIC markers. Our data propose an alternative approach to investigate tumor-initiating potential in gliomas and to advance the development of new therapies and diagnostics.
- Published
- 2010
- Full Text
- View/download PDF
17. Elevated levels of MIC-1/GDF15 in the cerebrospinal fluid of patients are associated with glioblastoma and worse outcome.
- Author
-
Shnaper S, Desbaillets I, Brown DA, Murat A, Migliavacca E, Schluep M, Ostermann S, Hamou MF, Stupp R, Breit SN, de Tribolet N, and Hegi ME
- Subjects
- Adolescent, Adult, Aged, Aged, 80 and over, Biomarkers, Tumor blood, Brain Neoplasms blood, Brain Neoplasms surgery, Case-Control Studies, Cohort Studies, Enzyme-Linked Immunosorbent Assay, Female, Glioblastoma blood, Glioblastoma surgery, Growth Differentiation Factor 15 blood, Growth Differentiation Factor 15 genetics, Humans, Immunoenzyme Techniques, Male, Middle Aged, Prognosis, RNA, Messenger genetics, RNA, Messenger metabolism, Reverse Transcriptase Polymerase Chain Reaction, Survival Rate, Treatment Outcome, Young Adult, Biomarkers, Tumor cerebrospinal fluid, Brain Neoplasms cerebrospinal fluid, Glioblastoma cerebrospinal fluid, Growth Differentiation Factor 15 cerebrospinal fluid
- Abstract
For patients with brain tumors identification of diagnostic and prognostic markers in easy accessible biological material, such as plasma or cerebrospinal fluid (CSF), would greatly facilitate patient management. MIC-1/GDF15 (growth differentiation factor 15) is a secreted protein of the TGF-beta superfamily and emerged as a candidate marker exhibiting increasing mRNA expression during malignant progression of glioma. Determination of MIC-1/GDF15 protein levels by ELISA in the CSF of a cohort of 94 patients with intracranial tumors including gliomas, meningioma and metastasis revealed significantly increased concentrations in glioblastoma patients (median, 229 pg/ml) when compared with control cohort of patients treated for non-neoplastic diseases (median below limit of detection of 156 pg/ml, p < 0.0001, Mann-Whitney test). However, plasma MIC-1/GDF15 levels were not elevated in the matching plasma samples from these patients. Most interestingly, patients with glioblastoma and increased CSF MIC-1/GDF15 had a shorter survival (p = 0.007, log-rank test). In conclusion, MIC-1/GDF15 protein measured in the CSF may have diagnostic and prognostic value in patients with intracranial tumors.
- Published
- 2009
- Full Text
- View/download PDF
18. Modulation of angiogenic and inflammatory response in glioblastoma by hypoxia.
- Author
-
Murat A, Migliavacca E, Hussain SF, Heimberger AB, Desbaillets I, Hamou MF, Rüegg C, Stupp R, Delorenzi M, and Hegi ME
- Subjects
- Brain Neoplasms metabolism, Cell Line, Tumor, Cohort Studies, Computational Biology methods, Gene Expression Profiling, Humans, Inflammation, Monocytes metabolism, Multigene Family, Oligonucleotide Array Sequence Analysis, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Gene Expression Regulation, Neoplastic, Glioblastoma metabolism, Hypoxia, Neovascularization, Pathologic
- Abstract
Glioblastoma are rapidly proliferating brain tumors in which hypoxia is readily recognizable, as indicated by focal or extensive necrosis and vascular proliferation, two independent diagnostic criteria for glioblastoma. Gene expression profiling of glioblastoma revealed a gene expression signature associated with hypoxia-regulated genes. The correlated gene set emerging from unsupervised analysis comprised known hypoxia-inducible genes involved in angiogenesis and inflammation such as VEGF and BIRC3, respectively. The relationship between hypoxia-modulated angiogenic genes and inflammatory genes was associated with outcome in our cohort of glioblastoma patients treated within prospective clinical trials of combined chemoradiotherapy. The hypoxia regulation of several new genes comprised in this cluster including ZNF395, TNFAIP3, and TREM1 was experimentally confirmed in glioma cell lines and primary monocytes exposed to hypoxia in vitro. Interestingly, the cluster seems to characterize differential response of tumor cells, stromal cells and the macrophage/microglia compartment to hypoxic conditions. Most genes classically associated with the inflammatory compartment are part of the NF-kappaB signaling pathway including TNFAIP3 and BIRC3 that have been shown to be involved in resistance to chemotherapy.Our results associate hypoxia-driven tumor response with inflammation in glioblastoma, hence underlining the importance of tumor-host interaction involving the inflammatory compartment.
- Published
- 2009
- Full Text
- View/download PDF
19. Tenascin-C is a novel RBPJkappa-induced target gene for Notch signaling in gliomas.
- Author
-
Sivasankaran B, Degen M, Ghaffari A, Hegi ME, Hamou MF, Ionescu MC, Zweifel C, Tolnay M, Wasner M, Mergenthaler S, Miserez AR, Kiss R, Lino MM, Merlo A, Chiquet-Ehrismann R, and Boulay JL
- Subjects
- Amino Acid Sequence, Base Sequence, Brain Neoplasms metabolism, Brain Neoplasms pathology, Cell Movement physiology, Glioblastoma metabolism, Glioblastoma pathology, Humans, Immunoglobulin J Recombination Signal Sequence-Binding Protein genetics, Immunohistochemistry, Molecular Sequence Data, Oligodendroglioma genetics, Oligodendroglioma metabolism, Oligodendroglioma pathology, Promoter Regions, Genetic, Receptor, Notch2 genetics, Receptor, Notch2 metabolism, Response Elements, Signal Transduction, Tenascin biosynthesis, Transcriptional Activation, Brain Neoplasms genetics, Glioblastoma genetics, Immunoglobulin J Recombination Signal Sequence-Binding Protein biosynthesis, Receptor, Notch2 biosynthesis, Tenascin genetics
- Abstract
Tenascin-C (TNC) expression is known to correlate with malignancy in glioblastoma (GBM), a highly invasive and aggressive brain tumor that shows limited response to conventional therapies. In these malignant gliomas as well as in GBM cell lines, we found Notch2 protein to be strongly expressed. In a GBM tumor tissue microarray, RBPJk protein, a Notch2 cofactor for transcription, was found to be significantly coexpressed with TNC. We show that the TNC gene is transactivated by Notch2 in an RBPJk-dependent manner mediated by an RBPJk binding element in the TNC promoter. The transactivation is abrogated by a Notch2 mutation, which we detected in the glioma cell line Hs683 that does not express TNC. This L1711M mutation resides in the RAM domain, the site of interaction between Notch2 and RBPJk. In addition, transfection of constructs encoding activated Notch2 or Notch1 increased endogenous TNC expression identifying TNC as a novel Notch target gene. Overexpression of a dominant negative form of the transcriptional coactivator MAML1 or knocking down RBPJk in LN319 cells led to a dramatic decrease in TNC protein levels accompanied by a significant reduction of cell migration. Because addition of purified TNC stimulated glioma cell migration, this represents a mechanism for the invasive properties of glioma cells controlled by Notch signaling and defines a novel oncogenic pathway in gliomagenesis that may be targeted for therapeutic intervention in GBM patients.
