Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Cancer Res ; 75(21): 4504-16, 2015 Nov 01.
Article in English | MEDLINE | ID: mdl-26363010

ABSTRACT

Glioblastoma is an aggressive brain tumor characterized by an abnormal blood vasculature that is hyperpermeable. Here, we report a novel role for CD93 in regulating angiogenesis in this setting by modulating cell-cell and cell-matrix adhesion of endothelial cells. Tissue microarray analysis demonstrated that vascular expression of CD93 was correlated with poor survival in a clinical cohort of patients with high-grade astrocytic glioma. Similarly, intracranial growth in the GL261 mouse model of glioma was delayed significantly in CD93(-/-) hosts, resulting in improved survival compared with wild-type mice. This effect was associated with increased vascular permeability and decreased vascular perfusion of tumors, indicating reduced vessel functionality in the absence of CD93. RNAi-mediated attenuation of CD93 in endothelial cells diminished VEGF-induced tube formation in a three-dimensional collagen gel. CD93 was required for efficient endothelial cell migration and proper cell polarization in vitro. Further, in endothelial cells where CD93 was attenuated, decreased cell spreading led to a severe reduction in cell adhesion, a lack of proper cell contacts, a loss of VE-cadherin, and aberrant actin stress fiber formation. Our results identify CD93 as a key regulator of glioma angiogenesis and vascular function, acting via cytoskeletal rearrangements required for cell-cell and cell-matrix adhesion.


Subject(s)
Brain Neoplasms/blood supply , Endothelium, Vascular/metabolism , Glioblastoma/blood supply , Membrane Glycoproteins/metabolism , Neovascularization, Pathologic/genetics , Receptors, Complement/metabolism , Actin Cytoskeleton/pathology , Animals , Antigens, CD/genetics , Brain Neoplasms/mortality , Brain Neoplasms/pathology , Cadherins/genetics , Capillary Permeability/genetics , Cell Adhesion/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Endothelial Cells/metabolism , Glioblastoma/mortality , Glioblastoma/pathology , Humans , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA Interference , RNA, Small Interfering , Receptors, Complement/genetics , Stress Fibers/metabolism , Vascular Endothelial Growth Factor A/metabolism
2.
FASEB J ; 29(1): 227-38, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25361735

ABSTRACT

Antiangiogenic treatment targeting the vascular endothelial growth factor (VEGF) signaling pathway is in clinical use, but its effect on vascular function and the tumor microenvironment is poorly understood. Here, we investigate cross-talk between VEGF and proinflammatory TNF-α signaling in endothelial cells and its impact on leukocyte recruitment. We found that cotreatment with VEGF decreased TNF-α-induced Jurkat cell adhesion to human microvascular endothelial cells by 40%. This was associated with inhibition of TNF-α-mediated regulation of 86 genes, including 2 T-lymphocyte-attracting chemokines, CXCL10 and CXCL11 [TNF-α concentration 1 ng/ml; 50% inhibition/inhibitory concentration (IC50) VEGF, 3 ng/ml]. Notably, VEGF directly suppressed TNF-α-induced gene expression through negative cross-talk with the NF-κB-signaling pathway, leading to an early decrease in IFN regulatory factor 1 (IRF-1) expression and reduced phosphorylation of signal transducer and activator of transcription 1 (p-Stat1) at later times. Inhibition of VEGF signaling in B16 melanoma tumor-bearing mice by sunitinib treatment resulted in up-regulation of CXCL10 and CXCL11 in tumor vessels, accompanied by up to 18-fold increased infiltration of CD3(+) T-lymphocytes in B16 tumors. Our results demonstrate a novel role of VEGF in negative regulation of NF-κB signaling and endothelial activation in the tumor microenvironment and provide evidence that pharmacological inhibition of VEGF signaling enhances T-lymphocyte recruitment through up-regulation of chemokines CXCL10 and CXCL11.


Subject(s)
Lymphocytes, Tumor-Infiltrating/metabolism , Lymphocytes, Tumor-Infiltrating/pathology , NF-kappa B/metabolism , Tumor Microenvironment/physiology , Vascular Endothelial Growth Factor A/metabolism , Animals , Cell Adhesion , Chemokine CXCL10/metabolism , Chemokine CXCL11/metabolism , Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Jurkat Cells , Male , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Microfluidic Analytical Techniques , Signal Transduction , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , Tumor Necrosis Factor-alpha/metabolism
3.
Cell Tissue Res ; 354(3): 647-69, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24072341

