Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 79
Filter
1.
Acta Neuropathol Commun ; 6(1): 18, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29490700

ABSTRACT

Despite multidisciplinary local and systemic therapeutic approaches, the prognosis for most patients with brain metastases is still dismal. The role of adaptive and innate anti-tumor response including the Human Leukocyte Antigen (HLA) machinery of antigen presentation is still unclear. We present data on the HLA class II-chaperone molecule CD74 in brain metastases and its impact on the HLA peptidome complexity.We analyzed CD74 and HLA class II expression on tumor cells in a subset of 236 human brain metastases, primary tumors and peripheral metastases of different entities in association with clinical data including overall survival. Additionally, we assessed whole DNA methylome profiles including CD74 promoter methylation and differential methylation in 21 brain metastases. We analyzed the effects of a siRNA mediated CD74 knockdown on HLA-expression and HLA peptidome composition in a brain metastatic melanoma cell line.We observed that CD74 expression on tumor cells is a strong positive prognostic marker in brain metastasis patients and positively associated with tumor-infiltrating T-lymphocytes (TILs). Whole DNA methylome analysis suggested that CD74 tumor cell expression might be regulated epigenetically via CD74 promoter methylation. CD74high and TILhigh tumors displayed a differential DNA methylation pattern with highest enrichment scores for antigen processing and presentation. Furthermore, CD74 knockdown in vitro lead to a reduction of HLA class II peptidome complexity, while HLA class I peptidome remained unaffected.In summary, our results demonstrate that a functional HLA class II processing machinery in brain metastatic tumor cells, reflected by a high expression of CD74 and a complex tumor cell HLA peptidome, seems to be crucial for better patient prognosis.


Subject(s)
Antigens, CD/metabolism , Brain Neoplasms/metabolism , Brain Neoplasms/secondary , Genes, MHC Class II , Sialyltransferases/metabolism , Antigens, CD/genetics , Biomarkers, Tumor/metabolism , Brain/metabolism , Brain/pathology , Brain Neoplasms/mortality , Cell Line, Tumor , Cohort Studies , DNA Methylation , Gene Knockdown Techniques , Humans , Kaplan-Meier Estimate , Melanoma/metabolism , Melanoma/pathology , Prognosis , Promoter Regions, Genetic , Sialyltransferases/genetics , T-Lymphocytes/metabolism , T-Lymphocytes/pathology
2.
Neuropathol Appl Neurobiol ; 40(2): 205-16, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24117486

ABSTRACT

AIMS: The Far Upstream Element [FUSE] Binding Protein 1 (FUBP1) regulates target genes, such as the cell cycle regulators MYC and p21. FUBP1 is up-regulated in many tumours and acts as an oncoprotein by stimulating proliferation and inhibiting apoptosis. Recently, FUBP1 mutations were identified in approximately 15% of oligodendrogliomas. To date, all reported FUBP1 mutations have been predicted to inactivate FUBP1, which suggests that in contrast to most other tumours FUBP1 may act as a tumour suppressor in oligodendrogliomas. METHODS: As no data are currently available concerning FUBP1 protein levels in gliomas, we examined the FUBP1 expression profiles of human glial tumours by immunohistochemistry and immunofluorescence. We analysed FUBP1 expression related to morphological differentiation, IDH1 and FUBP1 mutation status, 1p/19q loss of heterozygosity (LOH) as well as proliferation rate. RESULTS: Our findings demonstrate that FUBP1 expression levels are increased in all glioma subtypes as compared with normal central nervous system (CNS) control tissue and are associated with increased proliferation. In contrast, FUBP1 immunonegativity predicted FUBP1 mutation with a sensitivity of 100% and a specificity of 90% in our cohort and was associated with oligodendroglial differentiation, IDH1 mutation and 1p/19q loss of heterozygosity (LOH). Using this approach, we detected a to-date undescribed FUBP1 mutation in an oligodendroglioma. CONCLUSION: In summary, our data indicate an association between of FUBP1 expression and proliferation in gliomas. Furthermore, our findings present FUBP1 immunohistochemical analysis as a helpful additional tool for neuropathological glioma diagnostics predicting FUBP1 mutation.


Subject(s)
DNA Helicases/genetics , DNA Helicases/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Oligodendroglioma/genetics , Oligodendroglioma/metabolism , Cell Differentiation , Cell Proliferation , Chromosome Deletion , Chromosomes, Human, Pair 1 , Chromosomes, Human, Pair 19 , Codon, Nonsense , Glioma/genetics , Glioma/metabolism , Humans , Isocitrate Dehydrogenase/genetics , Loss of Heterozygosity , Neurons/metabolism , RNA-Binding Proteins
3.
Brain ; 130(Pt 12): 3336-41, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17998256

