Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters










Publication year range
1.
NMR Biomed ; 29(11): 1577-1589, 2016 11.
Article in English | MEDLINE | ID: mdl-27717037

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 T2 -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.


Subject(s)
Brain Neoplasms/diagnostic imaging , Brain Neoplasms/pathology , Diffusion Magnetic Resonance Imaging/methods , Early Detection of Cancer/methods , Glioma/diagnostic imaging , Glioma/pathology , 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
2.
Oncogene ; 35(1): 12-21, 2016 Jan 07.
Article in English | MEDLINE | ID: mdl-25772239

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.


Subject(s)
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 , 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
3.
Acta Neurochir Suppl ; 86: 495-8, 2003.
Article in English | MEDLINE | ID: mdl-14753493

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.


Subject(s)
Aquaporins/metabolism , Brain Neoplasms/metabolism , Brain/metabolism , Glioma/metabolism , Subarachnoid Hemorrhage/metabolism , Aquaporin 1 , Aquaporin 4 , Blood Group Antigens , Blotting, Western , Case-Control Studies , Humans , Immunohistochemistry
4.
Brain Pathol ; 11(3): 296-305, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11414472

ABSTRACT

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


Subject(s)
DNA-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic , Meningeal Neoplasms/genetics , Meningioma/genetics , Neoplasm Proteins/genetics , Nuclear Proteins/genetics , Aged , Codon/genetics , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/biosynthesis , Cyclins/genetics , DNA Mutational Analysis , DNA, Neoplasm/genetics , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/physiology , Disease Progression , Female , Genes, Tumor Suppressor , Humans , Male , Meningeal Neoplasms/metabolism , Meningeal Neoplasms/pathology , Meningioma/metabolism , Meningioma/pathology , Middle Aged , Neoplasm Proteins/biosynthesis , Nuclear Proteins/biosynthesis , Nuclear Proteins/physiology , RNA Splicing , RNA, Messenger/biosynthesis , RNA, Neoplasm/biosynthesis , Recombinant Fusion Proteins/physiology , Reverse Transcriptase Polymerase Chain Reaction , Saccharomyces cerevisiae/genetics , Transcriptional Activation , Tumor Protein p73 , Tumor Suppressor Protein p53/physiology , Tumor Suppressor Proteins
5.
Lab Invest ; 80(6): 973-80, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10879747

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.


Subject(s)
Aminopeptidases/metabolism , Angiotensins/physiology , Brain Neoplasms/blood supply , Brain Neoplasms/enzymology , Endothelium, Vascular/enzymology , Glioblastoma/blood supply , Glioblastoma/enzymology , Transforming Growth Factor beta/physiology , 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
6.
Cancer Res ; 60(11): 3019-24, 2000 Jun 01.
Article in English | MEDLINE | ID: mdl-10850451

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.


Subject(s)
Brain Neoplasms/genetics , Genes, Dominant , Genes, p53/genetics , Mutation , 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
7.
J Exp Med ; 191(10): 1789-98, 2000 May 15.
Article in English | MEDLINE | ID: mdl-10811871

ABSTRACT

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


Subject(s)
Glioblastoma/genetics , Hypoxia/genetics , Thrombospondin 1/genetics , Animals , Base Sequence , DNA Primers/genetics , Down-Regulation , Genes, p53 , Glioblastoma/blood supply , Glioblastoma/metabolism , Humans , Hypoxia/physiopathology , Male , Mice , Mice, Nude , Neoplasm Transplantation , Neovascularization, Pathologic/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Thrombospondin 1/biosynthesis , Transplantation, Heterologous , Tumor Cells, Cultured
8.
Cancer Res ; 60(5): 1403-9, 2000 Mar 01.
Article in English | MEDLINE | ID: mdl-10728706

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.


Subject(s)
Antigens, Neoplasm/analysis , Brain Neoplasms/immunology , Epoxide Hydrolases/analysis , Glioma/immunology , 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
9.
Oncogene ; 18(43): 5870-8, 1999 Oct 21.
Article in English | MEDLINE | ID: mdl-10557074

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.


Subject(s)
Astrocytoma/genetics , Brain Neoplasms/genetics , Mutation , Tumor Suppressor Protein p53/genetics , 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
10.
Oncogene ; 18(7): 1447-56, 1999 Feb 18.
Article in English | MEDLINE | ID: mdl-10050881

ABSTRACT

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


Subject(s)
Interleukin-8/genetics , Oxygen/physiology , Animals , Cell Hypoxia , Cobalt/pharmacology , Dactinomycin/pharmacology , Dichlororibofuranosylbenzimidazole/pharmacology , Endothelial Growth Factors/genetics , Gene Expression Regulation , Glioblastoma , Humans , Lymphokines/genetics , Mice , Nucleic Acid Synthesis Inhibitors/pharmacology , Oxidation-Reduction , RNA, Messenger , Response Elements , Transcription Factor AP-1/metabolism , Transcription, Genetic , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
11.
Oncogene ; 16(25): 3331-5, 1998 Jun 25.
Article in English | MEDLINE | ID: mdl-9681833

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.


