Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 92
Filter
1.
Biomed J ; 44(3): 245-251, 2021 06.
Article in English | MEDLINE | ID: mdl-34130944

ABSTRACT

COVID-19 is a recent pandemic that is still a major health problem of modern times and already more than 17.5 lakhs people succumbed to this deadly disease. This disease is caused by novel coronavirus which is named SARS-COV-2 by the International Committee on Taxonomy of Viruses. This virus originated from Wuhan city in Hubei province of China in December 2019 and within a short period spread across the many countries in the globe. There are a lot of basic as well as clinical research is going on to study the mode of transmission and the mechanism of action of SARS-COV-2 infection and its therapeutics. SARS-COV-2 is not only known to infect lungs, but it also infects other organs in the human body including the gastrointestinal (GI) tract, the liver, and the pancreas via the angiotensin-converting enzyme (ACE) 2, an important component of the renin-angiotensin system. In this short review, we are mainly discussing the mode of SARS-COV-2 transmission, physiological counterbalancing roles of ACE2 and ACE and the tissue patterns of ACE2 expression, and the overall effect of COVID19 on human gastrointestinal System. Therefore, this review sheds light on the possible mechanism of SARS-COV-2 infection in the GI system and its pathological symptoms raising a potential possibility of GI tract acting as a secondary site for SARS-CoV-2 tropism and infection. Finally, future studies to understand the fecal-oral transmission of the virus and the correlation of viral load and severity of GI symptoms are proposed to gain knowledge of the GI symptoms in COVID-19 to aid in early diagnosis and prognosis.


Subject(s)
COVID-19 , Gastrointestinal Tract/virology , Angiotensin-Converting Enzyme 2 , Humans , Pandemics , SARS-CoV-2 , Viral Tropism
2.
Neurogastroenterol Motil ; 33(2): e13968, 2021 02.
Article in English | MEDLINE | ID: mdl-32789934

ABSTRACT

BACKGROUND: Duchenne muscular dystrophy (DMD) is characterized by the lack of dystrophin in skeletal, cardiac, and smooth muscle. Slow colonic transit and constipation are common in DMD patients and animal models of DMD. However, the cause of this hypocontractility and the expression of contractile proteins in smooth muscle are unknown. The aim of the study was to investigate the expression of contractile proteins in the colonic smooth muscle and the function of the colon in control and mdx mice. METHODS: Muscle contraction was measured in muscle strips and isolated muscle cells. Peristaltic activity was measured in ex vivo preparations by spatiotemporal mapping, and gastrointestinal (GI) transit in vivo was measured by the distribution of fluorescent marker along the intestine and colon. mRNA expression of contractile proteins smoothelin, caldesmon, calponin, and tropomyosin was measured by qRT-PCR. RESULTS: Expression of mRNA for contractile proteins was decreased in colonic smooth muscle of mdx mice compared with control. Contraction in response to acetylcholine and KCl was decreased in colonic muscle strips and in isolated muscle cells of mdx mice. Distension of ex vivo colons with Krebs buffer induced peristalsis in both control and mdx mice; however, significantly fewer full peristaltic waves were recorded in the colons of mdx mice. GI transit was also inhibited in mdx mice. CONCLUSION AND INFERENCES: The data indicate that the lack of dystrophin causes decrease in colonic smooth muscle contractility, peristalsis, and GI transit and provides the basis for analysis of mechanisms involved in smooth muscle dysfunction in DMD.


Subject(s)
Colon/physiopathology , Gastrointestinal Transit/physiology , Muscle, Smooth/physiopathology , Muscular Dystrophy, Duchenne/physiopathology , Peristalsis/physiology , Animals , Colon/metabolism , Male , Mice , Mice, Inbred mdx , Muscle Contraction/physiology , Muscle, Smooth/metabolism , Muscular Dystrophy, Duchenne/metabolism
3.
Inflamm Bowel Dis ; 26(6): 809-819, 2020 05 12.
Article in English | MEDLINE | ID: mdl-32031621