- Published
- 2009
- Full Text
- View/download PDF
20. Anti-O6-methylguanine-methyltransferase (MGMT) immunohistochemistry in glioblastoma multiforme: observer variability and lack of association with patient survival impede its use as clinical biomarker.
- Author
-
Preusser M, Charles Janzer R, Felsberg J, Reifenberger G, Hamou MF, Diserens AC, Stupp R, Gorlia T, Marosi C, Heinzl H, Hainfellner JA, and Hegi M
- Subjects
- Adult, Aged, Antibodies, Antibody Specificity, Biomarkers, Tumor analysis, Brain Neoplasms physiopathology, Cohort Studies, DNA Methylation, DNA Modification Methylases analysis, DNA Modification Methylases genetics, DNA Repair Enzymes analysis, DNA Repair Enzymes genetics, Diagnosis, Differential, Glioblastoma physiopathology, Humans, Immunohistochemistry methods, Middle Aged, Observer Variation, Predictive Value of Tests, Promoter Regions, Genetic genetics, Prospective Studies, Reproducibility of Results, Survival Rate, Tumor Suppressor Proteins analysis, Tumor Suppressor Proteins genetics, Biomarkers, Tumor metabolism, Brain Neoplasms diagnosis, Brain Neoplasms enzymology, DNA Modification Methylases metabolism, DNA Repair Enzymes metabolism, Glioblastoma diagnosis, Glioblastoma enzymology, Tumor Suppressor Proteins metabolism
- Abstract
Silencing of O6-methylguanine-DNA methyltransferase (MGMT) protein expression because of MGMT gene promoter hypermethylation is considered to be associated with postoperative chemoradiotherapy benefits in glioblastoma multiforme (GBM) patients. The objective of this study was to clarify the usability of MGMT immunohistochemistry (IHC) as a clinical biomarker. We immunostained a tissue microarray containing biopsy samples of 164 GBM patients from the European Organization for Research and Treatment of Cancer and the National Cancer Institute of Canada (EORTC/NCIC) trial 26981/22981 using two commercial anti-MGMT antibodies (clones MT3.1 and MT23.2). Immunostaining results were semiquantitatively evaluated by four observers from three neuropathological laboratories using a predefined algorithm. We analyzed (i) inter- and intraobserver agreement on MGMT expression (kappa statistics); (ii) correlation of MGMT expression with MGMT promoter methylation status (kappa statistics); and (iii) correlation of MGMT expression with patient outcome (log-rank test). Interobserver agreement on MGMT expression varied from slight to almost perfect, whereas intraobserver agreement ranged from substantial to almost perfect. MGMT expression showed poor to moderate correlation with MGMT promoter methylation status. We found no significant association of MGMT expression with patient outcome. In our hands, observer variability as well as lack of association with the MGMT promoter methylation status and patient survival impeded the use of anti-MGMT immunohistochemistry as a clinical biomarker for routine diagnostic purposes.
- Published
- 2008
- Full Text
- View/download PDF
21. Infrequent promoter methylation of the MGMT gene in liver metastases from uveal melanoma.
- Author
-
Voelter V, Diserens AC, Moulin A, Nagel G, Yan P, Migliavacca E, Rimoldi D, Hamou MF, Kaina B, Leyvraz S, and Hegi ME
- Subjects
- Antineoplastic Agents therapeutic use, DNA Methylation, Humans, Immunohistochemistry, Liver Neoplasms drug therapy, Melanoma drug therapy, Predictive Value of Tests, Promoter Regions, Genetic, Protein Array Analysis, Biomarkers, Tumor genetics, DNA Modification Methylases genetics, DNA Repair Enzymes genetics, Liver Neoplasms genetics, Liver Neoplasms secondary, Melanoma genetics, Melanoma secondary, Tumor Suppressor Proteins genetics, Uveal Neoplasms pathology
- Abstract
Uveal melanoma is associated with a high mortality rate once metastases occur, with over >90% of metastatic patients dying within less than 1 year from metastases to the liver. The intraarterial hepatic (iah) administration of the alkylating agent fotemustine holds some promise with response rates of 36% and median survival of 15 months. Here, we investigated whether the DNA-repair-protein MGMT may be involved in the variability of response to fotemustine and temozolomide in uveal melanoma. Epigenetic inactivation of MGMT has been demonstrated to be a predictive marker for benefit from alkylating agent therapy in glioblastoma. We found a methylated MGMT promoter in 6% of liver metastases from 34 uveal melanoma patients. The mean MGMT activity measured in liver metastases with negligible liver tissue content was significantly lower than in liver tissue (146 versus 523 fmol/mg protein, p = 0.002). Expression of the MGMT protein was detectable in 50% of 88 metastases by immunohistochemistry on a tissue microarray. Expression was heterogeneous, and in accordance with MGMT activity data, usually lower than in the surrounding liver. Differential MGMT activity/expression between metastasis and liver tissue and more efficient depletion of MGMT with higher doses of alkylating agent therapy using iah delivery may provide the pharmacologic window for the higher response rate. However, these results do not support MGMT methylation status or protein expression as predictive markers for treatment outcome to iah chemotherapy with alkylating agents.
- Published
- 2008
- Full Text
- View/download PDF
22. Validation of real-time methylation-specific PCR to determine O6-methylguanine-DNA methyltransferase gene promoter methylation in glioma.
- Author
-
Vlassenbroeck I, Califice S, Diserens AC, Migliavacca E, Straub J, Di Stefano I, Moreau F, Hamou MF, Renard I, Delorenzi M, Flamion B, DiGuiseppi J, Bierau K, and Hegi ME
- Subjects
- Glioma genetics, Humans, Reproducibility of Results, Sensitivity and Specificity, DNA Methylation, Glioma diagnosis, O(6)-Methylguanine-DNA Methyltransferase genetics, Polymerase Chain Reaction methods, Promoter Regions, Genetic genetics
- Abstract
Epigenetic silencing of the DNA repair protein O(6)-methylguanine-DNA methyltransferase (MGMT) by promoter methylation predicts successful alkylating agent therapy, such as with temozolomide, in glioblastoma patients. Stratified therapy assignment of patients in prospective clinical trials according to tumor MGMT status requires a standardized diagnostic test, suitable for high-throughput analysis of small amounts of formalin-fixed, paraffin-embedded tumor tissue. A direct, real-time methylation-specific PCR (MSP) assay was developed to determine methylation status of the MGMT gene promoter. Assay specificity was obtained by selective amplification of methylated DNA sequences of sodium bisulfite-modified DNA. The copy number of the methylated MGMT promoter, normalized to the beta-actin gene, provides a quantitative test result. We analyzed 134 clinical glioma samples, comparing the new test with the previously validated nested gel-based MSP assay, which yields a binary readout. A cut-off value for the MGMT methylation status was suggested by fitting a bimodal normal mixture model to the real-time results, supporting the hypothesis that there are two distinct populations within the test samples. Comparison of the tests showed high concordance of the results (82/91 [90%]; Cohen's kappa = 0.80; 95% confidence interval, 0.82-0.95). The direct, real-time MSP assay was highly reproducible (Pearson correlation 0.996) and showed valid test results for 93% (125/134) of samples compared with 75% (94/125) for the nested, gel-based MSP assay. This high-throughput test provides an important pharmacogenomic tool for individualized management of alkylating agent chemotherapy.