ABSTRACT

A serum marker for malignant cerebral astrocytomas could improve both differential diagnosis and clinical management of brain tumour patients. To evaluate whether the serum concentration of glial fibrillary acidic protein (GFAP) may indicate glioblastoma multiforme (GBM) in patients with single supratentorial space-occupying lesions, we prospectively examined 50 consecutive patients with histologically proven GBM, World Health Organization (WHO) grade IV, 14 patients with anaplastic astrocytoma (WHO grade III), 4 patients with anaplastic oligodendroglioma, 13 patients with diffuse astrocytoma (WHO grade II), 17 patients with a single cerebral metastasis and 50 healthy controls. Serum was taken from the patients before tumour resection or stereotactic biopsy. Serum GFAP levels were determined using a commercially available ELISA test and were detectable in 40 out of the 50 GBM patients (median: 0.18 microg/l; range: 0-5.6 microg/l). The levels were significantly elevated compared with those of the non-GBM tumour patients and healthy controls (median: 0 mug/l; range: 0-0.024 microg/l; P < 0.0001, respectively). Non-GBM tumour patients and all healthy subjects showed zero serum GFAP levels. There was a significant correlation between tumour volume (Spearman Rho, CC = 0.47; 95% confidence interval, 0.2-0.67; P < 0.001), tumour necrosis volume (CC = 0.49; 95% confidence interval, 0.2-0.72; P = 0.004), the amount of necrotic GFAP positive cells (CC = 0.61; 95% confidence interval, 0.29-0.81; P = 0.007) and serum GFAP level among the GBM patients. A serum GFAP level of >0.05 microg/l was 76% sensitive and 100% specific for the diagnosis of GBM in patients with a single supratentorial mass lesion in this series. Therefore, it can be concluded that serum GFAP constitutes a diagnostic biomarker for GBM. Future studies should investigate whether serum GFAP could also be used to monitor therapeutic effects and whether it may have a prognostic value.


Subject(s)
Biomarkers, Tumor/blood , Brain Neoplasms/diagnosis , Glial Fibrillary Acidic Protein/blood , Glioblastoma/diagnosis , Adult , Aged , Aged, 80 and over , Brain Neoplasms/pathology , Female , Glioblastoma/pathology , Glioblastoma/secondary , Humans , Male , Middle Aged , Necrosis , Neoplasm Proteins/blood , Prospective Studies , Sensitivity and Specificity
4.
Acta Neurochir (Wien) ; 148(4): 473-7, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16341628

ABSTRACT

The incidence of diagnosed xanthogranuloma of the sellar region is very low [1, 2, 5, 6]. We report about two cases 1) in a 57-year-old female and 2) in a 5-year-old boy. In both cases radiographic findings revealed an inhomogeneous, contrast enhancing sellar lesion. Histopathology showed the typical features of a xanthogranuloma of the sellar region with cholesterol clefts, lympho-plasmacellular infiltrates, marked hemosiderin deposits, multinucleated foreign body giant cells around cholesterol clefts, accumulation of macrophages and only small epithelial cell clusters [6]. As xanthogranuloma of the sellar region are rarely diagnosed we want to draw attention to this rather unusual diagnosis.


Subject(s)
Pituitary Gland/pathology , Sella Turcica/pathology , Skull Base Neoplasms/diagnosis , Skull Base Neoplasms/physiopathology , Xanthogranuloma, Juvenile/diagnosis , Xanthogranuloma, Juvenile/physiopathology , Child, Preschool , Cholesterol/metabolism , Craniopharyngioma/diagnosis , Diagnosis, Differential , Female , Giant Cells, Foreign-Body/pathology , Hemosiderin/metabolism , Humans , Hypopituitarism/etiology , Hypopituitarism/physiopathology , Hypopituitarism/surgery , Lymphocytes/pathology , Macrophages/pathology , Male , Middle Aged , Neurosurgical Procedures , Pituitary Gland/physiopathology , Pituitary Gland/surgery , Postoperative Complications/etiology , Postoperative Complications/physiopathology , Postoperative Complications/therapy , Sella Turcica/physiopathology , Sella Turcica/surgery , Skull Base Neoplasms/surgery , Treatment Outcome , Xanthogranuloma, Juvenile/surgery
5.
J Neurol Neurosurg Psychiatry ; 75(3): 503-5, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14966177

ABSTRACT

Glial cell cytoplasmic inclusions were identified in a case of spinocerebellar ataxia type 2. These have not been reported before. The inclusions were found in low frequency in the dentate nucleus, cerebellar white matter, pontine transverse fibres, and the inferior olivary nucleus. They were of variable size and shape and expressed ubiquitin, thus resembling glial cytoplasmic inclusions in multiple system atrophy. However, their immunohistochemical profile was different as they did not show immunoreactivity for either tau protein or alpha-synuclein. There was no evidence of expanded polyglutamine tracts in these inclusions, which also failed to label with silver stains. As in many other neurodegenerative diseases, in spinocerebellar ataxia type 2 there may be pathogenic contributions of glial cells other than the common astrogliotic response to neuronal damage.


Subject(s)
Inclusion Bodies/pathology , Neuroglia/cytology , Spinocerebellar Ataxias/pathology , Adolescent , Brain/pathology , Cytoplasm , Humans , Male
7.
Mech Dev ; 108(1-2): 45-57, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11578860

ABSTRACT

The vascularization of the central nervous system occurs by angiogenic sprouting, a process in which different factors like vascular endothelial growth factor (VEGF) and angiopoietin (Ang)-1/2 must act in a coordinated fashion. We investigated how these factors participate in capillarization of the cerebellum, an area experiencing marked reorganization processes during its postnatal development. VEGF and Ang-1 mRNA were predominantly expressed by astrocytes, while Ang-2 mRNA was specifically induced at the tip of invading endothelial cell cords. Similar to the cerebral cortex, vascularization of the cerebellum occurred in an inside-out pattern, following closely the generation and differentiation of each cerebellar layer. VEGF and Ang-1/2 expression patterns shifted in a similar inside-out fashion, supporting their proposed function in vessel growth and maturation.


