Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Cardiovasc Res ; 113(14): 1776-1788, 2017 Dec 01.
Article in English | MEDLINE | ID: mdl-29016873

ABSTRACT

AIMS: The formation of cell-cell and cell-extra cellular matrix (ECM) contacts by endothelial cells (ECs) is crucial for the stability and integrity of a vascular network. We previously identified cingulin-like 1 (Cgnl1) in a transcriptomic screen for new angiogenic modulators. Here we aim to study the function of the cell-cell junction associated protein Cgnl1 during vessel formation. METHODS AND RESULTS: Unlike family member cingulin, Cgnl1 expression is enriched in ECs during vascular growth. Cgnl1 is important for the formation of multicellular tubule structures, as shown in vitro using loss-of function assays in a 3D matrix co-culture system that uses primary human ECs and supporting mural cells. Further studies revealed that Cgnl1 regulates vascular growth by promoting Ve-cadherin association with the actin cytoskeleton, thereby stabilizing adherens junctions. Cgnl1 also regulates focal adhesion assembly in response to ECM contact, promoting vinculin and paxillin recruitment and focal adhesion kinase signalling. In vivo, we demonstrate in a postnatal retinal vascular development model in mice that Cgnl1 function is crucial for sustaining neovascular growth and stability. CONCLUSIONS: Our data demonstrate a functional relevance for Cgnl1 as a defining factor in new vessel formation both in vitro and in vivo.


Subject(s)
Adherens Junctions/metabolism , Cytoskeletal Proteins/metabolism , Endothelial Cells/metabolism , GTP Phosphohydrolases/metabolism , Membrane Proteins/metabolism , Neovascularization, Physiologic/physiology , Actin Cytoskeleton/metabolism , Animals , Cell Adhesion/physiology , Cytoskeletal Proteins/genetics , Endothelium, Vascular/metabolism , Humans , Intercellular Junctions/metabolism , Membrane Proteins/genetics , Mice, Inbred C57BL
2.
PLoS One ; 12(6): e0178779, 2017.
Article in English | MEDLINE | ID: mdl-28628621

ABSTRACT

BACKGROUND: Intracoronary infusion of autologous bone marrow-derived mononuclear cells (BMMNC), after acute myocardial infarction (AMI), has been shown to improve myocardial function. However, therapeutic efficacy is limited, possibly because cell retention rates are low, suggesting that optimization of cell retention might increase therapeutic efficacy. Since retention of injected BMMNC is observed only within infarcted, but not remote, myocardium, we hypothesized that adhesion molecules on activated endothelium following reperfusion are essential. Consequently, we investigated the role of vascular cell adhesion molecule 1 (VCAM-1) in BMMNC retention in swine undergoing reperfused AMI produced by 120 min of percutaneous left circumflex coronary occlusion. METHODS AND RESULTS: VCAM-1 expression in the infarct and remote region was quantified at 1, 3, 7, 14, and 35 days, post-reperfusion (n≥6 swine per group). Since expression levels were significantly higher at 3 days (2.41±0.62%) than at 7 days (0.98±0.28%; p<0.05), we compared the degree of cell retention at those time points in a follow-up study, in which an average of 43·106 autologous BMMNCs were infused intracoronary at 3, or 7 days, post-reperfusion (n = 6 swine per group) and retention was histologically quantified one hour after intracoronary infusion of autologous BMMNCs. Although VCAM-1 expression correlated with retention of BMMNC within each time point, overall BMMNC retention was similar at day 3 and day 7 (2.3±1.3% vs. 3.1±1.4%, p = 0.72). This was not due to the composition of infused bone marrow cell fractions (analyzed with flow cytometry; n = 5 per group), as cell composition of the infused BMMNC fractions was similar. CONCLUSION: These findings suggest that VCAM-1 expression influences to a small degree, but is not the principal determinant of, BMMNC retention.


Subject(s)
Leukocytes, Mononuclear/transplantation , Myocardial Infarction/pathology , Vascular Cell Adhesion Molecule-1/metabolism , Acute Disease , Animals , Bone Marrow Cells/cytology , Cells, Cultured , Follow-Up Studies , Immunohistochemistry , Leukocytes, Mononuclear/cytology , Myocardial Infarction/metabolism , Myocardial Infarction/therapy , Swine , Time Factors , Up-Regulation , Vascular Cell Adhesion Molecule-1/genetics
3.
Cardiovasc Res ; 110(1): 129-39, 2016 May 01.
Article in English | MEDLINE | ID: mdl-26822228