- Published
- 2008
- Full Text
- View/download PDF
23. MGMT gene silencing and benefit from temozolomide in glioblastoma.
- Author
-
Hegi ME, Diserens AC, Gorlia T, Hamou MF, de Tribolet N, Weller M, Kros JM, Hainfellner JA, Mason W, Mariani L, Bromberg JE, Hau P, Mirimanoff RO, Cairncross JG, Janzer RC, and Stupp R
- Subjects
- Brain Neoplasms drug therapy, Brain Neoplasms mortality, Brain Neoplasms radiotherapy, Chemotherapy, Adjuvant, Disease-Free Survival, Glioblastoma drug therapy, Glioblastoma mortality, Glioblastoma radiotherapy, Humans, Polymerase Chain Reaction, Promoter Regions, Genetic, Randomized Controlled Trials as Topic, Survival Analysis, Temozolomide, Antineoplastic Agents, Alkylating therapeutic use, Brain Neoplasms genetics, DNA Methylation, Dacarbazine analogs & derivatives, Dacarbazine therapeutic use, Gene Silencing, Glioblastoma genetics, O(6)-Methylguanine-DNA Methyltransferase genetics
- Abstract
Background: Epigenetic silencing of the MGMT (O6-methylguanine-DNA methyltransferase) DNA-repair gene by promoter methylation compromises DNA repair and has been associated with longer survival in patients with glioblastoma who receive alkylating agents., Methods: We tested the relationship between MGMT silencing in the tumor and the survival of patients who were enrolled in a randomized trial comparing radiotherapy alone with radiotherapy combined with concomitant and adjuvant treatment with temozolomide. The methylation status of the MGMT promoter was determined by methylation-specific polymerase-chain-reaction analysis., Results: The MGMT promoter was methylated in 45 percent of 206 assessable cases. Irrespective of treatment, MGMT promoter methylation was an independent favorable prognostic factor (P<0.001 by the log-rank test; hazard ratio, 0.45; 95 percent confidence interval, 0.32 to 0.61). Among patients whose tumor contained a methylated MGMT promoter, a survival benefit was observed in patients treated with temozolomide and radiotherapy; their median survival was 21.7 months (95 percent confidence interval, 17.4 to 30.4), as compared with 15.3 months (95 percent confidence interval, 13.0 to 20.9) among those who were assigned to only radiotherapy (P=0.007 by the log-rank test). In the absence of methylation of the MGMT promoter, there was a smaller and statistically insignificant difference in survival between the treatment groups., Conclusions: Patients with glioblastoma containing a methylated MGMT promoter benefited from temozolomide, whereas those who did not have a methylated MGMT promoter did not have such a benefit., (Copyright 2005 Massachusetts Medical Society.)
- Published
- 2005
- Full Text
- View/download PDF
24. Growth promoting signaling by tenascin-C [corrected].
- Author
-
Ruiz C, Huang W, Hegi ME, Lange K, Hamou MF, Fluri E, Oakeley EJ, Chiquet-Ehrismann R, and Orend G
- Subjects
- Actins metabolism, Cell Growth Processes drug effects, Cell Line, Tumor, Cytoskeletal Proteins biosynthesis, Cytoskeletal Proteins genetics, DNA-Binding Proteins biosynthesis, DNA-Binding Proteins genetics, Down-Regulation drug effects, Gene Expression Profiling, Glioblastoma metabolism, Humans, Inhibitor of Differentiation Protein 2, Proto-Oncogene Proteins physiology, RNA, Messenger biosynthesis, RNA, Messenger genetics, Repressor Proteins biosynthesis, Repressor Proteins genetics, Signal Transduction drug effects, Transcription Factors biosynthesis, Transcription Factors genetics, Tropomyosin biosynthesis, Wnt Proteins, Gene Expression Regulation, Neoplastic drug effects, Glioblastoma genetics, Glioblastoma pathology, MAP Kinase Signaling System drug effects, Tenascin pharmacology
- Abstract
Tenascin-C is an adhesion-modulating extracellular matrix molecule that is highly expressed in tumor stroma and stimulates tumor cell proliferation. Adhesion of T98G glioblastoma cells to a fibronectin substratum is inhibited by tenascin-C. To address the mechanism of action, we performed a RNA expression analysis of T89G cells grown in the presence or absence of tenascin-C and found that tenascin-C down-regulates tropomyosin-1. Upon overexpression of tropomyosin-1, cell spreading on a fibronectin/tenascin-C substratum was restored, indicating that tenascin-C destabilizes actin stress fibers through down-regulation of tropomyosin-1. Tenascin-C also increased the expression of the endothelin receptor type A and stimulated the corresponding mitogen-activated protein kinase signaling pathway, which triggers extracellular signal-regulated kinase 1/2 phosphorylation and c-Fos expression. Tenascin-C additionally caused down-regulation of the Wnt inhibitor Dickkopf 1. In consequence, Wnt signaling was enhanced through stabilization of beta-catenin and stimulated the expression of the beta-catenin target Id2. Finally, our in vivo data derived from astrocytoma tissue arrays link increased tenascin-C and Id2 expression with high malignancy. Because increased endothelin and Wnt signaling, as well as reduced tropomyosin-1 expression, are closely linked to transformation and tumorigenesis, we suggest that tenascin-C specifically modulates these signaling pathways to enhance proliferation of glioma cells.
- Published
- 2004
- Full Text
- View/download PDF
25. INK4a/Arf is required for suppression of EGFR/DeltaEGFR(2-7)-dependent ERK activation in mouse astrocytes and glioma.
- Author
-
Lachat Y, Diserens AC, Nozaki M, Kobayashi H, Hamou MF, Godard S, De Tribolet N, and Hegi ME
- Subjects
- Animals, Enzyme Activation, Mice, Phosphorylation, Quinazolines, Tyrphostins pharmacology, Astrocytes enzymology, Cyclin-Dependent Kinase Inhibitor p16 physiology, ErbB Receptors antagonists & inhibitors, Glioma enzymology, Mitogen-Activated Protein Kinases metabolism
- Abstract
Amplification of the epidermal growth factor receptor (EGFR) or expression of its constitutively activated mutant, DeltaEGFR(2-7), in association with the inactivation of the INK4a/Arf gene locus is a frequent alteration in human glioblastoma. The notion of a cooperative effect between these two alterations has been demonstrated in respective mouse brain tumor models including our own. Here, we investigated underlying molecular mechanisms in early passage cortical astrocytes deficient for p16(INK4a)/p19(Arf) or p53, respectively, with or without ectopic expression of DeltaEGFR(2-7). Targeting these cells with the specific EGFR inhibitor tyrphostin AG1478 revealed that phosphorylation of ERK was only abrogated in the presence of an intact INK4a/Arf gene locus. The sensitivity to inhibit ERK phosphorylation was independent of ectopic expression of DeltaEGFR(2-7) and independent of the TP53 status. This resistance to downregulate the MAPK pathway in the absence of INK4a/Arf was confirmed in cell lines derived from our mouse glioma models with the respective initial genetic alterations. Thus, deletion of INK4a/Arf appears to keep ERK in its active, phosphorylated state insensitive to an upstream inhibitor specifically targeting EGFR/DeltaEGFR(2-7). This resistance may contribute to the cooperative tumorigenic effect selected for in human glioblastoma that may be of crucial clinical relevance for treatments specifically targeting EGFR/DeltaEGFR(2-7) in glioblastoma patients.