Subject(s)
Astrocytes/metabolism , Cerebellum/blood supply , Cerebellum/growth & development , Endothelial Growth Factors/genetics , Lymphokines/genetics , Membrane Glycoproteins/genetics , Proteins/genetics , Angiopoietin-1 , Angiopoietin-2 , Animals , Apoptosis , Astrocytes/cytology , Cell Division , Cerebellum/cytology , Endothelium, Vascular/cytology , Endothelium, Vascular/growth & development , Extracellular Matrix Proteins/genetics , Gene Expression Regulation, Developmental , Immunohistochemistry , In Situ Hybridization , Neovascularization, Physiologic/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptor Protein-Tyrosine Kinases/genetics , Receptor, TIE-2 , Receptors, Cell Surface/genetics , Receptors, Growth Factor/genetics , Receptors, TIE , Receptors, Vascular Endothelial Growth Factor , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
8.
Nat Med ; 7(5): 575-83, 2001 May.
Article in English | MEDLINE | ID: mdl-11329059

ABSTRACT

Vascular endothelial growth factor (VEGF) stimulates angiogenesis by activating VEGF receptor-2 (VEGFR-2). The role of its homolog, placental growth factor (PlGF), remains unknown. Both VEGF and PlGF bind to VEGF receptor-1 (VEGFR-1), but it is unknown whether VEGFR-1, which exists as a soluble or a membrane-bound type, is an inert decoy or a signaling receptor for PlGF during angiogenesis. Here, we report that embryonic angiogenesis in mice was not affected by deficiency of PlGF (Pgf-/-). VEGF-B, another ligand of VEGFR-1, did not rescue development in Pgf-/- mice. However, loss of PlGF impaired angiogenesis, plasma extravasation and collateral growth during ischemia, inflammation, wound healing and cancer. Transplantation of wild-type bone marrow rescued the impaired angiogenesis and collateral growth in Pgf-/- mice, indicating that PlGF might have contributed to vessel growth in the adult by mobilizing bone-marrow-derived cells. The synergism between PlGF and VEGF was specific, as PlGF deficiency impaired the response to VEGF, but not to bFGF or histamine. VEGFR-1 was activated by PlGF, given that anti-VEGFR-1 antibodies and a Src-kinase inhibitor blocked the endothelial response to PlGF or VEGF/PlGF. By upregulating PlGF and the signaling subtype of VEGFR-1, endothelial cells amplify their responsiveness to VEGF during the 'angiogenic switch' in many pathological disorders.


Subject(s)
Capillary Permeability , Endothelial Growth Factors/physiology , Lymphokines/physiology , Neoplasms, Experimental/blood supply , Neovascularization, Pathologic , Pregnancy Proteins/physiology , Animals , Base Sequence , DNA Primers , Embryonic and Fetal Development , Mice , Placenta Growth Factor , Plasma , Pregnancy Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors , Wound Healing/physiology
9.
Nat Genet ; 28(2): 131-8, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11381259

ABSTRACT

Hypoxia stimulates angiogenesis through the binding of hypoxia-inducible factors to the hypoxia-response element in the vascular endothelial growth factor (Vegf) promotor. Here, we report that deletion of the hypoxia-response element in the Vegf promotor reduced hypoxic Vegf expression in the spinal cord and caused adult-onset progressive motor neuron degeneration, reminiscent of amyotrophic lateral sclerosis. The neurodegeneration seemed to be due to reduced neural vascular perfusion. In addition, Vegf165 promoted survival of motor neurons during hypoxia through binding to Vegf receptor 2 and neuropilin 1. Acute ischemia is known to cause nonselective neuronal death. Our results indicate that chronic vascular insufficiency and, possibly, insufficient Vegf-dependent neuroprotection lead to the select degeneration of motor neurons.


Subject(s)
Cell Hypoxia/genetics , Endothelial Growth Factors/genetics , Lymphokines/genetics , Motor Neurons/pathology , Nerve Degeneration/genetics , Response Elements/genetics , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , Animals , Axons/physiology , Binding Sites , Electrophysiology , Endothelial Growth Factors/metabolism , Humans , Lymphokines/metabolism , Mice , Mice, Knockout , Motor Neurons/physiology , Muscle Contraction , Muscle Fibers, Skeletal/pathology , Muscular Atrophy/genetics , Muscular Atrophy/pathology , Nerve Degeneration/pathology , Nerve Degeneration/physiopathology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neuropilin-1 , Peripheral Nerves/pathology , Promoter Regions, Genetic , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Growth Factor/genetics , Receptors, Growth Factor/metabolism , Receptors, Vascular Endothelial Growth Factor , Sequence Deletion , Spinal Cord/physiology , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
10.
Cell Growth Differ ; 12(3): 137-45, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11306514