ABSTRACT

BACKGROUND: Endoplasmic reticulum (ER) stress is an essential response of epithelial and immune cells to inflammation in Crohn's disease. The presence and mechanisms that might regulate the ER stress response in subepithelial myofibroblasts (SEMFs) and its role in the development of fibrosis in patients with Crohn's disease have not been examined. METHODS: Subepithelial myofibroblasts were isolated from the affected ileum and normal ileum of patients with each Montreal phenotype of Crohn's disease and from normal ileum in non-Crohn's subjects. Binding of GRP78 to latent TGF-ß1 and its subcellular trafficking was examined using proximity ligation-hybridization assay (PLA). The effects of XBP1 and ATF6 on TGF-ß1 expression were measured using DNA-ChIP and luciferase reporter assay. Endoplasmic reticulum stress components, TGF-ß1, and collagen levels were analyzed in SEMF transfected with siRNA-mediated knockdown of DNMT1 and GRP78 or with DNMT1 inhibitor 5-Azacytidine or with overexpression of miR-199a-5p. RESULTS: In SEMF of strictured ileum from patients with B2 Crohn's disease, expression of ER stress sensors increased significantly. Tunicamycin elicited time-dependent increase in GRP78 protein levels, direct interaction with latent TGF-ß1, and activated TGF-ß1 signaling. The TGFB1 DNA-binding activity of ATF-6α and XBP1 were significantly increased and elicited increased TGFB1 transcription in SEMF-isolated from affected ileum. The levels of ER stress components, TGF-ß1, and collagen expression in SEMF were significantly decreased following knockdown of DNMT1 or GRP78 by 5-Azacytidine treatment or overexpression of miR-199a-5p. CONCLUSIONS: Endoplasmic reticulum stress is present in SEMF of patients susceptible to fibrostenotic Crohn's disease and can contribute to development of fibrosis. Targeting ER stress may represent a novel therapeutic target to prevent fibrosis in patients with fibrostenotic Crohn's disease.


Subject(s)
Crohn Disease/metabolism , Endoplasmic Reticulum Stress/physiology , Intestinal Mucosa/pathology , Myofibroblasts/metabolism , Transforming Growth Factor beta1/metabolism , Adult , Cells, Cultured , Crohn Disease/pathology , Endoplasmic Reticulum Chaperone BiP , Female , Fibrosis , Humans , Ileum/pathology , Male , Middle Aged , Signal Transduction , Young Adult
4.
Neurogastroenterol Motil ; 32(2): e13737, 2020 02.
Article in English | MEDLINE | ID: mdl-31721379

ABSTRACT

BACKGROUND: l-amino acids, such as monosodium glutamate (MSG), activate the umami receptor T1R1/T1R3. We previously showed increased peristalsis in response to activation of T1R1/T1R3 by MSG in mouse colon. However, the expression and function of these receptors in the different regions of the stomach are not clear. METHODS: Mouse gastric smooth muscle cells (SMCs) were isolated and cultured in Dulbecco's Modified Eagle Medium. Expression of T1R1 and T1R3 was measured by RT-PCR and Western blot. The effect of MSG with and without inosine monophosphate (IMP, an allosteric activator of T1R1/T1R3) on acetylcholine (ACh)-induced contraction was measured in muscle strips and isolated SMCs by scanning micrometry. The effect of MSG with or without IMP on activation of G proteins and ACh-induced Ca2+ release was measured in SMCs. KEY RESULTS: Monosodium glutamate inhibited ACh-induced contractions in muscle strips from both antrum and fundus and the effect of MSG was augmented by IMP; the effects were concentration-dependent and not affected by the nitric oxide synthase inhibitor, L-NNA, or tetrodotoxin suggesting a direct effect on SMCs. In isolated gastric SMCs, T1R1 and T1R3 transcripts and protein were identified. Addition of MSG with or without IMP inhibited ACh-induced Ca2+ release and muscle contraction; the effect on contraction was blocked by pertussis toxin suggesting activation of Gαi proteins. MSG in the presence of IMP selectively activated Gαi2 . CONCLUSIONS AND INFERENCES: Umami receptors (T1R1/T1R3) are present on SMCs of the stomach, and activation of these receptors induces muscle relaxation by decreasing [Ca2+ ]i via Gαi2 .


Subject(s)
Muscle, Smooth/metabolism , Receptors, G-Protein-Coupled/metabolism , Stomach , Animals , Female , Male , Mice , Mice, Inbred C57BL
5.
Cell Tissue Res ; 378(3): 471-483, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31410629