- Published
- 2004
- Full Text
- View/download PDF
26. Classification of human astrocytic gliomas on the basis of gene expression: a correlated group of genes with angiogenic activity emerges as a strong predictor of subtypes.
- Author
-
Godard S, Getz G, Delorenzi M, Farmer P, Kobayashi H, Desbaillets I, Nozaki M, Diserens AC, Hamou MF, Dietrich PY, Regli L, Janzer RC, Bucher P, Stupp R, de Tribolet N, Domany E, and Hegi ME
- Subjects
- Adolescent, Adult, Aged, Astrocytoma blood supply, Astrocytoma metabolism, Astrocytoma pathology, Brain Neoplasms blood supply, Brain Neoplasms metabolism, Brain Neoplasms pathology, Cell Hypoxia physiology, Child, Preschool, Female, Gene Expression Profiling, Gene Expression Regulation, Neoplastic, Glioblastoma genetics, Glioblastoma metabolism, Glioblastoma pathology, Humans, Insulin-Like Growth Factor Binding Protein 2 biosynthesis, Insulin-Like Growth Factor Binding Protein 2 genetics, Male, Middle Aged, Multigene Family, Oligonucleotide Array Sequence Analysis, Reproducibility of Results, Astrocytoma genetics, Brain Neoplasms genetics, Neovascularization, Pathologic genetics
- Abstract
The development of targeted treatment strategies adapted to individual patients requires identification of the different tumor classes according to their biology and prognosis. We focus here on the molecular aspects underlying these differences, in terms of sets of genes that control pathogenesis of the different subtypes of astrocytic glioma. By performing cDNA-array analysis of 53 patient biopsies, comprising low-grade astrocytoma, secondary glioblastoma (respective recurrent high-grade tumors), and newly diagnosed primary glioblastoma, we demonstrate that human gliomas can be differentiated according to their gene expression. We found that low-grade astrocytoma have the most specific and similar expression profiles, whereas primary glioblastoma exhibit much larger variation between tumors. Secondary glioblastoma display features of both other groups. We identified several sets of genes with relatively highly correlated expression within groups that: (a). can be associated with specific biological functions; and (b). effectively differentiate tumor class. One prominent gene cluster discriminating primary versus nonprimary glioblastoma comprises mostly genes involved in angiogenesis, including VEGF fms-related tyrosine kinase 1 but also IGFBP2, that has not yet been directly linked to angiogenesis. In situ hybridization demonstrating coexpression of IGFBP2 and VEGF in pseudopalisading cells surrounding tumor necrosis provided further evidence for a possible involvement of IGFBP2 in angiogenesis. The separating groups of genes were found by the unsupervised coupled two-way clustering method, and their classification power was validated by a supervised construction of a nearly perfect glioma classifier.
- Published
- 2003
27. Aquaporin 1 and aquaporin 4 expression in human brain after subarachnoid hemorrhage and in peritumoral tissue.
- Author
-
Badaut J, Brunet JF, Grollimund L, Hamou MF, Magistretti PJ, Villemure JG, and Regli L
- Subjects
- Aquaporin 1, Aquaporin 4, Blood Group Antigens, Blotting, Western, Case-Control Studies, Humans, Immunohistochemistry, Aquaporins metabolism, Brain metabolism, Brain Neoplasms metabolism, Glioma metabolism, Subarachnoid Hemorrhage metabolism
- Abstract
Aquaporins (AQPs) are a protein family of water channels which facilitate the water flux through the plasmatic membranes. The expression of AQPs has been described in rat brain by several studies. Despite recent reports that have shown an over-expression of AQP1 and 4 in human tumoral cells, little is known about AQP expression in human brain. The purpose of this study was to investigate the expression of AQP1 and AQP4 in human brain after subarachnoid hemorrhage (SAH) and in peritumoral tissue by western blot and immunohistochemistry. The results showed a marked increase of the expression of AQP1 and AQP4. This over-expression occurred on the astrocytic processes and polarization on astrocytic end-feet was lost. No expression was observed on neuronal cells. This study is the first demonstration of the induction of AQP1 and AQP4 on reactive astrocytes in an acute brain injury, such as SAH. These results reinforce the hypothesis that AQPs may be involved in the dynamics of brain edema formation or resolution. Further studies are needed to understand their functional role.
- Published
- 2003
- Full Text
- View/download PDF
28. Anoxia induces macrophage inhibitory cytokine-1 (MIC-1) in glioblastoma cells independently of p53 and HIF-1.
- Author
-
Albertoni M, Shaw PH, Nozaki M, Godard S, Tenan M, Hamou MF, Fairlie DW, Breit SN, Paralkar VM, de Tribolet N, Van Meir EG, and Hegi ME
- Subjects
- Animals, Brain Neoplasms genetics, Brain Neoplasms metabolism, Cell Hypoxia genetics, Cytokines genetics, Dexamethasone pharmacology, Doxycycline pharmacology, Genes, p53, Glioblastoma genetics, Glioblastoma metabolism, Growth Differentiation Factor 15, Humans, Hypoxia-Inducible Factor 1, Hypoxia-Inducible Factor 1, alpha Subunit, Mice, Mice, Nude, Neoplasm Proteins genetics, Neoplasm Proteins physiology, Neoplasm Transplantation, Recombinant Fusion Proteins physiology, Signal Transduction, Transplantation, Heterologous, Brain Neoplasms pathology, Cytokines biosynthesis, DNA-Binding Proteins physiology, Gene Expression Regulation, Neoplastic drug effects, Glioblastoma pathology, Neoplasm Proteins biosynthesis, Nuclear Proteins physiology, Oxygen pharmacology, Transcription Factors, Tumor Suppressor Protein p53 physiology
- Abstract
Human astrocytic brain tumors select for mutations in the p53 tumor suppressor gene early in malignant progression. p53 is activated upon various kinds of cellular stress leading to apoptosis or cell cycle arrest, but is also implicated in complex biological processes such as inhibition of angiogenesis and metastasis. In an effort to shed light on consequences mediated by p53 inactivation in gliomas, we established the Tet-On system for p53 in the LN-Z308 glioblastoma cell line. The macrophage inhibitory cytokine-1 (MIC-1) gene was identified as a most prominent p53 target gene upon gene expression profiling. Oxygen deprivation, an important cellular stress, revealed MIC-1 as an anoxia responsive gene in glioblastoma cell lines. MIC-1 up-regulation by anoxia is mediated through an alternative, p53 and hypoxia inducible factor 1 (HIF-1) independent pathway. Furthermore, ectopic expression of MIC-1 in LN-Z308 cell line completely abolished its inherent tumorigenicity in nude mice, while proliferation in vitro was not affected. In the present experimental model MIC-1 may exert its anti-tumorigenic properties via a paracrine mechanism mediated by host cells in vivo. Taken together, these data suggest that MIC-1 is an important downstream mediator of p53 function, while acting itself as an intercessor of cellular stress signaling and exerting anti-tumorigenic activities.