ABSTRACT

Mitogen-activated protein kinase (MAPK) signaling was examined in malignant melanoma cells exposed to hypoxia. Here we demonstrate that hypoxia induced a strong activation of the c-Jun NH2-terminal kinase (JNK), also termed stress-activated protein kinase (SAPK), in the melanoma cell line 530 in vitro. Other members of the MAPK family, e.g., extracellular signal-regulated kinase and p38, remained unaffected by the hypoxic stimulus. Activated JNK/SAPK could also be observed in the vicinity of hypoxic tumor areas in melanoma metastases as detected by immunohistochemistry. Functional analysis of JNK/SAPK activation in the melanoma cell line 530 revealed that activation of JNK/SAPK is involved in hypoxia-mediated tumor cell apoptosis. Both a dominant negative mutant of JNK/SAPK (SAPKbeta K-->R) and a dominant negative mutant of the immediate upstream activator of JNK/SAPK, SEK1 (SEK1 K-->R), inhibited hypoxia-induced apoptosis in transient transfection studies. In contrast, overexpression of the wild-type kinases had a slight proapoptotic effect. Inhibition of extracellular signal-regulated kinase and p38 pathways by the chemical inhibitors PD98058 and SB203580, respectively, had no effect on hypoxiainduced apoptosis. Under normoxic conditions, no influence on apoptosis regulation was observed after inhibition of all three MAPK pathways. In contrast to recent findings, JNK/SAPK activation did not correlate with Fas or Fas ligand (FasL) expression, suggesting that the Fas/FasL system is not involved in hypoxia-induced apoptosis in melanoma cells. Taken together, our data demonstrate that hypoxia-induced JNK/SAPK activation appears to play a critical role in apoptosis regulation of melanoma cells in vitro and in vivo, independent of the Fas/FasL system.


Subject(s)
Apoptosis/physiology , Hypoxia/enzymology , MAP Kinase Kinase 4 , Melanoma/enzymology , Mitogen-Activated Protein Kinases/metabolism , Neoplasm Invasiveness/physiopathology , Neovascularization, Pathologic/enzymology , Gene Expression Regulation, Neoplastic/physiology , Humans , Hypoxia/physiopathology , Immunohistochemistry , JNK Mitogen-Activated Protein Kinases , Melanoma/pathology , Melanoma/physiopathology , Mitogen-Activated Protein Kinase Kinases/metabolism , Necrosis , Neovascularization, Pathologic/physiopathology , Proto-Oncogene Proteins c-jun/metabolism , RNA, Messenger/metabolism , Transcription, Genetic/physiology , Transfection , Tumor Cells, Cultured/enzymology , Tumor Cells, Cultured/pathology , fas Receptor/genetics
12.
Int J Cancer ; 91(3): 273-82, 2001 Feb 01.
Article in English | MEDLINE | ID: mdl-11169947

ABSTRACT

Tumor growth is angiogenesis-dependent. Current evidence suggests that vascular endothelial growth factor (VEGF), a major regulator of embryonic and hypoxia-mediated angiogenesis, is necessary for tumor angiogenesis. VEGF is expressed in tumor cells in vivo, and its tyrosine kinase receptors VEGFR-1 and VEGFR-2 are up-regulated in the tumor endothelium. A second endothelial cell-specific ligand/receptor tyrosine kinase system, consisting of the tie2 receptor, its activating ligand angiopoietin-1 and the inhibitory ligand angiopoietin-2, has been characterized. We have examined 6 human primary breast-cancer samples and 4 murine breast-cancer cell lines (M6363, M6378, M6444, M6468), transplanted into nude mice, by in situ hybridization and/or Northern analysis. Expression of angiopoietin-1, angiopoietin-2 and tie2 was compared to VEGF and VEGFR-2 expression. Human tumors expressed VEGFR-2 and tie2 but varied considerably in VEGF and angiopoietin-1/-2 expression. In the murine tumor models, we observed high heterogeneity of receptor and ligand expression. M6363 and M6378 tumors were analyzed in detail because they showed different expression of components of the tie2/angiopoietin signaling system. M6363 tumors expressed VEGF, VEGFR-2 and angiopoietin-2 but not tie2 or angiopoietin-1, suggesting activation of VEGFR-2 and inhibition of tie2 signaling pathways, whereas M6378 tumors expressed VEGF, VEGFR-2, tie2 and angiopoietin-1 but little angiopoietin-2, suggesting activation of both VEGFR-2 and tie2 signaling pathways. In vivo studies using truncated dominant-negative tie2 and VEGFR-2 mutants revealed inhibition of M6363 tumor growth by 15% (truncated tie2) and 36% (truncated VEGFR-2), respectively. In contrast, M6378 tumor growth was inhibited by 57% (truncated tie2) and 47% (truncated VEGFR-2), respectively. These findings support the hypothesis that tumor angiogenesis is dependent on VEGFR-2 but suggest that, in addition, tie2-dependent pathways of tumor angiogenesis may exist. For adequate application of angiogenesis inhibitors in tumor patients, analysis of prevailing angiogenesis pathways may be a prerequisite.


