Regulation of Protein Tyrosine Phosphatase Oxidation in Cell Adhesion and Migration

2014 ◽  
Vol 20 (13) ◽  
pp. 1994-2010 ◽  
Author(s):  
Jeroen Frijhoff ◽  
Markus Dagnell ◽  
Rinesh Godfrey ◽  
Arne Östman
2013 ◽  
Vol 7 (5) ◽  
pp. 418-423 ◽  
Author(s):  
Carlos O Arregui ◽  
Ángela González ◽  
Juan E Burdisso ◽  
Ana E González Wusener

2010 ◽  
Vol 299 (5) ◽  
pp. C1144-C1152 ◽  
Author(s):  
Hidehisa Shimizu ◽  
Yoshimi Nakagawa ◽  
Chie Murakami ◽  
Naohito Aoki ◽  
Shokei Kim-Mitsuyama ◽  
...  

Vascular smooth muscle cell (VSMC) proliferation and migration and vascular endothelial cell (VEC) dysfunction are closely associated with the development of atherosclerosis. We previously demonstrated that protein tyrosine phosphatase ε M (PTPεM) promotes VEC survival and migration. The present study investigates the biological functions of PTPεM in VSMCs and determines whether PTPεM is implicated in diabetes-accelerated atherosclerosis. We overexpressed wild-type and inactive PTPεM and an small interfering RNA (siRNA) of PTPεM by using an adenovirus vector to investigate the effects of PTPεM upon platelet-derived growth factor (PDGF)- and high glucose (HG)-induced responses of rat VSMCs in vitro. We found that PTPεM decreased PDGF-induced DNA synthesis and migration by reducing the phosphorylation level of the PDGF β-receptor (PDGFRβ) with subsequently suppressed H2O2 generation. The HG content in the medium generated H2O2, upregulated PDGFRβ expression and its tyrosine-phosphorylation, and elevated NADPH oxidase 1 (Nox1) expression even without exogenous PDGF, all of which were downregulated by PTPεM. The PDGFR inhibitor AG1296 also blocked HG-induced Nox1 expression and H2O2 production. Moreover, HG suppressed PTPεM expression itself, which was blocked by the antioxidant N-acetyl-l-cysteine. The effects of PTPεM siRNA were the opposite of those of wild-type PTPεM. Therefore, PTPεM negatively regulates PDGFRβ-mediated signaling pathways that are crucial for the pathogenesis of atherosclerosis, and PTPεM may be involved in diabetes-accelerated atherosclerosis.


2009 ◽  
Vol 28 (22) ◽  
pp. 3564-3578 ◽  
Author(s):  
So-Hee Lim ◽  
Seok-Kyu Kwon ◽  
Myung Kyu Lee ◽  
Jeonghee Moon ◽  
Dae Gwin Jeong ◽  
...  

1999 ◽  
Vol 274 (18) ◽  
pp. 12905-12909 ◽  
Author(s):  
Masato Ogata ◽  
Tsuyoshi Takada ◽  
Yoshiko Mori ◽  
Masatsugu Oh-hora ◽  
Yohzo Uchida ◽  
...  

2020 ◽  
Vol 295 (22) ◽  
pp. 7669-7685 ◽  
Author(s):  
Panfeng Fu ◽  
Ramaswamy Ramchandran ◽  
Mark Shaaya ◽  
Longshuang Huang ◽  
David L. Ebenezer ◽  
...  

Increased permeability of vascular lung tissues is a hallmark of acute lung injury and is often caused by edemagenic insults resulting in inflammation. Vascular endothelial (VE)-cadherin undergoes internalization in response to inflammatory stimuli and is recycled at cell adhesion junctions during endothelial barrier re-establishment. Here, we hypothesized that phospholipase D (PLD)-generated phosphatidic acid (PA) signaling regulates VE-cadherin recycling and promotes endothelial barrier recovery by dephosphorylating VE-cadherin. Genetic deletion of PLD2 impaired recovery from protease-activated receptor-1–activating peptide (PAR-1–AP)-induced lung vascular permeability and potentiated inflammation in vivo. In human lung microvascular endothelial cells (HLMVECs), inhibition or deletion of PLD2, but not of PLD1, delayed endothelial barrier recovery after thrombin stimulation. Thrombin stimulation of HLMVECs increased co-localization of PLD2-generated PA and VE-cadherin at cell-cell adhesion junctions. Inhibition of PLD2 activity resulted in prolonged phosphorylation of Tyr-658 in VE-cadherin during the recovery phase 3 h post-thrombin challenge. Immunoprecipitation experiments revealed that after HLMVECs are thrombin stimulated, PLD2, VE-cadherin, and protein-tyrosine phosphatase nonreceptor type 14 (PTPN14), a PLD2-dependent protein-tyrosine phosphatase, strongly associate with each other. PTPN14 depletion delayed VE-cadherin dephosphorylation, reannealing of adherens junctions, and barrier function recovery. PLD2 inhibition attenuated PTPN14 activity and reversed PTPN14-dependent VE-cadherin dephosphorylation after thrombin stimulation. Our findings indicate that PLD2 promotes PTPN14-mediated dephosphorylation of VE-cadherin and that redistribution of VE-cadherin at adherens junctions is essential for recovery of endothelial barrier function after an edemagenic insult.


2011 ◽  
Vol 22 (19) ◽  
pp. 3609-3624 ◽  
Author(s):  
James R. Whiteford ◽  
Xiaojie Xian ◽  
Claire Chaussade ◽  
Bart Vanhaesebroeck ◽  
Sussan Nourshargh ◽  
...  

Syndecan-2 is a heparan sulfate proteoglycan that has a cell adhesion regulatory domain contained within its extracellular core protein. Cell adhesion to the syndecan-2 extracellular domain (S2ED) is β1 integrin dependent; however, syndecan-2 is not an integrin ligand. Here the protein tyrosine phosphatase receptor CD148 is shown to be a key intermediary in cell adhesion to S2ED, with downstream β1 integrin–mediated adhesion and cytoskeletal organization. We show that S2ED is a novel ligand for CD148 and identify the region proximal to the transmembrane domain of syndecan-2 as the site of interaction with CD148. A mechanism for the transduction of the signal from CD148 to β1 integrins is elucidated requiring Src kinase and potential implication of the C2β isoform of phosphatidylinositol 3 kinase. Our data uncover a novel pathway for β1 integrin–mediated adhesion of importance in cellular processes such as angiogenesis and inflammation.


Sign in / Sign up

Export Citation Format

Share Document