31 research outputs found

    Signaling By Protease-Activated Receptors in Gastrointestinal Smooth Muscle

    Get PDF
    In the present study, we have examined the expression of protease-activated receptors (PARS) and characterized their signaling pathways in rabbit gastric muscle cells. Immunoblot analysis revealed expression of PARl and PAR2 but not PAR3 or PAR4 in smooth muscle. The PARl agonist TFLLR activated Gq, G12, and Gi3, but not Gil, Gi2, G13, Gs or Gz, whereas the PAR2 agonist SLIGRL activated Gq, G13, Gil, and Gi2, but not Gi3, G12, Gs, or Gz. Both PARl and PAR2 agonists stimulated PI hydrolysis and Rho kinase activity and inhibited cAMP formation. PAR1-stimulated PI hydrolysis was abolished in cells expressing Gαq minigene, but was not affected in cells expressing Gαi minigene or in cells treated with pertussis toxin (PTx). PAR2-stimulated PI hydrolysis was partially inhibited in cells expressing Gαq or Gαi minigene and in cells treated with PTx. PAR1- and PAR2-stimulated Rho kinase activity was abolished in cells expressing Gα12 or Gα13 minigene, respectively. Both PARl and PAR2 agonists induced a transient initial contraction that was selectively blocked by the inhibition of PI hydrolysis with U73122 and MLC kinase activity with ML-9. PAR1-induced sustained contraction was preferentially inhibited by the PKC inhibitor bisindolylmaleimide and to a minor extent by the Rho kinase inhibitor Y27632, whereas PAR2-induced sustained contraction was preferentially inhibited by Y27632. Activation of both PARl and PAR2 induced MLC20 phosphorylation, whereas phosphorylation of MYPTl and CPI-17 are receptor-specific: only PARl induced CPI-17 phosphorylation and only PAR2 induced MYPTl phosphorylation.Activation of PARl and PAR2 also induced IκBα degradation and NF-κB activation; the effects were abolished by the blockade of RhoA activity by Clostridium botulinum C3 exoenzyme suggesting NF-κB is downstream of RhoA. PAR1- and PAR2-stimulated Rho kinase activity was significantly augmented by the inhibitors of PKA (PKI), IKK2 (IKKIV), or NF-κB (MG132), and in cells expressing dominant negative mutants of IKK (IKK(K44A), IκBα (IκBα (S32A/S36A)), or phosphorylation-deficient RhoA (RhoA(S188A)). In addition, activation of PARl induced Gα12 phosphorylation, which was abolished by bisindolylmaleimide, suggests that phosphorylation was mediated by PKC derived from the activation of RhoA. Only PAR1-stimulated Rho kinase activity was significantly augmented by the PKC inhibitor. The effect of PKC inhibitor was additive to that of the PKA inhibitor

    Diabetes-induced oxidative stress mediates upregulation of RhoA/Rho kinase pathway and hypercontractility of gastric smooth muscle.

    No full text
    The pathogenesis of diabetes-associated motility disorders are multifactorial and attributed to abnormalities in extrinsic and intrinsic innervation, and a decrease in the number of interstitial cells of Cajal, and nNOS expression and activity. Here we studied the effect of hyperglycemia on smooth muscle function. Using smooth muscles from the fundus of ob/ob mice and of wild type (WT) mice treated with 30 mM glucose (HG), we identified the molecular mechanism by which hyperglycemia upregulates RhoA/Rho kinase pathway and muscle contraction. RhoA expression, Rho kinase activity and muscle contraction were increased, while miR-133a expression was decreased in smooth muscle of ob/ob mice and in smooth muscle treated with HG. Intraperitoneal injections of pre-miR-133a decreased RhoA expression in WT mice and reversed the increase in RhoA expression in ob/ob mice. Intraperitoneal injections of antagomiR-133a increased RhoA expression in WT mice and augmented the increase in RhoA expression in ob/ob mice. The effect of pre-miR-133a or antagomiR-133a in vitro in smooth muscle treated with HG was similar to that obtained in vivo, suggesting that the expression of RhoA is negatively regulated by miR-133a and a decrease in miR-133a expression in diabetes causes an increase in RhoA expression. Oxidative stress (levels of reactive oxygen species and hydrogen peroxide, and expression of superoxide dismutase 1 and NADPH oxidase 4) was increased in smooth muscle of ob/ob mice and in HG-treated smooth muscle. Treatment of ob/ob mice with N-acetylcysteine (NAC) in vivo or addition of NAC in vitro to HG-treated smooth muscle reversed the effect of glucose on the expression of miR-133a and RhoA, Rho kinase activity and muscle contraction. NAC treatment also reversed the decrease in gastric emptying in ob/ob mice. We conclude that oxidative stress in diabetes causes a decrease in miR-133a expression leading to an increase in RhoA/Rho kinase pathway and muscle contraction

    Cross-regulation of VPAC 2

    No full text

    Inhibition of Gα q

    No full text
    corecore