Subject(s)
Adenocarcinoma, Mucinous/blood supply , Breast Neoplasms/blood supply , Carcinoma, Ductal, Breast/blood supply , Membrane Glycoproteins/metabolism , Neoplasm Proteins/metabolism , Neovascularization, Pathologic/metabolism , Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Growth Factor/metabolism , Adenocarcinoma, Mucinous/metabolism , Angiopoietin-1 , Angiopoietin-2 , Animals , Blotting, Northern , Breast Neoplasms/metabolism , Carcinoma, Ductal, Breast/metabolism , Female , Humans , In Situ Hybridization , Mice , Mice, Nude , RNA, Messenger/metabolism , Receptor, TIE-2 , Receptors, Vascular Endothelial Growth Factor , Signal Transduction , Tumor Cells, Cultured , Vascular Endothelial Growth Factor Receptor-1
13.
Oncogene ; 19(48): 5435-43, 2000 Nov 16.
Article in English | MEDLINE | ID: mdl-11114720

ABSTRACT

Hypoxia induces transcription of a range of physiologically important genes including erythropoietin and vascular endothelial growth factor. The transcriptional activation is mediated by the hypoxia-inducible factor-1 (HIF-1), a heterodimeric member of the basic helix-loop-helix PAS family, composed of alpha and beta subunits. HIF-1alpha shares 48 per cent identity with the recently identified HIF-2alpha protein that is also stimulated by hypoxia. In a previous study of hemangioblastomas, the most frequent manifestation of hereditary von Hippel-Lindau disease (VHL), we found elevated levels of vascular endothelial growth factor and HIF-2alpha mRNA in stromal cells of the tumors. Mutations of the VHL tumor suppressor gene are associated with a variety of tumors such as renal clear cell carcinomas (RCC). In this study, we analysed the expression of the hypoxia-inducible factors HIF-1alpha and HIF-2alpha in a range of VHL wildtype and VHL deficient RCC cell lines. In the presence of functional VHL protein, HIF-1alpha mRNA levels are elevated, whereas HIF-2alpha mRNA expression is increased only in cells lacking a functional VHL gene product. On the protein levels, however, in VHL deficient cell lines, both HIF-alpha subunits are constitutively expressed, whereas re-introduction of a functional VHL gene restores the instability of HIF-1alpha and HIF-2alpha proteins under normoxic conditions. Moreover, immunohistochemical analyses of RCCs and hemangioblastomas demonstrate up-regulation of HIF-1alpha and HIF-2alpha in the tumor cells. The data presented here provide evidence for a role of the VHL protein in regulation of angiogenesis and erythropoiesis mediated by the HIF-1alpha and HIF-2alpha proteins.


Subject(s)
Carcinoma, Renal Cell/metabolism , DNA-Binding Proteins/biosynthesis , Genes, Tumor Suppressor/physiology , Kidney Neoplasms/genetics , Ligases , Nuclear Proteins/biosynthesis , Oxygen/metabolism , Proteins/genetics , Trans-Activators/biosynthesis , Transcription Factors , Tumor Suppressor Proteins , Ubiquitin-Protein Ligases , Basic Helix-Loop-Helix Transcription Factors , Carcinoma, Renal Cell/genetics , Cerebellum/metabolism , Cerebellum/physiology , DNA-Binding Proteins/genetics , Endothelial Growth Factors/biosynthesis , Endothelial Growth Factors/genetics , Glucose Transporter Type 1 , Hemangioblastoma/genetics , Hemangioblastoma/metabolism , Humans , Hypoxia-Inducible Factor 1 , Hypoxia-Inducible Factor 1, alpha Subunit , Immunohistochemistry , Kidney Neoplasms/metabolism , Lymphokines/biosynthesis , Lymphokines/genetics , Monosaccharide Transport Proteins/biosynthesis , Monosaccharide Transport Proteins/genetics , Mutation , Nuclear Proteins/genetics , Polymerase Chain Reaction , RNA, Messenger/genetics , RNA, Messenger/metabolism , Trans-Activators/genetics , Tumor Cells, Cultured , Up-Regulation , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors , Von Hippel-Lindau Tumor Suppressor Protein , von Hippel-Lindau Disease/genetics
14.
Am J Pathol ; 157(5): 1473-83, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11073808

ABSTRACT

Vascular endothelial growth factor (VEGF), a key regulator of vasculogenesis and embryonic angiogenesis, was recently found to be up-regulated in an animal model of stroke. Unlike VEGF, angiopoietin (Ang)-1 and -2, their receptor tie-2, and the associated receptor tie-1 exert their functions at later stages of vascular development, i.e., during vascular remodeling and maturation. To assess the role of the angiopoietin/tie family in ischemia-triggered angiogenesis we analyzed their temporal and spatial expression pattern after middle cerebral artery occlusion (MCAO) using in situ hybridization and immunohistochemistry. Ang-1 mRNA was constitutively expressed in a subset of glial and neuronal cells with no apparent change in expression after MCAO. Ang-2 mRNA was up-regulated 6 hours after MCAO and was mainly observed in endothelial cell (EC) cord tips in the peri-infarct and infarct area. Up-regulation of both Ang-2 and VEGF coincided with EC proliferation. Interestingly, EC proliferation was preceded by a transient period of EC apoptosis, correlating with a change in VEGF/Ang-2 balance. Our observation of specific stages of vascular regression and growth after MCAO are in agreement with recent findings suggesting a dual role of Ang-2 in blood vessel formation, depending on the availability of VEGF.