Subject(s)
Chromosomes, Human, Pair 10/genetics , Glioma/genetics , Phosphoric Monoester Hydrolases , Protein Tyrosine Phosphatases/genetics , Tumor Suppressor Proteins , 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
12.
J Exp Med ; 186(8): 1201-12, 1997 Oct 20.
Article in English | MEDLINE | ID: mdl-9334359

ABSTRACT

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


Subject(s)
Chemotaxis, Leukocyte , Glioblastoma/metabolism , Glioblastoma/pathology , Interleukin-8/metabolism , Neovascularization, Physiologic/immunology , Up-Regulation , Anaerobiosis , Antigens, CD/biosynthesis , Blotting, Northern , Cell Hypoxia/immunology , Disease Progression , Glioblastoma/physiopathology , Humans , In Situ Hybridization , Interleukin-8/biosynthesis , Interleukin-8/genetics , Necrosis , RNA, Messenger/biosynthesis , Receptors, Interleukin/biosynthesis , Receptors, Interleukin-8A , Spheroids, Cellular
13.
Zhonghua Bing Li Xue Za Zhi ; 25(3): 132-4, 1996 Jun.
Article in Chinese | MEDLINE | ID: mdl-9275663

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.


Subject(s)
Brain Neoplasms/pathology , Growth Inhibitors/genetics , Interleukin-6 , Lymphokines/genetics , Medulloblastoma/pathology , Oligonucleotides, Antisense/pharmacology , 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
14.
Zhonghua Zhong Liu Za Zhi ; 17(4): 271-4, 1995 Jul.
Article in Chinese | MEDLINE | ID: mdl-7587893

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.


Subject(s)
Cerebellar Neoplasms/pathology , Interleukin-6/pharmacology , Medulloblastoma/pathology , 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
15.
Br J Cancer ; 72(1): 160-3, 1995 Jul.
Article in English | MEDLINE | ID: mdl-7541233

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.


Subject(s)
Brain Neoplasms/chemistry , Carrier Proteins/analysis , Receptors, Cell Surface/analysis , Receptors, Lymphocyte Homing/analysis , Adult , Aged , Base Sequence , Brain Neoplasms/secondary , Female , Humans , Hyaluronan Receptors , Male , Middle Aged , Molecular Sequence Data , Polymerase Chain Reaction
16.
Int J Cancer ; 58(2): 240-7, 1994 Jul 15.
Article in English | MEDLINE | ID: mdl-7517920

ABSTRACT

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


Subject(s)
Astrocytoma/metabolism , Brain Neoplasms/metabolism , Chemotactic Factors/biosynthesis , Glioblastoma/metabolism , Astrocytoma/genetics , Base Sequence , Blotting, Northern , Brain Neoplasms/genetics , Chemokine CCL2 , Chemotactic Factors/genetics , Chemotactic Factors/metabolism , Cytokines/biosynthesis , Cytokines/genetics , Cytokines/metabolism , Gene Expression , Glioblastoma/genetics , Humans , Molecular Sequence Data , Polymerase Chain Reaction , RNA, Messenger/genetics , RNA-Directed DNA Polymerase , Tumor Cells, Cultured
17.
Cell Immunol ; 155(2): 428-35, 1994 May.
Article in English | MEDLINE | ID: mdl-8181074

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.


Subject(s)
Apoptosis/drug effects , B-Lymphocytes/drug effects , Interleukin-6/pharmacology , 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
18.
J Neuroimmunol ; 50(2): 187-94, 1994 Mar.
Article in English | MEDLINE | ID: mdl-8120140

ABSTRACT

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


Subject(s)
Glioblastoma/metabolism , Interleukin-1/metabolism , Receptors, Interleukin/antagonists & inhibitors , Base Sequence , Cytokines/physiology , Glioblastoma/pathology , Humans , Immunohistochemistry , Intracellular Membranes/metabolism , Molecular Sequence Data , Oligonucleotide Probes/genetics , Polymerase Chain Reaction , RNA, Messenger/metabolism , Tumor Cells, Cultured
19.
No Shinkei Geka ; 22(2): 125-9, 1994 Feb.
Article in Japanese | MEDLINE | ID: mdl-8115006

ABSTRACT

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


Subject(s)
Astrocytoma/metabolism , Brain Neoplasms/metabolism , Genes, p53 , Tumor Suppressor Protein p53/analysis , Astrocytoma/genetics , Brain Neoplasms/genetics , DNA, Neoplasm/analysis , Enzyme-Linked Immunosorbent Assay , Humans , Mutation , Polymerase Chain Reaction
20.
Cancer Res ; 53(22): 5345-9, 1993 Nov 15.
Article in English | MEDLINE | ID: mdl-7693337

ABSTRACT

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


Subject(s)
Biomarkers, Tumor/analysis , Brain Neoplasms/immunology , Brain Neoplasms/secondary , Glioblastoma/immunology , RNA, Neoplasm/analysis , Receptors, Lymphocyte Homing/analysis , Base Sequence , Blotting, Northern , Humans , Hyaluronan Receptors , Molecular Sequence Data , RNA, Neoplasm/chemistry , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...