Cell-free hemoglobin-mediated human lung microvascular endothelial barrier dysfunction is not mediated by cell death

2021 ◽  
Vol 556 ◽  
pp. 199-206
Author(s):  
Toria Tomasek ◽  
Lorraine B. Ware ◽  
Julie A. Bastarache ◽  
Jamie E. Meegan
Cells ◽  
2021 ◽  
Vol 10 (6) ◽  
pp. 1489
Author(s):  
Ruben M. L. Colunga Biancatelli ◽  
Pavel Solopov ◽  
Betsy Gregory ◽  
John D. Catravas

Exposure to hydrochloric acid (HCl) leads acutely to asthma-like symptoms, acute respiratory distress syndrome (ARDS), including compromised alveolo-capillary barrier, and respiratory failure. To better understand the direct effects of HCl on pulmonary endothelial function, we studied the characteristics of HCl-induced endothelial barrier dysfunction in primary cultures of human lung microvascular endothelial cells (HLMVEC), defined the involved molecular pathways, and tested the potentially beneficial effects of Heat Shock Protein 90 (HSP90) inhibitors. HCl impaired barrier function in a time- and concentration-dependent manner and was associated with activation of Protein Kinase B (AKT), Ras homolog family member A (RhoA) and myosin light chain 2 (MLC2), as well as loss of plasmalemmal VE-cadherin, rearrangement of cortical actin, and appearance of inter-endothelial gaps. Pre-treatment or post-treatment of HLMVEC with AUY-922, a third-generation HSP90 inhibitor, prevented and restored HCl-induced endothelial barrier dysfunction. AUY-922 increased the expression of HSP70 and inhibited the activation (phosphorylation) of extracellular-signal regulated kinase (ERK) and AKT. AUY-922 also prevented the HCl-induced activation of RhoA and MLC2 and the internalization of plasmalemmal VE-cadherin. We conclude that, by increasing the expression of cytoprotective proteins, interfering with actomyosin contractility, and enhancing the expression of junction proteins, inhibition of HSP90 may represent a useful approach for the management of HCl-induced endothelial dysfunction and acute lung injury.


2017 ◽  
Vol 57 (3) ◽  
pp. 307-314 ◽  
Author(s):  
Sunit Singla ◽  
Justin R. Sysol ◽  
Benjamin Dille ◽  
Nicole Jones ◽  
Jiwang Chen ◽  
...  

2020 ◽  
Vol 34 (S1) ◽  
pp. 1-1
Author(s):  
Jamie E. Meegan ◽  
Toria Tomasek ◽  
Lorraine B. Ware ◽  
Julie A. Bastarache

Circulation ◽  
2014 ◽  
Vol 130 (suppl_2) ◽  
Author(s):  
Panfeng Fu ◽  
Anne E Cress ◽  
Ting Wang ◽  
Joe G Garcia ◽  
Viswanathan Natarajan

Paxillin, a multi-domain scaffold-adapter focal adhesion (FA) protein, plays an important role in facilitating protein networking and efficient signaling transduction. Paxillin is phosphorylated at multiple serine/threonine and tyrosine residues; however, the role of tyrosine phosphorylation of paxillin in endothelial barrier dysfunction and the acute respiratory distress syndrome (ARDS) remains unclear. In this study, we used paxillin-specific siRNA and site-specific non-phosphorylatable mutants of paxillin to abrogate the function of paxillin, both in vitro and in vivo, to determine its role in the regulation of lung endothelial permeability and ARDS. In vitro, lipopolysaccharide (LPS) challenge of human lung microvascular endothelial cells (ECs) resulted in paxillin accumulation at focal adhesions, enhanced tyrosine phosphorylation of paxillin at Y31 and Y118, and significant endothelial barrier dysfunction. However no significant changes in Y181 phosphorylation by LPS challenge was observed. Paxillin silencing (siRNA) attenuated LPS-induced endothelial barrier dysfunction and dissociation of VE-cadherin from adherens junctions. LPS-induced paxillin phosphorylation at Y31 and Y118 was mediated by c-Abl tyrosine kinase and not by Src or focal adhesion kinase (FAK) in human lung microvascular ECs. Furthermore, down-regulation of c-Abl (siRNA) significantly reduced LPS-mediated endothelial barrier dysfunction. Transfection of human lung microvascular ECs with paxillin Y31, Y118 and Y31/Y118 mutants mitigated LPS-induced barrier dysfunction and VE-cadherin destabilization at adherens junctions. In vivo, knockdown of paxillin with siRNA in mouse lungs ameliorated LPS-induced pulmonary protein leak and lung inflammation. Together, these results suggest that c-Abl-mediated tyrosine phosphorylation of paxillin at Y31 and Y118 regulates LPS-mediated pulmonary vascular permeability and injury.


PLoS ONE ◽  
2016 ◽  
Vol 11 (8) ◽  
pp. e0160875 ◽  
Author(s):  
Li-Yun Huang ◽  
Christine Stuart ◽  
Kazuyo Takeda ◽  
Felice D’Agnillo ◽  
Basil Golding

Sign in / Sign up

Export Citation Format

Share Document