Subject(s)
Arterial Occlusive Diseases/metabolism , Cerebral Arteries , Membrane Glycoproteins/metabolism , Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Cell Surface/metabolism , Angiopoietin-1 , Angiopoietin-2 , Animals , Apoptosis , Arterial Occlusive Diseases/physiopathology , Blood Vessels/physiopathology , Cell Division , Endothelium, Vascular/pathology , Endothelium, Vascular/physiopathology , Immunohistochemistry , In Situ Hybridization , Male , Membrane Glycoproteins/genetics , Proteins/genetics , RNA, Messenger/metabolism , Rats , Rats, Inbred F344 , Receptor Protein-Tyrosine Kinases/genetics , Receptor, TIE-1 , Receptor, TIE-2 , Receptors, Cell Surface/genetics , Receptors, TIE
15.
Neurosurgery ; 47(4): 921-9; discussion 929-30, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11014432

ABSTRACT

OBJECTIVE: The goal of the present study was to develop an orthotopic in vivo model for the investigation of vascular endothelial growth factor (VEGF)-dependent glioma growth and vascularization. METHODS: C6 glioma cells were infected with viruses encoding sense or antisense VEGF. Expression of the transgene was controlled by Northern blot analysis, Western blot analysis, and immunohistochemistry. Spheroids generated from both clones as well as from wild-type and mock-transfected cells were implanted in the brains of Sprague-Dawley rats. Growth and vascularization were assessed using magnetic resonance imaging after 7 and 11 days. Histology was studied using hematoxylin and eosin staining, immunohistochemistry with anti-von Willebrand staining, anti-VEGF, anti-CD8, and assessment of vessel density. RESULTS: Cell proliferation, migration, and invasion in vitro were very similar in all cell clones. Sense gliomas demonstrated by far the fastest growth in vivo, with intense contrast enhancement meeting criteria for highly malignant tumors. Histological examination revealed masses of von Willebrand- and VEGF-positive tumor vessels with a high vessel density. Antisense gliomas depicted the radiological features of low-grade gliomas, with slow growth and poor vascularization, although they were highly infiltrative. Wild-type and mock-transfected gliomas demonstrated similar growth and vascularization patterns intermediate between sense and antisense gliomas. Any influence of the allogeneic response of the hosts on different tumor sizes could be excluded. CONCLUSION: Our model elucidates glioma growth and vascularization as strongly VEGF dependent, which is consistent with human gliomas. Thus, this model is suitable for testing antiangiogenic strategies to interfere with the VEGF/VEGF receptor system, as well as for exploring VEGF-independent mechanisms using the antisense-transfected clone.


Subject(s)
Brain Neoplasms/blood supply , Brain Neoplasms/pathology , Endothelial Growth Factors/physiology , Glioma/blood supply , Glioma/pathology , Lymphokines/physiology , Animals , Blood Vessels/pathology , Brain Neoplasms/physiopathology , CD8 Antigens/metabolism , Cell Division/physiology , Cell Movement , Glioma/diagnosis , Glioma/physiopathology , Immunohistochemistry , Magnetic Resonance Imaging , Neoplasm Invasiveness/diagnosis , Neoplasm Transplantation , Rats , Rats, Sprague-Dawley , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors , von Willebrand Factor/metabolism
16.
Hum Mol Genet ; 9(9): 1351-5, 2000 May 22.
Article in English | MEDLINE | ID: mdl-10814716

ABSTRACT

Hyperkeratotic capillary-venous malformations (HCCVMs) are rare cutaneous lesions that occur in a small subgroup of patients with cerebral capillary malformation (CCM). CCMs cause neurological problems that range from headaches to life-threatening intracranial bleeding. CCMs and HCCVMs have a similar histopathological appearance of dilated capillary-venous channels. Genetic linkage of inherited CCMs has been established to three chromosomal loci, 3q25. 2-27, 7p13-15 and 7q21-22. The first mutations were identified in the CCM1 gene (located on 7q21-22), which encodes KRIT1 protein (KREV1 interaction trapped 1), presumably a membrane-bound protein with signalling activity. Although KRIT1 is known to interact with KREV1/RAP1A, a Ras-family GTPase, the exact function of KRIT1 in the formation of cerebral capillaries and veins is poorly understood. In this study, we screened five families with CCM for mutations in the KRIT1 gene. In one of the families, CCMs co-segregated with HCCVMs. We identified a KRIT1Delta(G103)mutation in this family, suggesting that this rare form of the condition is also caused by mutations in the CCM1 gene and that KRIT1 is probably important for cutaneous vasculature. Interestingly, this deletion introduces the earliest stop codon among identified mutations, suggesting a possible correlation between the molecular alteration and the cutaneous phenotype. Another novel mutation, KRIT1(IVS2+2(T-->C)), was found in a family with only cerebral capillary-venous malformations.


