Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
FASEB J ; 29(8): 3411-25, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25911611

ABSTRACT

De novo formation of blood vessels is a pivotal mechanism during cancer development. During the past few years, antiangiogenic drugs have been developed to target tumor vasculature. However, because of limitations and adverse effects observed with current therapies, there is a strong need for alternative antiangiogenic strategies. Using specific anti-junctional adhesion molecule (JAM)-B antibodies and Jam-b-deficient mice, we studied the role in antiangiogenesis of JAM-B. We found that antibodies against murine JAM-B, an endothelium-specific adhesion molecule, inhibited microvessel outgrowth from ex vivo aortic rings and in vitro endothelial network formation. In addition, anti-JAM-B antibodies blocked VEGF signaling, an essential pathway for angiogenesis. Moreover, increased aortic ring branching was observed in aortas isolated from Jam-b-deficient animals, suggesting that JAM-B negatively regulates proangiogenic pathways. In mice, JAM-B expression was detected in de novo-formed blood vessels of tumors, but anti-JAM-B antibodies unexpectedly did not reduce tumor growth. Accordingly, JAM-B deficiency in vivo had no impact on blood vessel formation, suggesting that targeting JAM-B in vivo may be offset by other proangiogenic mechanisms. In conclusion, despite the promising effects observed in vitro, targeting JAM-B during tumor progression seems to be inefficient as a stand-alone antiangiogenesis therapy.


Subject(s)
Cell Adhesion Molecules/metabolism , Neovascularization, Pathologic/metabolism , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Animals , Cell Line, Tumor , Female , Mice , Mice, Inbred C57BL , Signal Transduction/physiology
2.
Nat Commun ; 4: 2842, 2013.
Article in English | MEDLINE | ID: mdl-24280864

ABSTRACT

Thymic epithelial cells (TEC) are heterogeneous stromal cells that generate microenvironments required for the formation of T cells within the thymus. Defects in TEC lead to immunodeficiency or autoimmunity. Here we identify TEC as the major source of cysteine-rich protein 61 (CYR61), a matricellular protein implicated in cell proliferation and migration. Binding of CYR61 to LFA-1, ICAM-1 and integrin α6 supports the adhesion of TEC and thymocytes as well as their interaction. Treatment of thymic lobes with recombinant CYR61 expands the stromal compartment by inducing the proliferation of TEC and activates Akt signalling. Engraftment of CYR61-overexpressing thymic lobes into athymic nude mice drastically boosts the yield of thymic output via expansion of TEC. This increases the space for the recruitment of circulating hematopoietic progenitors and the development of T cells. Our discovery paves the way for therapeutic interventions designed to restore thymus stroma and T-cell generation.


Subject(s)
Cysteine-Rich Protein 61/metabolism , Epithelial Cells/cytology , Stem Cells/cytology , T-Lymphocytes/cytology , Thymus Gland/cytology , Animals , Cell Adhesion , Cell Proliferation , Cells, Cultured , Cysteine-Rich Protein 61/genetics , Epithelial Cells/metabolism , Humans , Integrin alpha6/genetics , Integrin alpha6/metabolism , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/metabolism , Lymphocyte Function-Associated Antigen-1/genetics , Lymphocyte Function-Associated Antigen-1/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Nude , Protein Binding , Stem Cells/metabolism , T-Lymphocytes/metabolism , Thymus Gland/metabolism
3.
Mol Cancer Ther ; 11(12): 2588-99, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23002094

ABSTRACT

Antiangiogenic drugs have been used as anticancer agents to target tumor endothelial cells or pericytes. Because of limited efficacy of the current monotherapies, there is a strong demand for the dual targeting of endothelial cells and pericytes. Here, we identify Olfactomedin-like 3 (Olfml3) as a novel proangiogenic cue within the tumor microenvironment. Tumor-derived Olfml3 is produced by both tumor endothelial cells and accompanying pericytes and deposited in the perivascular compartment. Blockade of Olfml3 by anti-Olfml3 antibodies is highly effective in reducing tumor vascularization, pericyte coverage, and tumor growth. In vitro, Olfml3 targeting is sufficient to inhibit endothelioma cell migration and sprouting. Olfml3 alone or through binding to BMP4 enhances the canonical SMAD1/5/8 signaling pathway required for BMP4-induced angiogenesis. Therefore, Olfml3 blockade provides a novel strategy to control tumor growth by targeting two distinct cell types within the tumor microenvironment using a single molecule.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Carcinoma, Lewis Lung/blood supply , Carcinoma, Lewis Lung/drug therapy , Glycoproteins/antagonists & inhibitors , Pericytes/drug effects , Pericytes/metabolism , Animals , Antibodies/immunology , Antibodies/pharmacology , Bone Morphogenetic Protein 4/metabolism , Carcinoma, Lewis Lung/metabolism , Carcinoma, Lewis Lung/pathology , Female , Gene Silencing , Glycoproteins/biosynthesis , Glycoproteins/genetics , Glycoproteins/immunology , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Immunohistochemistry , Mice , Mice, Inbred C57BL , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Pericytes/pathology , Signal Transduction , Smad Proteins/metabolism , Swine , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...