ABSTRACT

AIMS: Impairment of the endothelial barrier leads to microvascular breakdown in cardiovascular disease and is involved in intraplaque haemorrhaging and the progression of advanced atherosclerotic lesions that are vulnerable to rupture. The exact mechanism that regulates vascular integrity requires further definition. Using a microarray screen for angiogenesis-associated genes during murine embryogenesis, we identified thrombospondin type I domain 1 (THSD1) as a new putative angiopotent factor with unknown biological function. We sought to characterize the role of THSD1 in endothelial cells during vascular development and cardiovascular disease. METHODS AND RESULTS: Functional knockdown of Thsd1 in zebrafish embryos and in a murine retina vascularization model induced severe haemorrhaging without affecting neovascular growth. In human carotid endarterectomy specimens, THSD1 expression by endothelial cells was detected in advanced atherosclerotic lesions with intraplaque haemorrhaging, but was absent in stable lesions, implying involvement of THSD1 in neovascular bleeding. In vitro, stimulation with pro-atherogenic factors (3% O2 and TNFα) decreased THSD1 expression in human endothelial cells, whereas stimulation with an anti-atherogenic factor (IL10) showed opposite effect. Therapeutic evaluation in a murine advanced atherosclerosis model showed that Thsd1 overexpression decreased plaque vulnerability by attenuating intraplaque vascular leakage, subsequently reducing macrophage accumulation and necrotic core size. Mechanistic studies in human endothelial cells demonstrated that THSD1 activates FAK-PI3K, leading to Rac1-mediated actin cytoskeleton regulation of adherens junctions and focal adhesion assembly. CONCLUSION: THSD1 is a new regulator of endothelial barrier function during vascular development and protects intraplaque microvessels against haemorrhaging in advanced atherosclerotic lesions.


Subject(s)
Atherosclerosis/metabolism , Endothelial Cells/metabolism , Microvessels/metabolism , Neovascularization, Pathologic/metabolism , Thrombospondins/metabolism , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/metabolism , Carotid Artery Diseases/metabolism , Female , Humans , Male , Mice, Inbred C57BL , Phosphatidylinositol 3-Kinases/metabolism , Plaque, Atherosclerotic/pathology , Thrombospondin 1/metabolism
4.
Circulation ; 125(25): 3142-58, 2012 Jun 26.
Article in English | MEDLINE | ID: mdl-22661514

ABSTRACT

BACKGROUND: New vessel formation contributes to organ development during embryogenesis and tissue repair in response to mechanical damage, inflammation, and ischemia in adult organisms. Early angiogenesis includes formation of an excessive primitive network that needs to be reorganized into a secondary vascular network with higher hierarchical structure. Vascular pruning, the removal of aberrant neovessels by apoptosis, is a vital step in this process. Although multiple molecular pathways for early angiogenesis have been identified, little is known about the genetic regulators of secondary network development. METHODS AND RESULTS: Using a transcriptomics approach, we identified a new endothelial specific gene named FYVE, RhoGEF, and PH domain-containing 5 (FGD5) that plays a crucial role in vascular pruning. Loss- and gain-of-function studies demonstrate that FGD5 inhibits neovascularization, indicated by in vitro tube-formation, aortic-ring, and coated-bead assays and by in vivo coated-bead plug assays and studies in the murine retina model. FGD5 promotes apoptosis-induced vaso-obliteration via induction of the hey1-p53 pathway by direct binding and activation of cdc42. Indeed, FGD5 correlates with apoptosis in endothelial cells during vascular remodeling and was linked to rising p21(CIP1) levels in aging mice. CONCLUSION: We have identified FGD5 as a novel genetic regulator of vascular pruning by activation of endothelial cell-targeted apoptosis.


Subject(s)
Apoptosis Regulatory Proteins/physiology , Endothelium, Vascular/pathology , Guanine Nucleotide Exchange Factors/physiology , Human Umbilical Vein Endothelial Cells/pathology , Neovascularization, Pathologic/pathology , Neovascularization, Pathologic/prevention & control , Animals , Apoptosis Regulatory Proteins/genetics , Cell Proliferation , Cells, Cultured , Endothelium, Vascular/metabolism , Guanine Nucleotide Exchange Factors/genetics , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Mice , Mice, Inbred C57BL , Mice, SCID , Neovascularization, Pathologic/genetics , Retinal Diseases/genetics , Retinal Diseases/metabolism , Retinal Diseases/pathology , Transcriptome/genetics
5.
Arterioscler Thromb Vasc Biol ; 32(5): 1289-98, 2012 May.
Article in English | MEDLINE | ID: mdl-22426130