Subject(s)
Brain/blood supply , Capillaries/abnormalities , Microtubule-Associated Proteins , Mutation , Proto-Oncogene Proteins/genetics , Codon , DNA Mutational Analysis , Family Health , Female , Genetic Linkage , Humans , Hyperkeratosis, Epidermolytic/diagnosis , Hyperkeratosis, Epidermolytic/genetics , KRIT1 Protein , Male , Pedigree , Phenotype
17.
Cancer Res ; 60(5): 1388-93, 2000 Mar 01.
Article in English | MEDLINE | ID: mdl-10728704

ABSTRACT

Microvessel density (MVD) counting techniques have been widely used to assess the vasculature in tumors. MVD counts assess the presence of blood vessels but do not give an indication of the degree of angiogenesis and the functional status of the tumor neovasculature. To analyze angiogenesis and the functional status of the tumor vascular bed, we have quantitated endothelial cell proliferation and the recruitment of pericytes in human tumors [glioblastomas (n = 30), renal cell carcinomas (n = 22), colon carcinomas (n = 18), mammary carcinomas (n = 24), lung carcinomas (n = 15), and prostate carcinomas (n = 19)]. These findings were compared to the physiological angiogenesis in the cyclic bovine ovarian corpus luteum. Tissue sections were examined applying double-labeling immunohistochemical techniques to detect proliferating endothelial cells and to colocalize endothelial cells and pericytes. The following parameters were quantitated: (a) MVD count; (b) proliferating capillary index (PCI); (c) proliferating tumor versus endothelial cell index; and (d) microvessel pericyte coverage index (MPI). Based on endothelial cell proliferation, angiogenesis was found to be present in all tumors with characteristic and significant differences between the tumor types (glioblastomas, PCI = 9.6 +/- 6.1%; renal cell carcinomas, PCI = 9.4 +/- 5.2%; colon carcinomas, PCI = 7.8 +/- 5.2%; mammary carcinomas, PCI = 5.0 +/- 4.8%; lung carcinomas, PCI = 2.6 +/- 2.5%; prostate carcinomas, PCI = 2.0 +/- 1.4%). There was a considerable degree of heterogeneity in the intensity of angiogenesis within each tumor group, as indicated by large standard deviations. Even in the most angiogenic tumors, angiogenesis was found to be 4 to 20 times less intense as compared with the physiological angiogenesis in the growing ovarian corpus rubrum (PCI = 40.6 +/- 6.2%). Varying degrees of pericyte recruitment to the tumor microvasculature were determined in the different tumor types (glioblastomas, MPI = 12.7 +/- 7.9%; renal cell carcinomas, MPI = 17.9 +/- 7.8%; colon carcinomas, MPI = 65.4 +/- 10.5%; mammary carcinomas, MPI = 67.3 +/- 14.2%; lung carcinomas, MPI = 40.8 +/- 14.5%; prostate carcinomas, MPI = 29.6 +/- 9.5%). The data demonstrate distinct quantitative variations in the intensity of angiogenesis in malignant human tumors. Furthermore, the varying degrees of pericyte recruitment indicate differences in the functional status of the tumor vasculature in different tumors that may reflect varying degrees of maturation of the tumor vascular bed.


Subject(s)
Neoplasms/blood supply , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology , Animals , Antineoplastic Agents/therapeutic use , Cattle , Cell Differentiation , Cell Division , Endothelium, Vascular/pathology , Humans , Neoplasms/drug therapy , Neoplasms/pathology , Pericytes/pathology
18.
J Neurooncol ; 50(1-2): 109-20, 2000.
Article in English | MEDLINE | ID: mdl-11245271

ABSTRACT

Vascular endothelial growth factor (VEGF) is a regulator of angiogenesis, vasculogenesis and vascular permeability. In this contribution, molecular and biological properties of VEGF are described. Furthermore, this article focuses on the evidence that angiogenesis in brain tumors is mediated by VEGF. Among the topics discussed are expression patterns of VEGF and its receptors in different brain tumors, possible regulatory mechanism involved in the VEGF-driven tumor angiogenesis and the involvement of VEGF in the genesis of peritumoral edema. Finally, anti-angiogenesis approaches to target VEGF/VEGF receptors are discussed.


Subject(s)
Brain Neoplasms/blood supply , Endothelial Growth Factors/physiology , Lymphokines/physiology , Neoplasm Proteins/physiology , Neovascularization, Pathologic/physiopathology , Receptor Protein-Tyrosine Kinases/physiology , Receptors, Growth Factor/physiology , Angiogenesis Inhibitors/pharmacology , Angiogenesis Inhibitors/therapeutic use , Animals , Brain Edema/etiology , Brain Neoplasms/complications , Brain Neoplasms/pathology , Capillary Leak Syndrome , Endothelial Growth Factors/antagonists & inhibitors , Endothelium, Vascular/pathology , Gene Expression Regulation, Neoplastic , Glioma/blood supply , Glioma/complications , Glioma/pathology , Hemangioblastoma/blood supply , Hemangioblastoma/complications , Hemangioblastoma/pathology , Humans , Lymphokines/antagonists & inhibitors , Meningeal Neoplasms/blood supply , Meningeal Neoplasms/complications , Meningeal Neoplasms/pathology , Meningioma/blood supply , Meningioma/complications , Meningioma/pathology , Mice , Models, Biological , Neoplasm Proteins/antagonists & inhibitors , Neovascularization, Pathologic/drug therapy , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/physiology , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptors, Growth Factor/antagonists & inhibitors , Receptors, Vascular Endothelial Growth Factor , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
19.
Int J Dev Neurosci ; 17(5-6): 579-91, 1999.
Article in English | MEDLINE | ID: mdl-10571419