ABSTRACT

Regulation of gut function depends on the detection and response to luminal contents. Luminal L-amino acids (L-AA) are detected by several receptors including metabotropic glutamate receptors 1 and 4 (mGluR1 and mGluR4), calcium-sensing receptor (CaSR), GPRC family C group 6 subtype A receptor (GPRC6A) and umami taste receptor heterodimer T1R1/T1R3. Here, we show that murine mucosal homogenates and STC-1 cells, a murine enteroendocrine cell line, express mRNA for all L-AA receptors. Immunohistochemical analysis demonstrated the presence of all L-AA receptors on STC-1 with CaSR being most commonly expressed and T1R1 least expressed (35% versus 15% of cells); mGluRs and GPRC6a were intermediate (~ 20% of cells). Regarding coexpression of L-AA receptors, the mGluRs and T1R1 were similarly coexpressed with CaSR (10-12% of cells) whereas GPRC6a was coexpressed least (7% of cells). mGluR1 was coexpressed with GPRC6a in 11% of cells whereas coexpression between other receptors was less (2-8% of cells). CaSR and mGluR1 were coexpressed with glucagon-like peptide-1 (GLP-1) and peptide YY (PYY) in 20-25% of cells whereas T1R1 and GPRC6a were coexpressed with GLP-1 and PYY less (8-12% of cells). Only mGluR4 showed differential coexpression with GLP-1 (13%) and PYY (21%). L-Phenylalanine (10 mM) caused a 3-fold increase in GLP-1 release, which was strongly inhibited by siRNA to CaSR indicating functional coupling of CaSR to GLP-1 release. The results suggest that not all STC-1 cells express (and coexpress) L-AA receptors to the same extent and that the pattern of response likely depends on the pattern of expression of L-AA receptors.


Subject(s)
Colon , Enteroendocrine Cells/metabolism , Intestine, Small , Receptors, Amino Acid/metabolism , Animals , Cell Line , Colon/cytology , Colon/metabolism , Enteroendocrine Cells/cytology , Intestine, Small/cytology , Intestine, Small/metabolism , Mice , Mice, Inbred C57BL
6.
Neurogastroenterol Motil ; 31(2): e13495, 2019 02.
Article in English | MEDLINE | ID: mdl-30393912

ABSTRACT

BACKGROUND: Actin polymerization plays an important role in smooth muscle contraction. Integrin-linked kinase (ILK) was shown to mediate actin polymerization in airway smooth muscle. The role of ILK in actin polymerization in response to m2 receptor activation was not in gastric smooth muscle. METHODS: Phosphorylation of paxillin, neuronal Wiskott-Aldrich syndrome protein (N-WASp), and association of paxillin with GEF proteins (Cool2/αPix [Cool2/PAK-interacting exchange factor alpha], Cool1/ßPix [Cool1/PAK-interacting exchange factor beta], and DOCK 180 [Dedicator of cytokinesis]) and N-WASp with Arp2/3 complex were measured by western blot. Activation of Cdc42 was determined using an antibody for activated Cdc42. Actin polymerization was measured as an increase in F-actin/G-actin ratio. RESULTS: Phosphorylation of paxillin, an association of paxillin with GEF proteins, Cdc42 activity, and actin polymerization were increased in response to m2 receptor activation in gastric smooth muscle cells. The increases in paxillin phosphorylation, Cdc42 activity, and actin polymerization were inhibited by a PI3Kγ inhibitor (AS-605240), ILK siRNA, and ILK dominant negative mutant (ILK [R211]). Increase in actin polymerization was also inhibited by Cdc42 dominant negative mutant (Cdc42 [T17N]). Increases in the association of paxillin with GEF proteins, phosphorylation of N-WASp and its association with Arp2/3 complex were inhibited by ILK (R211). CONCLUSION: In gastric smooth muscle cells, activation of PI3Kγ by muscarinic m2 receptors causes ILK-dependent phosphorylation of paxillin, an association of paxillin with Cdc42 GEF proteins and activation of Cdc42, which, in turn, causes phosphorylation of N-WASp and its association with Arp2/3 complex leading to actin polymerization.


Subject(s)
Actins/metabolism , Muscle, Smooth/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Receptor, Muscarinic M2/metabolism , Stomach/physiology , Animals , Muscle Contraction/physiology , Polymerization , Rabbits
7.
Dig Dis Sci ; 64(5): 1171-1181, 2019 05.
Article in English | MEDLINE | ID: mdl-30560338