ABSTRACT

Many studies on the molecular control underlying normal cell behavior and cellular responses to disease stimuli and pharmacological intervention are conducted in single-cell culture systems, while the read-out of cellular engagement in disease and responsiveness to drugs in vivo is often based on overall tissue responses. As the majority of drugs under development aim to specifically interact with molecular targets in subsets of cells in complex tissues, this approach poses a major experimental discrepancy that prevents successful development of new therapeutics. In this review, we address the shortcomings of the use of artificial (single) cell systems and of whole tissue analyses in creating a better understanding of cell engagement in disease and of the true effects of drugs. We focus on microvascular endothelial cells that actively engage in a wide range of physiological and pathological processes. We propose a new strategy in which in vivo molecular control of cells is studied directly in the diseased endothelium instead of at a (far) distance from the site where drugs have to act, thereby accounting for tissue-controlled cell responses. The strategy uses laser microdissection-based enrichment of microvascular endothelium which, when combined with transcriptome and (phospho)proteome analyses, provides a factual view on their status in their complex microenvironment. Combining this with miniaturized sample handling using microfluidic devices enables handling the minute sample input that results from this strategy. The multidisciplinary approach proposed will enable compartmentalized analysis of cell behavior and drug effects in complex tissue to become widely implemented in daily biomedical research and drug development practice.


Subject(s)
Cell Culture Techniques/methods , Drug Evaluation, Preclinical/methods , Endothelial Cells/cytology , Endothelial Cells/drug effects , Single-Cell Analysis/methods , Animals , Humans , Pharmacology/methods
4.
J Pathol ; 228(3): 378-90, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22786655

ABSTRACT

Glioblastoma are aggressive astrocytic brain tumours characterized by microvascular proliferation and an abnormal vasculature, giving rise to brain oedema and increased patient morbidity. Here, we have characterized the transcriptome of tumour-associated blood vessels and describe a gene signature clearly associated with pleomorphic, pathologically altered vessels in human glioblastoma (grade IV glioma). We identified 95 genes differentially expressed in glioblastoma vessels, while no significant differences in gene expression were detected between vessels in non-malignant brain and grade II glioma. Differential vascular expression of ANGPT2, CD93, ESM1, ELTD1, FILIP1L and TENC1 in human glioblastoma was validated by immunohistochemistry, using a tissue microarray. Through qPCR analysis of gene induction in primary endothelial cells, we provide evidence that increased VEGF-A and TGFß2 signalling in the tumour microenvironment is sufficient to invoke many of the changes in gene expression noted in glioblastoma vessels. Notably, we found an enrichment of Smad target genes within the distinct gene signature of glioblastoma vessels and a significant increase of Smad signalling complexes in the vasculature of human glioblastoma in situ. This indicates a key role of TGFß signalling in regulating vascular phenotype and suggests that, in addition to VEGF-A, TGFß2 may represent a new target for vascular normalization therapy.


Subject(s)
Blood Vessels/physiopathology , Brain Neoplasms/physiopathology , Gene Expression Profiling , Glioblastoma/physiopathology , Transforming Growth Factor beta2/physiology , Vascular Endothelial Growth Factor A/physiology , Adult , Aged , Brain Neoplasms/blood supply , Brain Neoplasms/pathology , Case-Control Studies , Endothelium, Vascular/pathology , Endothelium, Vascular/physiopathology , Gene Expression Regulation, Neoplastic/physiology , Glioblastoma/blood supply , Glioblastoma/pathology , Humans , Laser Capture Microdissection , Microarray Analysis , Middle Aged , Neoplasm Grading , Pericytes/pathology , Pericytes/physiology , Signal Transduction/physiology , Transforming Growth Factor beta2/genetics , Vascular Endothelial Growth Factor A/genetics
5.
Anticancer Drugs ; 23(2): 161-72, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22075979

