Lung vascular endothelial barrier function is regulated primarily by adherens junctions (AJs) consisting of VE- cadherin, which mediates adhesion of endothelial cells through homotypic interaction, and associated catenins. Recently, another VE-cadherin partner Vascular Endothelial Protein Tyrosine Phosphatase, VE-PTP, has been identified in regulating endothelial barrier function. Disassembly of AJs through internalization and subsequent degradation of VE-cadherin disrupts the endothelial barrier resulting severe intractable protein-rich pulmonary edema, the central pathogenic feature of ARDS. Restoration of the endothelial barrier is essential for resolving edema, yet the mechanisms underlying re-assembly of AJs are poorly understood. Based on our Supporting Data, in Project 1 we posit that VE-PTP is a central regulator of re-assembly of AJs that plays a critical role in restoring endothelial barrier function through its ability to stabilize VE-cadherin at AJs. We also showed that signaling via Hypoxia-Inducible Factors (HIFs) was required for the synthesis of VE-PTP. Notably, in addition to VE-PTP binding to VE-cadherin, another pool of VE-PTP is associated with Tie2, the tyrosine kinase receptor present in the endothelial plasmalemma, and which functions to restrain Tie2 activity. On the basis of these observations, in Project 1 we will pursue the following Specific Aims: (i) we will determine the synergistic role of (a) HIF-dependent expression of VE-PTP as an adaptive mechanism promoting the stabilization of VE-cadherin at AJs, and (b) HIF-dependent activation of Rac1 and Cdc42 at AJs in sealing and strengthening the AJ barrier and thus restoring lung fluid balance; (ii) we will determine the role of VE-PTP interaction with Tie2 in regulating the repair of AJs through the activation of Tie2 signaling and the translocation of VE-PTP to VE-cadherin, and resulting in VE-cadherin stabilization; and (iii) we will determine the therapeutic value of (a) blocking prolyl hydroxylases (PHDs) to activate HIF-mediated transcription of VE-PTP, and (b) inhibiting Tie2 interaction with VE-PTP in restoring lung vascular barrier function and fluid balance in models of inflammatory lung injury. We will apply a rigorous multidisciplinary approach to define the signaling mechanisms activated by VE-PTP interaction with VE-cadherin in restoring the integrity of lung endothelial barrier, with the intent of identifying new targets to normalize lung fluid balance and the course of inflammatory lung injury.

Public Health Relevance

VE-PTP interaction with VE-cadherin at adherens junctions (AJs) of the endothelium is required for AJs re- annealing. We will study the function of VE-PTP that may have an essential role in the restoration of lung endothelial junctional barrier and fluid balance after inflammatory injury.

Agency
National Institute of Health (NIH)
Institute
National Heart, Lung, and Blood Institute (NHLBI)
Type
Research Program Projects (P01)
Project #
5P01HL060678-17
Application #
9324310
Study Section
Special Emphasis Panel (ZHL1)
Program Officer
Xiao, Lei
Project Start
Project End
Budget Start
2017-07-01
Budget End
2018-06-30
Support Year
17
Fiscal Year
2017
Total Cost
Indirect Cost
Name
University of Illinois at Chicago
Department
Type
DUNS #
098987217
City
Chicago
State
IL
Country
United States
Zip Code
60612
Marsboom, Glenn; Rehman, Jalees (2018) Hypoxia Signaling in Vascular Homeostasis. Physiology (Bethesda) 33:328-337
Lv, Yang; Kim, Kyungho; Sheng, Yue et al. (2018) YAP Controls Endothelial Activation and Vascular Inflammation Through TRAF6. Circ Res 123:43-56
Christoforidis, Theodore; Driver, Tom G; Rehman, Jalees et al. (2018) Generation of controllable gaseous H2S concentrations using microfluidics. RSC Adv 8:4078-4083
Di, Anke; Xiong, Shiqin; Ye, Zhiming et al. (2018) The TWIK2 Potassium Efflux Channel in Macrophages Mediates NLRP3 Inflammasome-Induced Inflammation. Immunity 49:56-65.e4
Chen, Zhenlong; D S Oliveira, Suellen; Zimnicka, Adriana M et al. (2018) Reciprocal regulation of eNOS and caveolin-1 functions in endothelial cells. Mol Biol Cell 29:1190-1202
Le Master, Elizabeth; Huang, Ru-Ting; Zhang, Chongxu et al. (2018) Proatherogenic Flow Increases Endothelial Stiffness via Enhanced CD36-Mediated Uptake of Oxidized Low-Density Lipoproteins. Arterioscler Thromb Vasc Biol 38:64-75
Potje, Simone R; Chen, Zhenlong; Oliveira, Suellen D'Arc S et al. (2017) Nitric oxide donor [Ru(terpy)(bdq)NO]3+ induces uncoupling and phosphorylation of endothelial nitric oxide synthase promoting oxidant production. Free Radic Biol Med 112:587-596
Tsang, Kit Man; Hyun, James S; Cheng, Kwong Tai et al. (2017) Embryonic Stem Cell Differentiation to Functional Arterial Endothelial Cells through Sequential Activation of ETV2 and NOTCH1 Signaling by HIF1?. Stem Cell Reports 9:796-806
Marsboom, Glenn; Chen, Zhenlong; Yuan, Yang et al. (2017) Aberrant caveolin-1-mediated Smad signaling and proliferation identified by analysis of adenine 474 deletion mutation (c.474delA) in patient fibroblasts: a new perspective on the mechanism of pulmonary hypertension. Mol Biol Cell 28:1177-1185
Andresen Eguiluz, Roberto C; Kaylan, Kerim B; Underhill, Gregory H et al. (2017) Substrate stiffness and VE-cadherin mechano-transduction coordinate to regulate endothelial monolayer integrity. Biomaterials 140:45-57

Showing the most recent 10 out of 200 publications