ABSTRACT

BACKGROUND: Isovaleric acid (IVA) is a 5-carbon branched-chain fatty acid present in fermented foods and produced in the colon by bacterial fermentation of leucine. We previously reported that the shorter, straight-chain fatty acids acetate, propionate and butyrate differentially affect colonic motility; however, the effect of branched-chain fatty acids on gut smooth muscle and motility is unknown. AIMS: To determine the effect of IVA on contractility of colonic smooth muscle. METHODS: Murine colonic segments were placed in a longitudinal orientation in organ baths in Krebs buffer and fastened to force transducers. Segments were contracted with acetylcholine (ACh), and the effects of IVA on ACh-induced contraction were measured in the absence and presence of tetrodotoxin (TTx) or inhibitors of nitric oxide synthase [L-N-nitroarginine (L-NNA)] or adenylate cyclase (SQ22536). The effect of IVA on ACh-induced contraction was also measured in isolated muscle cells in the presence or absence of SQ22536 or protein kinase A (PKA) inhibitor (H-89). Direct activation of PKA was measured in isolated muscle cells. RESULTS: In colonic segments, ACh-induced contraction was inhibited by IVA in a concentration-dependent fashion; the IVA response was not affected by TTx or L-NNA but inhibited by SQ22536. Similarly, in isolated colonic muscle cells, ACh-induced contraction was inhibited by IVA in a concentration-dependent fashion and the effect blocked by SQ22536 and H-89. IVA also increased PKA activity in isolated smooth muscle cells. CONCLUSIONS: The branched-chain fatty acid IVA acts directly on colonic smooth muscle and causes muscle relaxation via the PKA pathway.


Subject(s)
Colon/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic AMP/metabolism , Fatty Acids, Volatile/pharmacology , Muscle Relaxation/physiology , Muscle, Smooth/metabolism , Pentanoic Acids/pharmacology , Animals , Colon/drug effects , Dose-Response Relationship, Drug , Female , Hemiterpenes , Male , Mice , Mice, Inbred C57BL , Muscle Relaxation/drug effects , Muscle, Smooth/drug effects , Organ Culture Techniques , Signal Transduction/drug effects , Signal Transduction/physiology
8.
Peptides ; 112: 48-55, 2019 02.
Article in English | MEDLINE | ID: mdl-30508636

ABSTRACT

The insulinotropic effects of the incretin hormone, glucagon-like peptide-1 (GLP-1) are mediated via GLP-1 receptors (GLP-1R) present on pancreatic ß cells. GLP-1 causes a decrease in the motility of stomach and intestine which involves both central and peripheral nervous systems. The expression and function of GLP-1R in gastrointestinal smooth muscle, however, are not clear. Muscle strips and isolated muscle cells were prepared from mouse colon and the effect of GLP-1(7-36) amide on acetylcholine (ACh)-induced contraction was measured. Muscle cells in culture were used to identify the expression of GLP-1R and the signaling pathways activated by GLP-1(7-36) amide. GLP-1R was expressed in the mucosal and non-mucosal tissue preparations derived from colon, and in smooth muscle cell cultures devoid of other cells such as enteric neurons. In colonic muscle strips, the addition of GLP-1(7-36) amide caused dose-dependent inhibition of acetylcholine-induced contractions. The effect of GLP-1(7-36) amide was partly inhibited by the neuronal blocker tetrodotoxin and nitric oxide (NO) synthase inhibitor l-NNA suggesting both NO-dependent neural and NO-independent direct effects on smooth muscle. In isolated colonic smooth muscle cells, GLP-1(7-36) amide caused an increase in Gαs activity, cAMP levels, and PKA activity, and inhibited ACh-induced contraction. The effect of GLP-1(7-36) amide on Gαs activity and cAMP levels was blocked by NF449, an inhibitor of Gαs, and the effect of GLP-1(7-36) amide on contraction was blocked by NF449 and myristoylated PKI, an inhibitor of PKA. We conclude that colonic smooth muscle cells express GLP-1R, and GLP-1(7-36) amide inhibits acetylcholine-induced contraction via GLP-1R coupled to the Gαs/cAMP/PKA pathway.


Subject(s)
Acetylcholine/metabolism , Glucagon-Like Peptide-1 Receptor/metabolism , Muscle Contraction , Muscle, Smooth/metabolism , Signal Transduction , Animals , Colon/metabolism , Colon/physiology , Cyclic AMP/metabolism , Gene Expression , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide-1 Receptor/genetics , Glucagon-Like Peptide-1 Receptor/physiology , Mice , Muscle, Smooth/physiology
9.
PLoS One ; 13(12): e0209359, 2018.
Article in English | MEDLINE | ID: mdl-30571746

