180 Inventory of “atherosclerotic genes” induced or repressed in endothelial cells and smooth muscle cells by differential display RT-PCR

1997 ◽  
Vol 11 ◽  
pp. 51
Author(s):  
Anton J.G. Horrevoets ◽  
Carlie J.M. de Vries ◽  
A.E. van Achterberg ◽  
Ruud D. Fontijn ◽  
Benien van Aken ◽  
...  
2014 ◽  
Vol 34 (suppl_1) ◽  
Author(s):  
Mao Luo ◽  
Min Zeng ◽  
Meiping Ren ◽  
Rong Li ◽  
Xin Deng ◽  
...  

Objective: Platelets can regulate endothelial cell genes expression through their adhesion to the subendothelium in response to vascular injury. Recently it has been reported that some endothelial cells (ECs)-secreted genes can modulate vascular smooth muscle cells (VSMCs) phenotypic transformation by ECs/VSMCs co-culture. However, little is known about the effects of platelets adhesion to ECs interaction on VSMCs phenotype. In this study, we investigated the role of some genes secreted by platelets adhesion to EC in regulating SMC phenotypic transformation. Methods and Results: By Q-RT-PCR, expression of PAI-1, MMP-2 and MMP-9 were up-regulated in ECs after platelets adhesion to ECs comparing to ECs monoculture alone. In migration (scratch) and proliferation (CCK-8) assays, platelets adhesion to ECs increased EC migration-promoting activity and proliferative activity. After adhesion of platelets to ECs/VSMCs co-culture, expression of VSMCs contractile apparatus SM-MHC, Smoothelin-B, SMA and SM22a were significantly decreased comparing to ECs/VSMCs co-culture by Q-RT-PCR, but decreasing slightly in adhesion with platelets from type 2 diabetes. Conversely, it increased the expression of synthetic marker Smemb, CCND-1, CCND-2. Further analysis found that platelets adhesion to ECs/VSMCs co-culture significantly increased VSMCs migration-promoting activity and proliferative activity. In Gelatin Zymography assays, there were high levels of MMP-2 and MMP-9 after adhesion of platelets to ECs/VSMCs co-culture comparing to ECs/VSMCs co-culture alone. Conclusion: These results offer important insights into the mechanisms controlling phenotypic modulation of VSMCs by platelets adhesion to ECs, and help to explain the effects of platelets-ECs-VSMCs interaction on VSMCs contractile/synthetic, migration, and proliferation, suggesting that the adhesion of platelets to ECs is a key process in regulating phenotypic switch of VSMCs.


1994 ◽  
Vol 72 (01) ◽  
pp. 044-053 ◽  
Author(s):  
N Chomiki ◽  
M Henry ◽  
M C Alessi ◽  
F Anfosso ◽  
I Juhan-Vague

SummaryIndividuals with elevated levels of plasminogen activator inhibitor type 1 are at risk of developing atherosclerosis. The mechanisms leading to increased plasma PAI-1 concentrations are not well understood. The link observed between increased PAI-1 levels and insulin resistance has lead workers to investigate the effects of insulin or triglyceride rich lipoproteins on PAI-1 production by cultured hepatocytes or endothelial cells. However, little is known about the contribution of these cells to PAI-1 production in vivo. We have studied the expression of PAI-1 in human liver sections as well as in vessel walls from different territories, by immunocytochemistry and in situ hybridization.We have observed that normal liver endothelial cells expressed PAI-1 while parenchymal cells did not. However, this fact does not refute the role of parenchymal liver cells in pathological states.In healthy vessels, PAI-1 mRNA and protein were detected primarily at the endothelium from the lumen as well as from the vasa vasorum. In normal arteries, smooth muscle cells were able to produce PAI-1 depending on the territory tested. In deeply altered vessels, PAI-1 expression was observed in neovessels scattering the lesions, in some intimal cells and in smooth muscle cells. Local increase PAI-1 mRNA described in atherosclerotic lesions could be due to the abundant neovascularization present in the lesion as well as a raised expression in smooth muscle cells. The increased PAI-1 in atherosclerosis could lead to fibrin deposit during plaque rupture contributing further to the development and progression of the lesion.