- Published
- 2002
- Full Text
- View/download PDF
29. p73 is not mutated in meningiomas as determined with a functional yeast assay but p73 expression increases with tumor grade.
- Author
-
Nozaki M, Tada M, Kashiwazaki H, Hamou MF, Diserens AC, Shinohe Y, Sawamura Y, Iwasaki Y, de Tribolet N, and Hegi ME
- Subjects
- 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, DNA-Binding Proteins genetics, Gene Expression Regulation, Neoplastic, Meningeal Neoplasms genetics, Meningioma genetics, Neoplasm Proteins genetics, Nuclear Proteins genetics
- 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.
- Published
- 2001
- Full Text
- View/download PDF
30. p53 transdominance but no gain of function in mouse brain tumor model.
- Author
-
Hegi ME, Klein MA, Rüedi D, Chène P, Hamou MF, and Aguzzi A
- Subjects
- Alleles, Animals, Brain metabolism, Brain pathology, Brain Neoplasms metabolism, Brain Neoplasms pathology, Cells, Cultured, DNA Mutational Analysis, Female, Genotype, Humans, Mice, Mice, Inbred C57BL, Mice, Transgenic, Phenotype, Plasmids, Precipitin Tests, Tumor Suppressor Protein p53 metabolism, Brain Neoplasms genetics, Genes, Dominant, Genes, p53 genetics, Mutation
- Abstract
Although p53 mutations in tumors typically result in loss of transactivation of p53 target genes some mutants display gain-of-function activity. The latter has important implications for the design of rational cancer therapy. We previously described a germ-line p53 mutation (deletion of codon 236, Y236delta) associated with a familial brain tumor syndrome. To determine whether this tissue-specific tumor predisposition reflects a gain-of-function activity of Y236delta or an effect of genetic background we have developed a mouse brain tumor model. Primary neuroectodermal cells deficient for p53 (+/- or -/-) and transduced with Y236delta using a retroviral vector were transplanted into the brain of adult wild-type mice. This neurografting paradigm circumvents the problem of early lethal tumors at extracerebral sites associated with germ-line p53 deficiency. Brain tumors arising in this mouse model were highly invasive, reflecting an important feature of the human disease. Tumors arose from p53+/- cells only when transduced with Y236delta. In keeping with in vitro data showing that Y236delta has dominant-negative activity, these tumors retained the endogenous wild-type p53 allele but accumulated high levels of Y236delta. However, the presence of Y236delta in transplanted p53-/- cells had no effect on the tumor frequency, 15% versus 27% without the mutant. In conclusion, Y236delta is transdominant but exerts no gain-of-function activity mediating a more penetrant tumor phenotype.
- Published
- 2000
31. Regulation of aminopeptidase A in human brain tumor vasculature: evidence for a role of transforming growth factor-beta.
- Author
-
Juillerat-Jeanneret L, Lohm S, Hamou MF, and Pinet F
- Subjects
- Animals, Brain Neoplasms pathology, Cell Division, Cerebrovascular Circulation, Endothelium, Vascular drug effects, Endothelium, Vascular pathology, Glioblastoma pathology, Glutamyl Aminopeptidase, Humans, Neovascularization, Pathologic, Rats, Rats, Inbred F344, Transforming Growth Factor beta pharmacology, Aminopeptidases metabolism, Angiotensins physiology, Brain Neoplasms blood supply, Brain Neoplasms enzymology, Endothelium, Vascular enzymology, Glioblastoma blood supply, Glioblastoma enzymology, Transforming Growth Factor beta physiology
- Abstract
Angiotensin peptides are potent vasoconstrictors, cell growth factors, and neuromodulators in normal and pathological situations. To assess the potential role of the angiotensins in brain tumor-associated vessels, the expression of the enzymes of the angiotensin cascade were evaluated in these tumors. The production of these bioactive peptides is dependent on the activities of exopeptidases, including several aminopeptidases and carboxypeptidases, producing angiotensin (Ang) I, II, III, IV and Ang 1-7. Human cerebral parenchymal and glioblastoma cells expressed renin, and tumor vasculature, but not glioblastoma cells, expressed angiotensin-converting enzyme. High aminopeptidase A (APA) activity, but no aminopeptidase N/B activity, was observed in human brain tumor vasculature, suggesting a predominant production of Ang III. Grafting of rat glioma cells in rat brains yielded tumors with high APA and low aminopeptidase N/B activities in tumor vessels, confirming human results. Tumor growth and APA activity in tumor vessels were not affected by chronic angiotensin-converting enzyme inhibition. The brain-derived EC219 endothelial cells expressed high APA activity, which was not involved in endothelial cell proliferation, but was down-regulated by exposure of cells to transforming growth factor-beta (TGF beta) or to TGF beta-secreting tumor cells, suggesting a role for this peptide in the control of APA activity in cerebral vasculature. Thus, APA is a potential marker of chronic dysfunction, involving loss of TGF beta function, of the metabolic blood-brain barrier, but not of neovascularization.
- Published
- 2000
- Full Text
- View/download PDF
32. Thrombospondin-1 is downregulated by anoxia and suppresses tumorigenicity of human glioblastoma cells.
- Author
-
Tenan M, Fulci G, Albertoni M, Diserens AC, Hamou MF, El Atifi-Borel M, Feige JJ, Pepper MS, and Van Meir EG
- Subjects
- 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, Glioblastoma genetics, Hypoxia genetics, Thrombospondin 1 genetics
- 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.
- Published
- 2000
- Full Text
- View/download PDF
33. Identification of the putative brain tumor antigen BF7/GE2 as the (de)toxifying enzyme microsomal epoxide hydrolase.
- Author
-
Kessler R, Hamou MF, Albertoni M, de Tribolet N, Arand M, and Van Meir EG
- Subjects
- Animals, Antibodies, Monoclonal immunology, Antibodies, Neoplasm immunology, Antibody Specificity, Antigens, Neoplasm metabolism, Brain Neoplasms enzymology, Epoxide Hydrolases immunology, Epoxide Hydrolases metabolism, Glioma enzymology, Humans, Mice, Rats, Transfection, Tumor Cells, Cultured, Antigens, Neoplasm analysis, Brain Neoplasms immunology, Epoxide Hydrolases analysis, Glioma immunology
- Abstract
Malignant gliomas are the main cause of death from primary brain tumors. Despite surgery, radiation, and chemotherapy, patients have a median survival of less than a few years; therefore, it is clearly imperative to investigate new ways of treatment. The development of new therapeutic strategies for brain tumors is dependent on a better understanding of the differences between normal and tumoral brain cells. Our group had described previously a Mr 48,000 antigen defined by reactivity with two monoclonal antibodies (GE2 and BF7) obtained by immunization of mice with human glioblastoma cells. Here, we describe the identification of the GE2/BF7 antigen as microsomal epoxide hydrolase (mEH), a drug-metabolizing enzyme that is involved both in toxification and detoxification of carcinogens. We initially used immunoaffinity purification using GE2 and BF7 and analyzed the purified proteins by microsequencing. Edman degradation identified 15 amino acids of the NH2-terminal sequence that were 100% identical to mEH. To further confirm the identity of the BF7/GE2 antigen as mEH, we showed that the protein immunopurified with GE2 and BF7 was recognized by an anti-mEH antibody and that in vitro and in vivo synthesized human mEH is recognized by BF7 and GE2 antibodies. Furthermore, anti-mEH antibody recognizes an antigen expressed both in gliomas and reactive astrocytes, as do BF7 and GE2. Finally, we demonstrate that in contrast to what has been reported in rat embryo fibroblasts, p53 does not regulate mEH mRNA expression in glioma cells.