ABSTRACT

In gastrointestinal smooth muscle, acetylcholine induced muscle contraction is biphasic, initial peak followed by sustained contraction. Contraction is regulated by phosphorylation of 20 kDa myosin light chain (MLC) at Ser19, interaction of actin and myosin, and actin polymerization. The present study characterized the signaling mechanisms involved in actin polymerization during initial and sustained muscle contraction in response to muscarinic M3 receptor activation in gastric smooth muscle cells by targeting the effectors of initial (phospholipase C (PLC)-ß/Ca2+ pathway) and sustained (RhoA/focal adhesion kinase (FAK)/Rho kinase pathway) contraction. The initial Ca2+ dependent contraction and actin polymerization is mediated by sequential activation of PLC-ß1 via Gαq, IP3 formation, Ca2+ release and Ca2+ dependent phosphorylation of proline-rich-tyrosine kinase 2 (Pyk2) at Tyr402. The sustained Ca2+ independent contraction and actin polymerization is mediated by activation of RhoA, and phosphorylation of FAK at Tyr397. Both phosphorylation of Pyk2 and FAK leads to phosphorylation of paxillin at Tyr118 and association of phosphorylated paxillin with the GEF proteins p21-activated kinase (PAK) interacting exchange factor α, ß (α and ß PIX) and DOCK 180. These GEF proteins stimulate Cdc42 leading to the activation of nucleation promoting factor N-WASP (neuronal Wiskott-Aldrich syndrome protein), which interacts with actin related protein complex 2/3 (Arp2/3) to induce actin polymerization and muscle contraction. Acetylcholine induced muscle contraction is inhibited by actin polymerization inhibitors. Thus, our results suggest that a novel mechanism for the regulation of smooth muscle contraction is mediated by actin polymerization in gastrointestinal smooth muscle which is independent of MLC20 phosphorylation.


Subject(s)
Actins/metabolism , Muscle Contraction/physiology , Muscle, Smooth/physiology , Protein Multimerization/physiology , Stomach/physiology , Animals , Calcium/metabolism , Cations, Divalent/metabolism , Mice , Mice, Inbred C57BL , Myosin Light Chains/metabolism , Phosphorylation/physiology , Rabbits , Receptor, Muscarinic M3 , Signal Transduction/physiology
10.
Article in English | MEDLINE | ID: mdl-28971603

ABSTRACT

Hydrogen sulfide (H2 S) plays an important role in smooth muscle relaxation. Here, we investigated the expression of enzymes in H2 S synthesis and the mechanism regulating colonic smooth muscle function by H2 S. Expression of cystathionine-γ-lyase (CSE), but not cystathionine-ß-synthase (CBS), was identified in the colonic smooth muscle of rabbit, mouse, and human. Carbachol (CCh)-induced contraction in rabbit muscle strips and isolated muscle cells was inhibited by l-cysteine (substrate of CSE) and NaHS (an exogenous H2 S donor) in a concentration-dependent fashion. H2 S induced S-sulfhydration of RhoA that was associated with inhibition of RhoA activity. CCh-induced Rho kinase activity also was inhibited by l-cysteine and NaHS in a concentration-dependent fashion. Inhibition of CCh-induced contraction by l-cysteine was blocked by the CSE inhibitor, dl-propargylglycine (DL-PPG) in dispersed muscle cells. Inhibition of CCh-induced Rho kinase activity by l-cysteine was blocked by CSE siRNA in cultured cells and DL-PPG in dispersed muscle cells. Stimulation of Rho kinase activity and muscle contraction in response to CCh was also inhibited by l-cysteine or NaHS in colonic muscle cells from mouse and human. Collectively, our studies identified the expression of CSE in colonic smooth muscle and determined that sulfhydration of RhoA by H2 S leads to inhibition of RhoA and Rho kinase activities and muscle contraction. The mechanism identified may provide novel therapeutic approaches to mitigate gastrointestinal motility disorders.


Subject(s)
Colon/cytology , Cystathionine gamma-Lyase/metabolism , Hydrogen Sulfide/pharmacology , Myocytes, Smooth Muscle/metabolism , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/metabolism , Animals , Cells, Cultured , Colon/drug effects , Colon/metabolism , Cystathionine beta-Synthase/metabolism , Down-Regulation , Humans , Mice , Muscle Contraction/drug effects , Muscle, Smooth/cytology , Muscle, Smooth/drug effects , Muscle, Smooth/metabolism , Myocytes, Smooth Muscle/drug effects , Rabbits , Signal Transduction/drug effects , Sulfides/metabolism
11.
J Sex Med ; 14(10): 1177-1186, 2017 10.
Article in English | MEDLINE | ID: mdl-28923309