ABSTRACT

The precise molecular effects that antiangiogenic drugs exert on tumor vasculature remain to be poorly understood. We therefore set out to investigate the molecular and architectural changes that occur in the vasculature of two different tumor types that both respond to vascular endothelial growth factor receptor 2 (VEGFR2) inhibitor therapy. Mice bearing Lewis lung carcinoma (LLC) or B16.F10 melanoma were treated with vandetanib (ZD6474), a VEGFR2/epidermal growth factor receptor (EGFR)/REarranged during Transfection (RET) kinase inhibitor, resulting in a significant 80% reduction in tumor outgrowth. Although in LLC the vascular density was not affected by vandetanib treatment, it was significantly decreased in B16.F10. In LLC, vandetanib treatment induced a shift in vascular gene expression toward stabilization, as demonstrated by upregulation of Tie2 and N-cadherin and downregulation of Ang2 and integrin ß3. In contrast, only eNOS and P-selectin responded to vandetanib treatment in B16.F10 vasculature. Strikingly, vandetanib reduced protein expression of VEGFR2 in both models, whereas mRNA remained unaffected. Analysis of miR-296 expression allowed us to exclude a role for the recently proposed microRNA-296 in VEGFR2 posttranslational control in LLC and B16.F10 in vivo. Our data demonstrate that VEGFR2/EGFR inhibition through vandetanib slows down both LLC and B16.F10 tumor growth. Yet, the underlying molecular changes in the vasculature that orchestrate the antitumor effect differ between tumor types. Importantly, in both models, vandetanib treatment induced loss of its pharmacological target, which was not directly related to miR-296 expression. Validation of our observations in tumor biopsies from VEGFR2 inhibitor-treated patients will be essential to unravel the effects of VEGFR2 inhibitor therapy on tumor vasculature in relation to therapeutic efficacy.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Carcinoma, Lewis Lung/drug therapy , Melanoma, Experimental/drug therapy , MicroRNAs/physiology , Neovascularization, Pathologic/drug therapy , Piperidines/therapeutic use , Quinazolines/therapeutic use , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Angiogenesis Inhibitors/administration & dosage , Angiogenesis Inhibitors/pharmacology , Animals , Carcinoma, Lewis Lung/blood supply , Carcinoma, Lewis Lung/genetics , Carcinoma, Lewis Lung/pathology , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/drug effects , Immunohistochemistry , Injections, Intraperitoneal , Male , Melanoma, Experimental/blood supply , Melanoma, Experimental/genetics , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Piperidines/administration & dosage , Piperidines/pharmacology , Quinazolines/administration & dosage , Quinazolines/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Vascular Endothelial Growth Factor Receptor-2/biosynthesis , Xenograft Model Antitumor Assays
6.
ISRN Oncol ; 2011: 409308, 2011.
Article in English | MEDLINE | ID: mdl-22235379

ABSTRACT

In established tumors, angiogenic endothelial cells (ECs) coexist next to "quiescent" EC in matured vessels. We hypothesized that angio-gene expression of B16.F10 melanoma would differ depending on the growth stage. Unraveling the spatiotemporal nature thereof is essential for drug regimen design aimed to affect multiple neovascularization stages. We determined the angiogenic phenotype-represented by 52 angio-genes-and vascular morphology of small, intermediate, and large s.c. growing mouse B16.F10 tumors and demonstrated that expression of these genes did not differ between the different growth stages. Yet vascular morphology changed dramatically from small vessels without lumen in small to larger vessels with increased lumen size in intermediate/large tumors. Separate analysis of these vascular morphologies revealed a significant difference in αSMA expression in relation to vessel morphology, while no relation with VEGF, HIF-1α, nor Dll4 expression levels was observed. We conclude that the tumor vasculature remains actively engaged in angiogenesis during B16.F10 melanoma outgrowth and that the major change in tumor vascular morphology does not follow molecular concepts generated in other angiogenesis models.

7.
Cell Tissue Res ; 335(1): 205-22, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18677515

ABSTRACT

Microvascular endothelial cells display a large degree of heterogeneity in function depending on their location in the vascular tree. The existence of organ-specific, microvascular-bed-specific, and even intravascular variations in endothelial cell gene expression emphasizes their high cell-to-cell variability, which is furthermore extremely adaptable to altering conditions. The ability of microvascular endothelial cells to respond dynamically to pathology-related microenvironmental changes is particularly apparent in tumor-growth-associated angiogenesis. An understanding of how they behave, how their behavior varies between and within tumors, and how their behavior is related to responsiveness to drugs is critical for the development of effective anti-angiogenic treatment strategies. In this review, we introduce some general issues concerning organ-imprinted microvascular heterogeneity and highlight the importance of studying microvascular endothelial cell behavior in an in vivo context. This is followed by an overview of state-of-the-art knowledge regarding the nature of the variation in microenvironmental conditions in pre-clinical and clinical tumors and their consequences for tumor endothelial behavior. We provide recent insights into the in vivo molecular activation status of the endothelium and, finally, outline our current understanding of the way that anti-angiogenic drugs affect tumor endothelial cells in relation to their anti-tumor effects.


