Reactive oxygen species mediate Rac-induced loss of cell-cell adhesion in primary human endothelial cells

2002 ◽  
Vol 115 (9) ◽  
pp. 1837-1846 ◽  
Author(s):  
Sandra van Wetering ◽  
Jaap D. van Buul ◽  
Safira Quik ◽  
Frederik P. J. Mul ◽  
Eloise C. Anthony ◽  
...  

The integrity of the endothelium is dependent on cell-cell adhesion, which is mediated by vascular-endothelial (VE)-cadherin. Proper VE-cadherin-mediated homotypic adhesion is, in turn, dependent on the connection between VE-cadherin and the cortical actin cytoskeleton. Rho-like small GTPases are key molecular switches that control cytoskeletal dynamics and cadherin function in epithelial as well as endothelial cells. We show here that a cell-penetrating, constitutively active form of Rac (Tat-RacV12) induces a rapid loss of VE-cadherin-mediated cell-cell adhesion in endothelial cells from primary human umbilical veins (pHUVEC). This effect is accompanied by the formation of actin stress fibers and is dependent on Rho activity. However,transduction of pHUVEC with Tat-RhoV14, which induces pronounced stress fiber and focal adhesion formation, did not result in a redistribution of VE-cadherin or an overall loss of cell-cell adhesion. In line with this observation, endothelial permeability was more efficiently increased by Tat-RacV12 than by Tat-RhoV14. The loss of cell-cell adhesion, which is induced by Tat-RacV12, occurred in parallel to and was dependent upon the intracellular production of reactive oxygen species (ROS). Moreover, Tat-RacV12 induced an increase in tyrosine phosphorylation of a component the VE-cadherin-catenin complex, which was identified as α-catenin. The functional relevance of this signaling pathway was further underscored by the observation that endothelial cell migration, which requires a transient reduction of cell-cell adhesion, was blocked when signaling through ROS was inhibited. In conclusion, Rac-mediated production of ROS represents a previously unrecognized means of regulating VE-cadherin function and may play an important role in the (patho)physiology associated with inflammation and endothelial damage as well as with endothelial cell migration and angiogenesis.

2011 ◽  
Vol 301 (3) ◽  
pp. C695-C704 ◽  
Author(s):  
Youxue Wang ◽  
Qun S. Zang ◽  
Zijuan Liu ◽  
Qian Wu ◽  
David Maass ◽  
...  

Endothelial migration is a crucial aspect of a variety of physiologic and pathologic conditions including atherosclerosis and vascular repair. Reactive oxygen species (ROS) function as second messengers during endothelial migration. Multiple intracellular sources of ROS are regulated by cellular context, external stimulus, and the microenvironment. However, the predominant source of ROS during endothelial cell (EC) migration and the mechanisms by which ROS regulate cell migration are incompletely understood. In this study, we tested the hypothesis that mitochondria-derived ROS (mtROS) regulate EC migration. In cultured human umbilical vein endothelial cells, VEGF increased mitochondrial metabolism, promoted mtROS production, and induced cell migration. Either the targeted mitochondrial delivery of the antioxidant, vitamin E (Mito-Vit-E), or the depletion of mitochondrial DNA abrogated VEGF-mediated mtROS production. Overexpression of mitochondrial catalase also inhibited VEGF-induced mitochondrial metabolism, Rac activation, and cell migration. Furthermore, these interventions suppressed VEGF-stimulated EC migration and blocked Rac1 activation in endothelial cells. Constitutively active Rac1 reversed Mito-Vit-E-induced inhibition of EC migration. Mito-Vit-E also attenuated carotid artery reendothelialization in vivo. These results provide strong evidence that mtROS regulate EC migration through Rac-1.


Circulation ◽  
2002 ◽  
Vol 106 (8) ◽  
pp. 981-986 ◽  
Author(s):  
Carmen Urbich ◽  
Elisabeth Dernbach ◽  
Alexandra Aicher ◽  
Andreas M. Zeiher ◽  
Stefanie Dimmeler

2017 ◽  
Vol 35 (1) ◽  
pp. 65
Author(s):  
Kanjana Jittiporn ◽  
Wisuda Suvitayavat ◽  
Primchanien Moongkarndi ◽  
Rulth B Caldwell

Objective: This study aimed to determine the effect of mangosteen extract on hypoxia induced reactive oxygen species production and vascular endothelial growth factor (VEGF) induced retinal endothelial cell migration.Material and Method: This research studied bovine retinal endothelial cells. The non-toxic concentration of mangosteen extract of water soluble part was verified using trypan blue staining. The effects of mangosteen extract on hypoxia induced reactive oxygen species production and retinal endothelial cells migration were determined using 2’, 7’ dichlorodihydrofluorescein diacetate and scrape/wound assay, respectively. The mechanism of mangosteen extract on retinal endothelial cell migration was determined using western blotting. The analysis of variance was used to determine the differences among group means.Results: The concentrations of mangosteen extract at 25, 50 and 100 mg/ml were non-toxic and these concentrations were used in further experiments. Mangosteen extract at a dose of 100 mg/ml significantly attenuated hypoxia induced reactive oxygen species formation. At all doses, mangosteen extract also significantly inhibited retinal endothelial cell migration. However, the mechanism of mangosteen extract on VEGF signaling did not affect the phosphorylation of VEGF receptor 2 (VEGFR2).Conclusion: Mangosteen extract has anti-oxidant and anti-migration effects.