ABSTRACT

BACKGROUND: The pathophysiology of increased severity of erectile dysfunction in men with diabetes and their poor response to oral pharmacotherapy are unclear. Defective vascular endothelium and consequent impairment in the formation and action of nitric oxide (NO) are implicated as potential mechanisms. Endothelial NO synthase, critical for NO generation, is localized to caveolae, plasma membrane lipid rafts enriched in structural proteins, and caveolins. Type 2 diabetes mellitus (T2DM)-induced changes in caveolin expression are recognized to play a role in cardiovascular dysfunction. AIMS: To evaluate DM-related changes to male erectile tissue in a mouse model that closely resembles human T2DM and study the specific role of caveolins in penile blood flow and microvascular perfusion using mice lacking caveolin (Cav)-1 or Cav-3. METHODS: We used wild-type C57BL6 (control) and Cav-1 and Cav-3 knockout (KO) male mice. T2DM was induced by streptozotocin followed by a high-fat diet for 4 months. Penile expressions of Cav-1, Cav-3, and endothelial NO synthase were determined by western blot, and phosphodiesterase type 5 activity was measured using [3H] cyclic guanosine monophosphate as a substrate. For hemodynamic studies, Cav-1 and Cav-3 KO mice were anesthetized, and penile blood flow (peak systolic velocity and end-diastolic velocity; millimeters per second) was determined using a high-frequency and high-resolution digital imaging color Doppler system. Penile tissue microcirculatory blood perfusion (arbitrary perfusion units) was measured using a novel PeriCam PSI system. OUTCOMES: Penile erectile tissues were harvested for histologic studies to assess Cav-1, Cav-3, and endothelial NO synthase expression, phosphodiesterase type 5 activity, and blood flow, and perfusion measurements were assessed for hemodynamic studies before and after an intracavernosal injection of prostaglandin E1 (50 ng). RESULTS: In T2DM mice, decreased Cav-1 and Cav-3 penile protein expression and increased phosphodiesterase type 5 activity were observed. Decreased response to prostaglandin E1 in peak systolic velocity (33 ± 4 mm/s in Cav-1 KO mice vs 62 ± 5 mm/s in control mice) and perfusion (146 ± 12 AU in Cav-1 KO mice vs 256 ± 12 AU in control mice) was observed. Hemodynamic changes in Cav-3 KO mice were insignificant. CLINICAL TRANSLATION: Our findings provide novel mechanistic insights into erectile dysfunction severity and poor pharmacotherapy that could have potential application to patients with T2DM. STRENGTHS AND LIMITATIONS: Use of KO mice and novel hemodynamic techniques are the strengths. A limitation is the lack of direct evaluation of penile hemodynamics in T2DM mice. CONCLUSION: Altered penile Cav-1 expression in T2DM mice and impaired penile hemodynamics in Cav-1 KO mice suggests a regulatory role for Cav-1 in DM-related erectile dysfunction. Parikh J, Zemljic-Harpf A, Fu J, et al. Altered Penile Caveolin Expression in Diabetes: Potential Role in Erectile Dysfunction. J Sex Med 2017;14:1177-1186.


Subject(s)
Caveolin 1/metabolism , Diabetes Mellitus, Type 2/complications , Erectile Dysfunction/metabolism , Nitric Oxide Synthase Type III/metabolism , Animals , Cyclic GMP/metabolism , Diabetes Mellitus, Type 2/metabolism , Endothelium, Vascular/metabolism , Male , Mice , Mice, Knockout , Microcirculation , Penile Erection/physiology , Penis/blood supply
12.
Am J Physiol Gastrointest Liver Physiol ; 313(4): G330-G341, 2017 Oct 01.
Article in English | MEDLINE | ID: mdl-28705807