ABSTRACT

OBJECTIVE: In cardiovascular regulation, heme oxygenase-1 (HO-1) activity has been shown to inhibit vascular smooth muscle cell (VSMC) proliferation by promoting cell cycle arrest at the G1/S phase. However, the effect of HO-1 on VSMC migration remains unclear. We aim to elucidate the mechanism by which HO-1 regulates PDGFBB-induced VSMC migration. METHODS AND RESULTS: Transduction of HO-1 cDNA adenoviral vector severely impeded human VSMC migration in a scratch, transmembrane, and directional migration assay in response to PDGFBB stimulation. Similarly, HO-1 overexpression in the remodeling process during murine retinal vasculature development attenuated VSMC coverage over the major arterial branches as compared with sham vector-transduced eyes. HO-1 expression in VSMCs significantly upregulated VEGFA and VEGFR2 expression, which subsequently promoted the formation of inactive PDGFRß/VEGFR2 complexes. This compromised PDGFRß phosphorylation and impeded the downstream cascade of FAK-p38 signaling. siRNA-mediated silencing of VEGFA or VEGFR2 could reverse the inhibitory effect of HO-1 on VSMC migration. CONCLUSIONS: These findings identify a potent antimigratory function of HO-1 in VSMCs, a mechanism that involves VEGFA and VEGFR2 upregulation, followed by assembly of inactive VEGFR2/PDGFRß complexes that attenuates effective PDGFRß signaling.


Subject(s)
Heme Oxygenase-1/pharmacology , Muscle, Smooth, Vascular/metabolism , Platelet-Derived Growth Factor/metabolism , RNA, Messenger/genetics , Receptor, Platelet-Derived Growth Factor beta/metabolism , Up-Regulation/drug effects , Vascular Endothelial Growth Factor Receptor-2/genetics , Cell Movement , Cell Proliferation , Heme Oxygenase-1/metabolism , Humans , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Platelet-Derived Growth Factor/pharmacology , Signal Transduction , Vascular Endothelial Growth Factor Receptor-2/biosynthesis
6.
Circ Res ; 109(4): 382-95, 2011 Aug 05.
Article in English | MEDLINE | ID: mdl-21700929

ABSTRACT

RATIONALE: Neovascularization is required for embryonic development and plays a central role in diseases in adults. In atherosclerosis, the role of neovascularization remains to be elucidated. In a genome-wide microarray-screen of Flk1+ angioblasts during murine embryogenesis, the v-ets erythroblastosis virus E26 oncogene homolog 2 (Ets2) transcription factor was identified as a potential angiogenic factor. OBJECTIVES: We assessed the role of Ets2 in endothelial cells during atherosclerotic lesion progression toward plaque instability. METHODS AND RESULTS: In 91 patients treated for carotid artery disease, Ets2 levels showed modest correlations with capillary growth, thrombogenicity, and rising levels of tumor necrosis factor-α (TNFα), monocyte chemoattractant protein 1, and interleukin-6 in the atherosclerotic lesions. Experiments in ApoE(-/-) mice, using a vulnerable plaque model, showed that Ets2 expression was increased under atherogenic conditions and was augmented specifically in the vulnerable versus stable lesions. In endothelial cell cultures, Ets2 expression and activation was responsive to the atherogenic cytokine TNFα. In the murine vulnerable plaque model, overexpression of Ets2 promoted lesion growth with neovessel formation, hemorrhaging, and plaque destabilization. In contrast, Ets2 silencing, using a lentiviral shRNA construct, promoted lesion stabilization. In vitro studies showed that Ets2 was crucial for TNFα-induced expression of monocyte chemoattractant protein 1, interleukin-6, and vascular cell adhesion molecule 1 in endothelial cells. In addition, Ets2 promoted tube formation and amplified TNFα-induced loss of vascular endothelial integrity. Evaluation in a murine retina model further validated the role of Ets2 in regulating vessel inflammation and endothelial leakage. CONCLUSIONS: We provide the first evidence for the plaque-destabilizing role of Ets2 in atherosclerosis development by induction of an intraplaque proinflammatory phenotype in endothelial cells.


Subject(s)
Aortic Diseases/metabolism , Carotid Artery Diseases/metabolism , Endothelial Cells/metabolism , Inflammation/metabolism , Proto-Oncogene Protein c-ets-2/metabolism , Analysis of Variance , Animals , Aortic Diseases/immunology , Aortic Diseases/pathology , Aortic Diseases/physiopathology , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Carotid Artery Diseases/immunology , Carotid Artery Diseases/pathology , Carotid Artery Diseases/physiopathology , Cells, Cultured , Chemokine CCL2/metabolism , Disease Models, Animal , Endothelial Cells/immunology , Hemorrhage/metabolism , Humans , Inflammation/immunology , Inflammation/pathology , Inflammation/physiopathology , Inflammation Mediators/metabolism , Interleukin-6/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/physiopathology , Neovascularization, Physiologic , Phenotype , Proto-Oncogene Protein c-ets-2/genetics , Retinal Vessels/metabolism , Retinal Vessels/pathology , Rupture , Time Factors , Transfection , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation , Vascular Cell Adhesion Molecule-1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...