Subject(s)
Endothelial Cells/metabolism , Gene Expression Regulation , Neoplasms/blood supply , Neoplasms/metabolism , Neovascularization, Pathologic/metabolism , Angiogenesis Inhibitors/therapeutic use , Animals , Endothelial Cells/pathology , Gene Expression Regulation/drug effects , Humans , Microvessels/metabolism , Microvessels/pathology , Neoplasms/drug therapy , Neoplasms/pathology , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology , Organ Specificity
8.
Cancer Res ; 67(9): 4138-48, 2007 May 01.
Article in English | MEDLINE | ID: mdl-17483324

ABSTRACT

Tumor angiogenesis requires intricate regulation of gene expression in endothelial cells. We recently showed that DNA methyltransferase (DNMT) and histone deacetylase (HDAC) inhibitors directly repress endothelial cell growth and tumor angiogenesis, suggesting that epigenetic modifications mediated by DNMTs and HDAC are involved in regulation of endothelial cell gene expression during tumor angiogenesis. To understand the mechanisms behind the epigenetic regulation of tumor angiogenesis, we used microarray analysis to perform a comprehensive screen to identify genes down-regulated in tumor-conditioned versus quiescent endothelial cells, and reexpressed by 5-aza-2'-deoxycytidine (DAC) and trichostatin A (TSA). Among the 81 genes identified, 77% harbored a promoter CpG island. Validation of mRNA levels of a subset of genes confirmed significant down-regulation in tumor-conditioned endothelial cells and reactivation by treatment with a combination of DAC and TSA, as well as by both compounds separately. Silencing of these genes in tumor-conditioned endothelial cells correlated with promoter histone H3 deacetylation and loss of H3 lysine 4 methylation, but did not involve DNA methylation of promoter CpG islands. For six genes, down-regulation in microdissected human tumor endothelium was confirmed. Functional validation by RNA interference revealed that clusterin, fibrillin 1, and quiescin Q6 are negative regulators of endothelial cell growth and angiogenesis. In summary, our data identify novel angiogenesis-suppressing genes that become silenced in tumor-conditioned endothelial cells in association with promoter histone modifications and reactivated by DNMT and HDAC inhibitors through reversal of these epigenetic modifications, providing a mechanism for epigenetic regulation of tumor angiogenesis.


Subject(s)
Colonic Neoplasms/blood supply , Endothelial Cells/physiology , Gene Silencing , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Caco-2 Cells , Clusterin/genetics , Colonic Neoplasms/genetics , DNA Methylation , DNA Modification Methylases/antagonists & inhibitors , Decitabine , Endothelial Cells/drug effects , Endothelial Cells/pathology , Epigenesis, Genetic , Fibrillin-1 , Fibrillins , Gene Expression Regulation, Neoplastic/drug effects , Histone Deacetylase Inhibitors , Humans , Hydroxamic Acids/pharmacology , Microfilament Proteins/genetics , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Oxidoreductases Acting on Sulfur Group Donors , Promoter Regions, Genetic , Reverse Transcriptase Polymerase Chain Reaction , Thioredoxins/genetics
9.
Drug Metab Dispos ; 34(4): 591-9, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16415126

ABSTRACT

Tissue slices have been shown to be a valuable tool to predict metabolism of novel drugs. However, besides the numerous advantages of their use for this purpose, some potential drawbacks also exist, including reported poor penetration of drugs into the inner cell layers of slices and loss of metabolic capacity during prolonged incubation, leading to underprediction of metabolic clearance. In the present study, we empirically identified (and quantified) sources of underprediction using rat tissue slices of lung, intestine, kidney, and liver and found that thin liver slices (+/-100 mum) metabolized model substrates (7-hydroxycoumarin, testosterone, warfarin, 7-ethoxycoumarin, midazolam, haloperidol, and quinidine) as rapidly as isolated hepatocytes. Furthermore, it was found that organ slices remain metabolically active for sufficient periods of incubation, enabling study of the kinetics of low clearance compounds. In addition, we determined the influence of albumin on the clearance prediction of six model substrates. For three of these substrates, the intrinsic clearance in the presence of albumin was approximately 3 times higher than that obtained from incubations without albumin, but corrected for unbound fraction. This resulted in a much more accurate prediction of in vivo whole body metabolic clearance for these compounds. Collectively, these results show that drawbacks of the use of slices for clearance prediction are largely surmountable. Provided that thin liver slices and physiological albumin concentration are used, whole body metabolic clearance is predicted with acceptable (2-fold) accuracy with organ slices. These results emphasize the applicability of organ slices in this field of research.


Subject(s)
Coumarins/metabolism , Serum Albumin, Bovine/metabolism , Testosterone/metabolism , Umbelliferones/metabolism , Animals , Drug Evaluation, Preclinical , Intestinal Mucosa/metabolism , Kidney/metabolism , Kinetics , Liver/metabolism , Male , Protein Binding , Rats , Rats, Sprague-Dawley , Rats, Wistar , Reproducibility of Results , Tissue Culture Techniques
SELECTION OF CITATIONS
SEARCH DETAIL
...