ABSTRACT

Hydrogen sulfide (H2S), like nitric oxide (NO), causes smooth muscle relaxation, but unlike NO, does not stimulate soluble guanylyl cyclase (sGC) activity and generate cyclic guanosine 5'-monophosphate (cGMP). The aim of this study was to investigate the interplay between NO and H2S in colonic smooth muscle. In colonic smooth muscle from rabbit, mouse, and human, l-cysteine, substrate of cystathionine-γ-lyase (CSE), or NaHS, an H2S donor, inhibited phosphodiesterase 5 (PDE5) activity and augmented the increase in cGMP levels, IP3 receptor phosphorylation at Ser1756 (measured as a proxy for PKG activation), and muscle relaxation in response to NO donor S-nitrosoglutathione (GSNO), suggesting augmentation of cGMP/PKG pathway by H2S. The inhibitory effect of l-cysteine, but not NaHS, on PDE5 activity was blocked in cells transfected with CSE siRNA or treated with CSE inhibitor d,l-propargylglycine (dl-PPG), suggesting activation of CSE and generation of H2S in response to l-cysteine. H2S levels were increased in response to l-cysteine, and the effect of l-cysteine was augmented by GSNO in a cGMP-dependent protein kinase-sensitive manner, suggesting augmentation of CSE/H2S by cGMP/PKG pathway. As a result, GSNO-induced relaxation was inhibited by dl-PPG. In flat-sheet preparation of colon, l-cysteine augmented calcitonin gene-related peptide release in response to mucosal stimulation, and in intact segments, l-cysteine increased the velocity of pellet propulsion. These results demonstrate that in colonic smooth muscle, there is a novel interplay between NO and H2S. NO generates H2S via cGMP/PKG pathway, and H2S, in turn, inhibits PDE5 activity and augments NO-induced cGMP levels. In the intact colon, H2S promotes colonic transit.NEW & NOTEWORTHY Hydrogen sulfide (H2S) and nitric oxide (NO) are important regulators of gastrointestinal motility. The studies herein provide the cross talk between NO and H2S signaling to mediate smooth muscle relaxation and colonic transit. H2S inhibits phosphodiesterase 5 activity to augment cGMP levels in response to NO, which, in turn, via cGMP/PKG pathway, generates H2S. These studies suggest that interventions targeted at restoring NO and H2S homeostasis within the smooth muscle may provide novel therapeutic approaches to mitigate motility disorders.


Subject(s)
Colon/physiology , Cyclic GMP-Dependent Protein Kinases/metabolism , Cyclic GMP/metabolism , Hydrogen Sulfide/metabolism , Muscle Contraction/physiology , Muscle, Smooth/physiology , Nitric Oxide/metabolism , Animals , Female , Gastrointestinal Motility , Humans , Male , Mice , Mice, Inbred C57BL , Rabbits , Signal Transduction/physiology , Species Specificity , Up-Regulation/physiology
13.
PLoS One ; 12(7): e0178574, 2017.
Article in English | MEDLINE | ID: mdl-28678840

ABSTRACT

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.


Subject(s)
Muscle Contraction/physiology , Muscle, Smooth/physiology , Oxidative Stress , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/metabolism , Acetylcysteine/pharmacology , Animals , Blotting, Western , Cells, Cultured , Diabetes Mellitus/genetics , Diabetes Mellitus/metabolism , Diabetes Mellitus/physiopathology , Free Radical Scavengers/pharmacology , Gastric Mucosa/metabolism , Gene Expression/drug effects , Glucose/pharmacology , Hyperglycemia/genetics , Hyperglycemia/metabolism , Hyperglycemia/physiopathology , Mice, Inbred C57BL , Mice, Obese , MicroRNAs/genetics , Muscle, Smooth/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics , Stomach/physiology , Up-Regulation , rho-Associated Kinases/genetics , rhoA GTP-Binding Protein/genetics
14.
PLoS One ; 12(2): e0171335, 2017.
Article in English | MEDLINE | ID: mdl-28192441

ABSTRACT

During postnatal development rats demonstrate an age-dependent increase in NaCl chorda tympani (CT) responses and the number of functional apical amiloride-sensitive epithelial Na+ channels (ENaCs) in salt sensing fungiform (FF) taste receptor cells (TRCs). Currently, the intracellular signals that regulate the postnatal development of salt taste have not been identified. We investigated the effect of cAMP, a downstream signal for arginine vasopressin (AVP) action, on the postnatal development of NaCl responses in 19-23 day old rats. ENaC-dependent NaCl CT responses were monitored after lingual application of 8-chlorophenylthio-cAMP (8-CPT-cAMP) under open-circuit conditions and under ±60 mV lingual voltage clamp. Behavioral responses were tested using 2 bottle/24h NaCl preference tests. The effect of [deamino-Cys1, D-Arg8]-vasopressin (dDAVP, a specific V2R agonist) was investigated on ENaC subunit trafficking in rat FF TRCs and on cAMP generation in cultured adult human FF taste cells (HBO cells). Our results show that in 19-23 day old rats, the ENaC-dependent maximum NaCl CT response was a saturating sigmoidal function of 8-CPT-cAMP concentration. 8-CPT-cAMP increased the voltage-sensitivity of the NaCl CT response and the apical Na+ response conductance. Intravenous injections of dDAVP increased ENaC expression and γ-ENaC trafficking from cytosolic compartment to the apical compartment in rat FF TRCs. In HBO cells dDAVP increased intracellular cAMP and cAMP increased trafficking of γ- and δ-ENaC from cytosolic compartment to the apical compartment 10 min post-cAMP treatment. Control 19-23 day old rats were indifferent to NaCl, but showed clear preference for appetitive NaCl concentrations after 8-CPT-cAMP treatment. Relative to adult rats, 14 day old rats demonstrated significantly less V2R antibody binding in circumvallate TRCs. We conclude that an age-dependent increase in V2R expression produces an AVP-induced incremental increase in cAMP that modulates the postnatal increase in TRC ENaC and the neural and behavioral responses to NaCl.