2005 ◽  
Vol 25 (11) ◽  
pp. 2295-2300 ◽  
Author(s):  
Satoshi Ikeda ◽  
Minako Yamaoka-Tojo ◽  
Lula Hilenski ◽  
Nikolay A. Patrushev ◽  
Ghulam M. Anwar ◽  
...  

2015 ◽  
Vol 98 ◽  
pp. 74-81 ◽  
Author(s):  
Sotiria Tsirmoula ◽  
Margarita Lamprou ◽  
Maria Hatziapostolou ◽  
Nelly Kieffer ◽  
Evangelia Papadimitriou

2014 ◽  
Vol 464 (1) ◽  
pp. 157-168 ◽  
Author(s):  
Anne H.-H. Tseng ◽  
Li-Hong Wu ◽  
Shyan-Shu Shieh ◽  
Danny Ling Wang

This article reports that hypoxia elicits SIRT3 to deacetylate FOXO3 in endothelial cells. This drives an increase in the expression of mitochondrial antioxidant enzymes, reduces accumulation of reactive oxygen species in mitochondria and thereby confers cellular capacity to adapt to hypoxia.


2007 ◽  
Vol 31 (1) ◽  
pp. 104-113 ◽  
Author(s):  
Gangaraju Rajashekhar ◽  
Matthew Grow ◽  
Antje Willuweit ◽  
Carolyn E. Patterson ◽  
Matthias Clauss

Activation of the vascular endothelium with cytokines such as TNF is widely used to study the role of the vasculature in proinflammatory disease. To gain insight into mechanisms of prolonged vascular endothelial activation we compared changes in gene expression induced by continuous activation in stable tmTNF-expressing cells with changes due to acute TNF challenge in vitro. Affymetrix Genechip analysis was performed on RNA from control, acute and continuous TNF-activated endothelial cells. Only 36% of the significant changes in gene expression were convergent between the acute and continuously activated endothelial cells compared with the control. From the divergently regulated genes, for example the cytokine ENA-78 was specifically induced in chronically activated cells, while E-selectin, a cell adhesion molecule, was upregulated only in acutely activated endothelial cells. Antioxidant SOD gene induction was noted in acute activation, while a regulatory NADPH oxidase subunit was selectively upregulated in continuously activated endothelium in accordance with significant reactive oxygen species induction occurred only in these cells. Accordingly, p38 and ERK1/2, two MAP kinases downstream of reactive oxygen species, were activated in stable transmembrane-spanning precursor (tm) TNF-expressing cells and were refractory to activation with soluble TNF or VEGF. In consequence, the increased p38 MAP kinase activity contributed to increased endothelial cell migration in tmTNF-expressing cells. These data suggest that continuous activation of endothelial cells leads to specific expression and functional changes, consistent with alterations observed in dysfunctional endothelium exposed to or involved in chronic inflammation.


2008 ◽  
Vol 99 (02) ◽  
pp. 363-372 ◽  
Author(s):  
Christopher J. Kuckleburg ◽  
Raksha Tiwari ◽  
Charles J. Czuprynski

SummaryA common feature of severe sepsis is vascular inflammation and damage to the endothelium. Because platelets can be directly activated by bacteria and endotoxin, these cells may play an important role in determining the outcome of sepsis. For example, inhibiting platelet interactions with the endothelium has been shown to attenuate endothelial cell damage and improve survival during sepsis. Although not entirely understood, the interactions between bacteria-activated platelets and the endothelium may play a key role in the vascular pathology of bacterial sepsis. Haemophilus somnus is a bacterial pathogen that causes diffuse vascular inflammation and endothelial damage. In some cases H.somnus infection results in an acute and fatal form of vasculitis in the cerebral microvasculature known as thrombotic meningoencephalitis (TME). In this study, we have characterized the mechanisms involved in endothelial cell apoptosis induced by activated platelets. We observed that direct contact between H.somnus-activated platelets and endothelial cells induced significant levels of apoptosis; however, Fas receptor activation on bovine endothelial cells was not able to induce apoptosis unless protein synthesis was disrupted. Endothelial cell apoptosis by H.somnus-activated platelets required activation of both caspase-8 and caspase-9, as inhibitors of either caspase inhibited apoptosis. Furthermore, activated platelets induced endothelial cell production of reactive oxygen species (ROS) and disrupting ROS activity in endothelial cells significantly inhibited apoptosis. These findings suggest that bacterial activation of platelets may contribute to endothelial cell dysfunction observed during sepsis, specifically by inducing endothelial cell apoptosis.


Sign in / Sign up

Export Citation Format

Share Document