ABSTRACT

With increasing size tumors are continually dependent on a functional blood vessel system to guarantee the supply with oxygen and nutrients. Vascular endothelial growth factor (VEGF) is a key mediator not only of developmental but also of hypoxia-mediated and tumor-induced angiogenesis. Gene therapy using antisense VEGF with the aim to inhibit tumor angiogenesis may be a successful strategy for the treatment of highly vascular and invasive malignant gliomas. We investigated whether retrovirus producer cells encoding antisense VEGF can be used for in vivo gene transfer. The full length mouse VEGF164 cDNA was cloned in a sense and antisense direction into the retroviral expression vector pLEN. pLEN-VEGF (sense) and pLEN-FGEV (antisense) expression vectors were used to transfect the packaging cell line GP + E86 and to establish ecotropic virus producer cell lines. GP + E86:LEN-FGEV (#5) cells showed high expression of antisense VEGF mRNA, whereas GP+ E86:LEN-VEGF (#8) showed high expression of sense VEGF mRNA and active VEGF protein. Co-implantation of GS-9L cells with retrovirus producing cells containing the antisense VEGF construct into the brains of syngeneic rats showed a statistically significant inhibition of tumor growth and prolongation of survival time, while co-implantation of retrovirus producer cells containing the sense VEGF expression vector resulted in an increasing tumor growth and reduced survival time of the rats compared to control animals. Histological analysis of the tumors co-implanted with GP + E86:LEN-FGEV (#5) cells showed the suppression of angiogenesis, high degree of necrosis and no evidence of a significant immune response. Expression of antisense VEGF mRNA in these tumors was confirmed by in situ hybridization analysis. This is the first report demonstrating the potential utility of virus producer cells as in vivo gene transfer vehicles for antisense VEGF gene therapy of malignant gliomas.


Subject(s)
Antisense Elements (Genetics) , Brain Neoplasms/therapy , Genetic Therapy , Glioma/therapy , Retroviridae/genetics , Virus Replication , Animals , Blotting, Northern , Blotting, Western , Cell Line , Genetic Code , Mice , Rats , Survival Rate , Toxicity Tests
20.
J Pathol ; 189(1): 66-71, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10451490

ABSTRACT

It has previously been suggested that in human brain tumours, endothelial cell proliferation during angiogenesis is regulated by a paracrine mechanism involving vascular endothelial growth factor (VEGF) and its receptors (VEGF receptor 1 and VEGF receptor 2). The mechanism of growth factor up-regulation is based on hypoxic activation of mRNA expression and mRNA stabilization and genetic events, leading to an increase of growth factor gene expression. The role of the other newly discovered VEGF family members with a high specificity for endothelial cells in the pathogenesis of glial neoplasms is unknown. To investigate which other members of the VEGF family are overexpressed in human brain tumours, the mRNA levels of placenta growth factor (PlGF), VEGF-A, and VEGF-B genes were determined by northern blot analysis in surgically obtained human meningiomas. In the 16 meningiomas examined, the mRNA for PlGF was highly expressed in four tumours and VEGF-A mRNA was highly abundant in three tumour samples. There was no close correlation between PlGF mRNA levels and VEGF-A expression levels. VEGF-B mRNA was abundantly expressed in all tumour samples at uniform levels. In a PlGF-positive tumour sample, immunoreactive VEGFR-1 and VEGFR-2 were detected in endothelial cells of the blood vessels. PlGF protein was detectable in most but not all capillaries of the tumour. PlGF is thus highly up-regulated in a subset of human meningiomas and may therefore have functions, in some tumour vessels, connected to endothelial cell maturation and tube formation. These findings suggest that PlGF, in addition to VEGF-A, may be another positive factor in tumour angiogenesis in human meningiomas.


Subject(s)
Meningeal Neoplasms/chemistry , Meningioma/chemistry , Neoplasm Proteins/analysis , Pregnancy Proteins/analysis , RNA, Messenger/analysis , Adult , Aged , Aged, 80 and over , Blotting, Northern , Child , Endothelial Growth Factors/analysis , Female , Humans , Immunoblotting , Immunohistochemistry , Lymphokines/analysis , Male , Meningeal Neoplasms/pathology , Meningioma/pathology , Middle Aged , Neoplasm Proteins/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Placenta Growth Factor , Platelet Endothelial Cell Adhesion Molecule-1/analysis , Pregnancy Proteins/genetics , Receptor Protein-Tyrosine Kinases/analysis , Receptors, Growth Factor/analysis , Receptors, Vascular Endothelial Growth Factor , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factor B , Vascular Endothelial Growth Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...