1982 ◽  
Vol 48 (01) ◽  
pp. 101-103 ◽  
Author(s):  
B Kirchhof ◽  
J Grünwald

SummaryEndothelial and smooth muscle cells cultured from minipig aorta were examined for their inhibitory activity on thrombin and for their thrombin generating capacity.Endothelial cells showed both a thrombin inhibition and an activation of prothrombin in the presence of Ca++, which was enhanced in the presence of phospholipids. Smooth muscle cells showed an activation of prothrombin but at a lower rate. Both coagulation and amidolytic micro-assays were suitable for studying the thrombin-vessel wall interaction.


1985 ◽  
Vol 53 (02) ◽  
pp. 165-169 ◽  
Author(s):  
Walter E Laug

SummaryTPure cultures of bovine endothelial cells (EC) produce and secrete large amounts of plasminogen activators (PA). Cocultivation of EC with vascular smooth muscle cells (SMC) resulted in a significant decrease of PA activities secreted by the EC, whereas the cellular PA activities remained unaffected. Secreted PA activities were absent in the growth medium as long as the SMC to EC ratio was 2:1 or higher. The PA inhibitory activity of the SMC was rapid and cell-to-cell contact was not necessary.The PA inhibitory activity was present in homogenates of SMC as well as in the medium conditioned by them but not in the extracellular matrix elaborated by these cells. Serum free medium conditioned by SMC neutralized both tissue type (t-PA) and urokinase like (u-PA) plasminogen activators. Gel electrophoretic analysis of SMC conditioned medium followed by reverse fibrin autography demonstrated PA inhibitory activities in the molecular weight (Mr) range of 50,000 to 52,000 similar to those present in media conditioned by bovine endothelial cells or fibroblasts. Regular fibrin zymography of SMC conditioned medium incubated with u-PA or t-PA revealed the presence of a component with a calculated approximate Mr of 45,000 to 50,000 which formed SDS resistant complexes with both types of PA.These data demonstrate that vascular SMC produce and secrete (a) inhibitor(s) of PAs which may influence the fibrinolytic potential of EC.


1996 ◽  
Vol 16 (10) ◽  
pp. 1263-1268 ◽  
Author(s):  
Antonio López Farré ◽  
Juan R. Mosquera ◽  
Lourdes Sánchez de Miguel ◽  
Inmaculada Millás ◽  
Trinidad de Frutos ◽  
...  

Cells ◽  
2021 ◽  
Vol 10 (6) ◽  
pp. 1485
Author(s):  
Adrian Sowka ◽  
Pawel Dobrzyn

Studies of adipose tissue biology have demonstrated that adipose tissue should be considered as both passive, energy-storing tissue and an endocrine organ because of the secretion of adipose-specific factors, called adipokines. Adiponectin is a well-described homeostatic adipokine with metabolic properties. It regulates whole-body energy status through the induction of fatty acid oxidation and glucose uptake. Adiponectin also has anti-inflammatory and antidiabetic properties, making it an interesting subject of biomedical studies. Perivascular adipose tissue (PVAT) is a fat depot that is conterminous to the vascular wall and acts on it in a paracrine manner through adipokine secretion. PVAT-derived adiponectin can act on the vascular wall through endothelial cells and vascular smooth muscle cells. The present review describes adiponectin’s structure, receptors, and main signaling pathways. We further discuss recent studies of the extent and nature of crosstalk between PVAT-derived adiponectin and endothelial cells, vascular smooth muscle cells, and atherosclerotic plaques. Furthermore, we argue whether adiponectin and its receptors may be considered putative therapeutic targets.


Sign in / Sign up

Export Citation Format

Share Document