- Published
- 2000
34. Cells with TP53 mutations in low grade astrocytic tumors evolve clonally to malignancy and are an unfavorable prognostic factor.
- Author
-
Ishii N, Tada M, Hamou MF, Janzer RC, Meagher-Villemure K, Wiestler OD, Tribolet N, and Van Meir EG
- Subjects
- Adolescent, Adult, Aged, Astrocytoma classification, Astrocytoma physiopathology, Astrocytoma radiotherapy, Brain Neoplasms classification, Brain Neoplasms physiopathology, Brain Neoplasms radiotherapy, Child, Preschool, Clone Cells, Disease Progression, Female, Humans, Male, Middle Aged, Prognosis, Recurrence, Astrocytoma genetics, Brain Neoplasms genetics, Mutation, Tumor Suppressor Protein p53 genetics
- Abstract
It is important to understand how low grade tumors recur and progress to malignant lesions since this dramatically shortens patient survival. Here, we evaluated the concept that malignant progression and poor prognosis of low grade astrocytic tumors are TP53 dependent through clonal expansion of mutated cells. TP53 status was established in primary and recurrent tumors from 36 patients with WHO grade II astrocytic tumors and two tumor types were found. Tumors from 14 patients (39%; type 1) had TP53 mutated cells, and 92% of these recurred with 57% progressing to malignancy. The evolution of TP53 mutated cells before and after progression was examined using a clonal analysis procedure in yeast. Malignant progression was accompanied by an increased percentage of mutant TP53 (red) yeast colonies resulting from monoclonal expansion of cells with mutated TP53. The presence of TP53 mutations in WHO grade II astrocytic tumors was associated with malignant progression (P=0. 034, chi2 test) and shorter progression-free survival (PFS; 47.6+/-9. 6 months for TP53-mutated tumors vs 67.8+/-8.2 months for TP53-wild type tumors, P<0.05, log-rank test). Tumors from 22 patients (61%; type 2) were without TP53 mutations, and 64% of these recurred without a change in TP53 status, although 41% progressed to malignancy. This suggests that TP53 mutation is not an initiating or progression event in the majority of low grade astrocytic tumors. Our study also indicates that irradiation for WHO grade II astrocytic tumors might be associated with poor outcome (P<0.0001) and this was independent of TP53 status. These findings have important implications in the clinical management of patients with low grade astocytoma and provide new support to the clonal evolution model for tumor progression.
- Published
- 1999
- Full Text
- View/download PDF
35. Regulation of interleukin-8 expression by reduced oxygen pressure in human glioblastoma.
- Author
-
Desbaillets I, Diserens AC, de Tribolet N, Hamou MF, and Van Meir EG
- Subjects
- 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, Interleukin-8 genetics, Oxygen physiology
- 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.
- Published
- 1999
- Full Text
- View/download PDF
36. Somatic deletion mapping on chromosome 10 and sequence analysis of PTEN/MMAC1 point to the 10q25-26 region as the primary target in low-grade and high-grade gliomas.
- Author
-
Maier D, Zhang Z, Taylor E, Hamou MF, Gratzl O, Van Meir EG, Scott RJ, and Merlo A
- Subjects
- Amino Acid Substitution, Chromosome Mapping, DNA, Neoplasm analysis, DNA, Neoplasm genetics, Dinucleotide Repeats genetics, Gene Deletion, Genes, Tumor Suppressor genetics, Genetic Markers genetics, Glioma pathology, Humans, Loss of Heterozygosity, Mutation genetics, PTEN Phosphohydrolase, Sequence Analysis, DNA, Severity of Illness Index, Chromosomes, Human, Pair 10 genetics, Glioma genetics, Phosphoric Monoester Hydrolases, Protein Tyrosine Phosphatases genetics, Tumor Suppressor Proteins
- Abstract
The 10q25-26 region between the dinucleotide markers D10S587 and D10S216 is deleted in glioblastomas and, as we have recently shown, in low-grade oligodendrogliomas. We further refined somatic mapping on 10q23-tel and simultaneously assessed the role of the candidate tumor suppressor gene PTEN/MMAC1 in glial neoplasms by sequence analysis of eight low-grade and 24 high-grade gliomas. These tumors were selected for partial or complete loss of chromosome 10 based on deletion mapping with increased microsatellite marker density at 10q23-tel. Three out of eight (38%) low-grade and 3/24 (13%) high-grade gliomas exclusively target 10q25-26. We did not find a tumor only targeting 10q23.3, and most tumors (23/32, 72%) showed large deletions on 10q including both regions. The sequence analysis of PTEN/MMAC1 revealed nucleotide alterations in 1/8 (12.5%) low-grade gliomas in a tumor with LOH at l0q21-qtel and in 5/21 (24%) high-grade gliomas displaying LOH that always included 10q23-26. Our refined mapping data point to the 10q25-26 region as the primary target on 10q, an area that also harbors the DMBT1 candidate tumor suppressor gene. The fact that we find hemizygous deletions at 10q25-qtel in low-grade astrocytomas and oligodendrogliomas - two histologically distinct entities of gliomas - suggests the existence of a putative suppressor gene involved early in glial tumorigenesis.
- Published
- 1998
- Full Text
- View/download PDF
37. Upregulation of interleukin 8 by oxygen-deprived cells in glioblastoma suggests a role in leukocyte activation, chemotaxis, and angiogenesis.
- Author
-
Desbaillets I, Diserens AC, Tribolet N, Hamou MF, and Van Meir EG
- Subjects
- 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, Chemotaxis, Leukocyte, Glioblastoma metabolism, Glioblastoma pathology, Interleukin-8 metabolism, Neovascularization, Physiologic immunology, Up-Regulation
- 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.
- Published
- 1997
- Full Text
- View/download PDF
38. [Inhibitory effect of antisense LIF oligonucleotide on the outgrowth of human medulloblastoma cells].
- Author
-
Liu J, Li H, and Hamou MF
- Subjects
- Adolescent, Brain Neoplasms genetics, Cell Division drug effects, DNA, Neoplasm biosynthesis, Gene Expression drug effects, Humans, Leukemia Inhibitory Factor, Male, Medulloblastoma genetics, Tumor Cells, Cultured drug effects, Brain Neoplasms pathology, Growth Inhibitors genetics, Interleukin-6, Lymphokines genetics, Medulloblastoma pathology, Oligonucleotides, Antisense pharmacology
- Abstract
In order to analyse the functional importance of leukemia inhibitory factor (LIF) in the prolliferation of human medulloblastoma cells, antisense LIF oligonucleotides were used to block LIF gene expression in established medulloblastoma cell line Med-3. The effects of this measure on Med-3 cells were then investigated by using different approaches. The results revealed that LIF expression in the cells treated by antisense LIF reduced below the threshold being detectable with RT-CPR and anti-LIF immunocytochemistry also showed a negative lebelling. Meanwhile, proliferation rate of antisense treated cells decreased distinctively. In contrast, the LIF gene expression as well as proliferation of Med-3 cells treated by sense oligonucleotides remained similar as that of the normally cultured cells. The data suggest a LIF oriented gene/immune trial for the therapy of medulloblastomas and other LIF growth-associated tumors of humans.
