Hyperosmolar glucose induces vasoconstriction through Rho/Rho-kinase pathway in the rat aorta

2011 ◽  
Vol 27 (3) ◽  
pp. 244-251 ◽  
Author(s):  
İsmail Ün ◽  
A. Hakan Kurt ◽  
Kansu Büyükafşar
BMB Reports ◽  
2011 ◽  
Vol 44 (6) ◽  
pp. 415-420 ◽  
Author(s):  
Do-Hyung Kim ◽  
Young-Mi Seok ◽  
In-Kyeom Kim ◽  
In-Kyu Lee ◽  
Seong-Yun Jeong ◽  
...  

2010 ◽  
Vol 399 (2) ◽  
pp. 292-299 ◽  
Author(s):  
Koji Nobe ◽  
Hiromi Nobe ◽  
Hiroko Yoshida ◽  
Michael S. Kolodney ◽  
Richard J. Paul ◽  
...  

2007 ◽  
Vol 75 (1) ◽  
pp. 3-4 ◽  
Author(s):  
S PETERS ◽  
M MICHEL
Keyword(s):  

Cytoskeleton ◽  
2018 ◽  
Vol 75 (11) ◽  
pp. 461-471 ◽  
Author(s):  
Yaprak Dilber Şimay ◽  
Aysun Özdemir ◽  
Burçin İbişoğlu ◽  
Mustafa Ark

2003 ◽  
Vol 285 (5) ◽  
pp. R1145-R1152 ◽  
Author(s):  
Christopher J. Wingard ◽  
Shahid Husain ◽  
Jan Williams ◽  
Sharita James

Maintenance of the detumescent state of the penis is believed to involve the actions of several vasoconstrictors. However, our mechanistic understanding of any synergistic vasoconstrictor influences is extremely limited. We tested the hypothesis that a vasoconstrictor combination of endothelin (ET-1) and phenylephrine (PE) augments the constrictor responses in rat corporal cavernosal tissues by a mechanism involving the RhoA-Rho kinase pathway. Independently, ET-1 (1 nM-30 μM) and PE (100 nM-100 μM) both caused dose-dependent contractions of isolated rat cavernosal tissues. In combination, ET-1 (30 nM) augmented the contractile effect of PE and shifted the calculated EC50 for PE (90 ± 12 to 45 ± 5 μM). The active stress generated by cavernosal strips during the ET-1 + PE combined stimulation (4.9 ± 0.2 mN/mm2) was greater than the combined stress generated with ET-1 (0.4 ± 0.1 mN/mm2) or PE (3.3 ± 0.2 mN/mm2) stimulations alone. Blockade of ETA receptors (30 nM; A-127722) reversed the augmented stress generation and the Rho-kinase inhibitor Y-27632 differentially and dose-dependently relaxed the tissue. The combined constrictor effect was associated with a fourfold increase of RhoA in the membrane faction of the tissue homogenates. We conclude that the ET-1 + PE combination potentiate vasoconstriction through mutual activation of the RhoA-Rho kinase pathway. The interactions of these agonists likely play important roles in the maintenance of the flaccid state and contribute to some forms of erectile dysfunction.


2013 ◽  
Vol 304 (5) ◽  
pp. G527-G535 ◽  
Author(s):  
Senthilkumar Rajagopal ◽  
Divya P. Kumar ◽  
Sunila Mahavadi ◽  
Sayak Bhattacharya ◽  
Ruizhe Zhou ◽  
...  

The present study characterized the TGR5 expression and the signaling pathways coupled to this receptor that mediates the relaxation of gastric smooth muscle. TGR5 was detected in gastric muscle cells by RT-PCR and Western blotting. Treatment of cells with the TGR5-selective ligand oleanolic acid (OA) activated Gαs, but not Gαq, Gαi1, Gαi2, or Gαi3, and increased cAMP levels. OA did not elicit contraction, but caused relaxation of carbachol-induced contraction of gastric muscle cells from wild-type mice, but not tgr5−/− mice. OA, but not a selective exchange protein activated by cAMP (Epac) ligand (8-pCPT-2′-O-Me-cAMP), caused phosphorylation of RhoA and the phosphorylation was blocked by the PKA inhibitor, myristoylated PKI, and by the expression of phosphorylation-deficient mutant RhoA (S188A). Both OA and Epac ligand stimulated Ras-related protein 1 (Rap1) and inhibited carbachol (CCh)-induced Rho kinase activity. Expression of RhoA (S188A) or PKI partly reversed the inhibition of Rho kinase activity by OA but had no effect on inhibition by Epac ligand. However, suppression of Rap1 with siRNA blocked the inhibition of Rho kinase by Epac ligand, and partly reversed the inhibition by OA; the residual inhibition was blocked by PKI. Muscle relaxation in response to OA, but not Epac ligand, was partly reversed by PKI. We conclude that activation of TGR5 causes relaxation of gastric smooth muscle and the relaxation is mediated through inhibition of RhoA/Rho kinase pathway via both cAMP/Epac-dependent stimulation of Rap1 and cAMP/PKA-dependent phosphorylation of RhoA at Ser188. TGR5 receptor activation on smooth muscle reveals a novel mechanism for the regulation of gut motility by bile acids.


Sign in / Sign up

Export Citation Format

Share Document