- Published
- 1996
39. [Growth promoting effect of IL-6 on medulloblastoma cells in vitro].
- Author
-
Liu J, Li H, and Hamou MF
- Subjects
- Base Sequence, Cell Division, Cerebellar Neoplasms genetics, DNA, Neoplasm biosynthesis, Gene Expression, Humans, Interleukin-6 genetics, Medulloblastoma genetics, Molecular Sequence Data, Morpholines, Signal Transduction, Tumor Cells, Cultured drug effects, Cerebellar Neoplasms pathology, Interleukin-6 pharmacology, Medulloblastoma pathology
- Abstract
Expression of IL-6 and IL-6 signal transducer genes (IL-6R and gp130) in human medulloblastoma cells were investigated by Northern blot analysis, RT/PCR and immunocytochemical staining. The results revealed that among 13 tumor cases and 3 established medulloblastoma cell lines, 95% of them were found to express full set of IL-6 signal transducer genes, IL-6R and gp 130, but none of them expressed IL-6. Incubation of medulloblastoma cells in vitro with recombinant IL-6 could apparently stimulate cell growth and enhance DNA synthesis. Our data thus for the first time demonstrate that though IL-6 is not expressed by medulloblastoma cells themselves, IL-6 can act on medulloblastoma cells through its signal transducers by a paracrine pathway. Since medulloblastomas are surrounded by gliosis and infiltrated by immune activated cells which are known to express IL-6, the influence of those IL-6 producing cells on the development and progression of medulloblastomas should be taken into account.
- Published
- 1995
40. Differential CD44 expression patterns in primary brain tumours and brain metastases.
- Author
-
Li H, Liu J, Hofmann M, Hamou MF, and de Tribolet N
- Subjects
- Adult, Aged, Base Sequence, Brain Neoplasms secondary, Female, Humans, Hyaluronan Receptors, Male, Middle Aged, Molecular Sequence Data, Polymerase Chain Reaction, Brain Neoplasms chemistry, Carrier Proteins analysis, Receptors, Cell Surface analysis, Receptors, Lymphocyte Homing analysis
- Abstract
Splicing variants of CD44 (CD44v) are increasingly recognised as metastasis-promoting factors in rodent and some human cancers. However, the frequency for CD44v expression in human cancers and their metastases and the status of CD44v expression in low or non-metastatic tumours is still uncertain. To address this issue, we investigated CD44 expression patterns in brain metastases (BMTs) spread from more than ten organs and five types of primary brain tumours (PBTs) by Northern blot, reverse transcription-polymerase chain reaction (RT-PCR) and immunocytochemical analysis. The results demonstrated that all of the 56 PBTs examined express standard form of CD44 (CD44s) but none of them express CD44v. In contrast, 22 of 26 BMTs studied were found with CD44v expression. Our data thus present direct evidence of a general distribution of CD44 in BMTs but suggest that such expression is an extremely rare event in PBTs. Therefore, the presence or absence of CD44v expression may be related to high or low metastatic potential of human malignancies.
- Published
- 1995
- Full Text
- View/download PDF
41. Human astrocytomas and glioblastomas express monocyte chemoattractant protein-1 (MCP-1) in vivo and in vitro.
- Author
-
Desbaillets I, Tada M, de Tribolet N, Diserens AC, Hamou MF, and Van Meir EG
- Subjects
- 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, Astrocytoma metabolism, Brain Neoplasms metabolism, Chemotactic Factors biosynthesis, Glioblastoma metabolism
- 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.
- Published
- 1994
- Full Text
- View/download PDF
42. IL-6 stimulates growth and inhibits constitutive, protein synthesis-independent apoptosis of murine B-cell hybridoma 7TD1.
- Author
-
Liu J, Li H, de Tribolet N, Jaufeerally R, Hamou MF, and Van Meir EG
- Subjects
- Animals, Aurintricarboxylic Acid pharmacology, B-Lymphocytes cytology, Cycloheximide pharmacology, Hybridomas, Mice, Proto-Oncogene Proteins biosynthesis, Proto-Oncogene Proteins c-bcl-2, Proto-Oncogene Proteins c-myc biosynthesis, Apoptosis drug effects, B-Lymphocytes drug effects, Interleukin-6 pharmacology
- Abstract
Interleukin-6 (IL-6) is a well-known survival/growth factor for murine B-cell hybridoma 7TD1 cells. In this study, we analyzed the behavior of 7TD1 cells in the absence or presence of IL-6. After IL-6 deprivation, the cells became growth arrested and demonstrated signs considered to be hallmarks of apoptosis. To assess the underlying molecular mechanism of IL-6-regulated apoptosis, two chemical compounds, cycloheximide (CHX) and aurintricarboxylic acid (ATA), were used to treat the cells. ATA, a DNA endonuclease inhibitor, efficiently prevented DNA fragmentation and increased cell survival in the absence of IL-6. CHX, an inhibitor of protein synthesis, failed to prevent apoptosis and blocked cell survival signals induced by IL-6. These results suggest, first, that IL-6 promotes 7TD1 cell survival by inhibiting an endogenous constitutive cell death program and, second, that this inhibition requires de novo protein synthesis.
- Published
- 1994
- Full Text
- View/download PDF
43. Production of interleukin-1 receptor antagonist by human glioblastoma cells in vitro and in vivo.
- Author
-
Tada M, Diserens AC, Desbaillets I, Jaufeerally R, Hamou MF, and de Tribolet N
- Subjects
- 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, Glioblastoma metabolism, Interleukin-1 metabolism, Receptors, Interleukin antagonists & inhibitors
- 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.
- Published
- 1994
- Full Text
- View/download PDF
44. [Overexpression of p53 protein in gliomas].
- Author
-
Kikuchi T, Diserens AC, Hamou MF, de Tribolet N, and van Meir E
- Subjects
- Astrocytoma genetics, Brain Neoplasms genetics, DNA, Neoplasm analysis, Enzyme-Linked Immunosorbent Assay, Humans, Mutation, Polymerase Chain Reaction, Astrocytoma metabolism, Brain Neoplasms metabolism, Genes, p53, Tumor Suppressor Protein p53 analysis
- 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.
- Published
- 1994
45. Variant CD44 adhesion molecules are expressed in human brain metastases but not in glioblastomas.
- Author
-
Li H, Hamou MF, de Tribolet N, Jaufeerally R, Hofmann M, Diserens AC, and Van Meir EG
- Subjects
- Base Sequence, Blotting, Northern, Humans, Hyaluronan Receptors, Molecular Sequence Data, RNA, Neoplasm chemistry, Tumor Cells, Cultured, Biomarkers, Tumor analysis, Brain Neoplasms immunology, Brain Neoplasms secondary, Glioblastoma immunology, RNA, Neoplasm analysis, Receptors, Lymphocyte Homing analysis
- 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.
- Published
- 1993
46. Expression and release of interleukin-1 by human glioblastoma cells in vitro and in vivo.
- Author
-
Gauthier T, Hamou MF, Monod L, Gallay P, Carrel S, and de Tribolet N
- Subjects
- Brain Neoplasms pathology, Cell Line, Glial Fibrillary Acidic Protein physiology, Glioma pathology, Humans, Immunoenzyme Techniques, Interleukin-1 metabolism, RNA, Neoplasm genetics, Transcription, Genetic, Brain Neoplasms genetics, Gene Expression Regulation, Neoplastic physiology, Glioma genetics, Interleukin-1 genetics, Tumor Cells, Cultured physiology
- Abstract
The present study demonstrates interleukin-1 (IL-1) production by human glioblastoma cells both in vitro and in vivo. The presence of IL-1 alpha and IL-1 beta transcripts was analyzed in 4 cell lines. IL-1 alpha mRNA was expressed constitutively in one cell line whereas constitutive IL-1 beta mRNA could not be detected in any of the cell lines. IL-1 alpha transcripts could be induced with phorbol myristate acetate (PMA) or PMA plus lipopolysaccharide (LPS) in 2 of 4 cell lines and IL-1 beta mRNA in 2 of 4 cell lines. Culture fluid from these cell lines was tested for the presence of IL-1 using a specific radio-immuno-assay for either IL-1 alpha or IL-1 beta. In agreement with the results on RNA, one of 4 cell lines was found to constitutively produce IL-1 alpha but not IL-1 beta. After treatment with PMA and LPS, IL-1 alpha was detected in the culture fluid from two other lines and IL-1 beta in the medium from three lines. That the IL-1 produced by these cell lines was biologically active was confirmed in a two step thymocyte proliferation assay. IL-1 like activity was detected in all samples that were positive in the radio-immuno-assay. Finally, immunohistological analysis on fresh frozen tumour sections provided evidence for IL-1 production by glioblastoma cells in vivo. Fourteen out of 28 glioblastomas were stained with an anti-IL-1 alpha monoclonal antibody while none of them was stained with an anti-IL-1 beta antibody.
- Published
- 1993
- Full Text
- View/download PDF
47. Differential expression of the CD44 molecule in human brain tumours.
- Author
-
Kuppner MC, Van Meir E, Gauthier T, Hamou MF, and de Tribolet N
- Subjects
- Flow Cytometry, Gene Expression, Glioma immunology, Humans, Immunoenzyme Techniques, Medulloblastoma immunology, Meningioma immunology, Molecular Weight, RNA, Messenger genetics, Receptors, Lymphocyte Homing chemistry, Brain Neoplasms immunology, Receptors, Lymphocyte Homing metabolism
- Abstract
Expression of the CD44 molecule was examined in a variety of human brain tumours, brain metastases and normal brain. Immunohistological staining with several CD44 antibodies demonstrated differential expression of the CD44 molecule among different brain tumour types. CD44 was strongly expressed in high-grade gliomas and weakly expressed in meningiomas, medulloblastomas and normal brain. Northern blot analysis revealed the presence of 3 major CD44 mRNAs of 1.6, 2.2, and 5.0 kb in glioblastomas and a mRNA of 5.6 kb in meningiomas. CD44 expression was also detected by flow cytometric analysis on cultured cells derived from a variety of human brain tumours including glioblastomas and meningiomas.
- Published
- 1992
- Full Text
- View/download PDF
48. Expression of cALLa/NEP on gliomas: a possible marker of malignancy.
- Author
-
Monod L, Hamou MF, Ronco P, Verroust P, and de Tribolet N
- Subjects
- Adolescent, Adult, Biopsy, Brain pathology, Child, Female, Humans, Immunoenzyme Techniques, Male, Middle Aged, Neoplasm Staging, Antigens, Differentiation analysis, Antigens, Neoplasm analysis, Biomarkers, Tumor analysis, Brain Neoplasms pathology, Glioma pathology, Neprilysin analysis
- Abstract
First described on pre-B leukemia cells, the common acute lymphoblastic leukemia antigen (cALLa) is also expressed on glioma cells in vitro. Its identity to neutral endopeptidase (NEP) (E.C.3.24.11) was corroborated by our finding that cALLa positive glioma cells had NEP activity. To study cALLa/NEP distribution on glial tumours in vivo, we examined 76 brain tumour biopsies by immunostaining techniques on frozen tissue sections using anti-cALLa (FAH99) and anti-NEP (135 A 3) monoclonal antibodies. We found that 96% of grade 4 gliomas (25/26) expressed NEP. Whereas only 45% (4/9) of grade 3 or anaplastic astrocytomas did. In low grade gliomas, we found 2 positive tumours out of 21 tested (10%). Double immunostaining procedures revealed that NEP was co-expressed with GFAP. However no NEP could be detected on non-glial brain tumours nor on reactive astrocytes. These results suggest that cALLa/NEP expression could be linked to malignant progression of gliomas.
- Published
- 1992
- Full Text
- View/download PDF
49. Human glioblastoma cells release interleukin 6 in vivo and in vitro.
- Author
-
Van Meir E, Sawamura Y, Diserens AC, Hamou MF, and de Tribolet N
- Subjects
- 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, Glioma metabolism, Interleukin-6 metabolism
- 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.
- Published
- 1990
50. Activation and adhesion molecule expression on lymphoid infiltrates in human glioblastomas.
- Author
-
Kuppner MC, Hamou MF, and de Tribolet N
- Subjects
- Antigens, CD physiology, Antigens, Differentiation analysis, Cell Adhesion, Cell Adhesion Molecules physiology, Cell Movement, Histocompatibility Antigens analysis, Humans, Immunohistochemistry, Integrin beta1, Leukocyte Common Antigens, Lymphocyte Function-Associated Antigen-1 analysis, Receptors, Lymphocyte Homing analysis, Antigens, CD analysis, Brain Neoplasms immunology, Cell Adhesion Molecules analysis, Glioma immunology, Lymphocytes immunology
- Abstract
Frozen tissue sections obtained from human glioblastomas, brain tumor metastases and normal brain were examined for the expression of molecules known to be involved in lymphocyte activation and/or adhesion and migration. The molecules studied included CD3, CD45R, UCHL-1 (CD45RO), lymphocyte function-associated antigen 1 (LFA-1) (CD11a, CD18), intercellular adhesion molecule 1 (ICAM-1) (CD54), 4B4 (CD29), CD44, CD2, and LFA-3 (CD58). CD3+ lymphocytes infiltrating human glioblastomas and brain tumor metastases expressed LFA-1 alpha and beta. Many cells were also UCHL-1+ whereas only a small percentage were CD45R+. CD2+ lymphocytes were also present. Tumor-infiltrating lymphocytes (TIL) were found to be negative for CD29, which was, however, expressed on intratumoral vessels in addition to vessels found in normal brain. Glioblastoma cells and intratumoral vessels expressed ICAM-1 whereas no ICAM-1 was found on TIL or on normal brain. Glioblastoma cells also expressed high levels of both CD44 and LFA-3 whereas TIL were negative for these antigens. CD44 was also expressed on certain regions of normal brain. Antibodies to LFA-1 alpha and -beta and ICAM-1 could significantly block the binding of lymphokine-activated killer (LAK) cells or TIL to human glioblastoma cells suggesting that these molecules play a role in the binding and subsequent migration of lymphocytes into brain tumor tissue.
- Published
- 1990
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.