Subject(s)
Chorda Tympani Nerve/drug effects , Cyclic AMP/pharmacology , Sodium Chloride/pharmacology , Taste/drug effects , Adult , Age Factors , Animals , Blotting, Western , Cells, Cultured , Chorda Tympani Nerve/physiology , Cyclic AMP/analogs & derivatives , Cyclic AMP/metabolism , Deamino Arginine Vasopressin/pharmacology , Epithelial Sodium Channels/genetics , Epithelial Sodium Channels/metabolism , Food Preferences/drug effects , Food Preferences/physiology , Gene Expression/drug effects , Humans , Microscopy, Confocal , Rats, Sprague-Dawley , Receptors, Vasopressin/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Taste/physiology , Taste Buds/drug effects , Taste Buds/metabolism , Taste Buds/physiology , Thionucleotides/metabolism , Thionucleotides/pharmacology
15.
Biochem Biophys Res Commun ; 483(3): 923-929, 2017 02 12.
Article in English | MEDLINE | ID: mdl-27746176

ABSTRACT

GATA transcription factors regulate an array of genes important in cell proliferation and differentiation. Here we report the identification of regulator of G protein signaling 4 (RGS4) as a novel target for GATA-6 transcription factor. Although three sites (a, b, c) within the proximal region of rabbit RGS4 promoter for GATA transcription factors were predicted by bioinformatics analysis, only GATA-a site (16 bp from the core TATA box) is essential for RGS4 transcriptional regulation. RT-PCR analysis demonstrated that only GATA-6 was highly expressed in rabbit colonic smooth muscle cells but GATA-4/6 were expressed in cardiac myocytes and GATA-1/2/3 expressed in blood cells. Adenovirus-mediated expression of GATA-6 but not GATA-1 significantly increased the constitutive and IL-1ß-induced mRNA expression of the endogenous RGS4 in colonic smooth muscle cells. IL-1ß stimulation induced GATA-6 nuclear translocation and increased GATA-6 binding to RGS4 promoter. These data suggest that GATA factor could affect G protein signaling through regulating RGS4 expression, and GATA signaling may develop as a future therapeutic target for RGS4-related diseases.


Subject(s)
GATA6 Transcription Factor/metabolism , RGS Proteins/metabolism , Active Transport, Cell Nucleus/drug effects , Animals , Base Sequence , Binding Sites/genetics , Cells, Cultured , Colon/cytology , Colon/drug effects , Colon/metabolism , GATA1 Transcription Factor/genetics , GATA1 Transcription Factor/metabolism , GATA6 Transcription Factor/genetics , Gene Expression Regulation/drug effects , Interleukin-1beta/metabolism , Interleukin-1beta/pharmacology , Mutagenesis, Site-Directed , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Promoter Regions, Genetic , RGS Proteins/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rabbits , Signal Transduction
16.
PLoS One ; 11(11): e0166565, 2016.
Article in English | MEDLINE | ID: mdl-27846263

ABSTRACT

In addition to the T2R bitter taste receptors, neuronal nicotinic acetylcholine receptors (nAChRs) have recently been shown to be involved in the bitter taste transduction of nicotine, acetylcholine and ethanol. However, at present it is not clear if nAChRs are expressed in enteroendocrine cells other than beta cells of the pancreas and enterochromaffin cells, and if they play a role in the synthesis and release of neurohumoral peptides. Accordingly, we investigated the expression and functional role of nAChRs in enteroendocrine STC-1 cells. Our studies using RT-PCR, qRT-PCR, immunohistochemical and Western blotting techniques demonstrate that STC-1 cells express several α and ß nAChR subunits. Exposing STC-1 cells to nicotine acutely (24h) or chronically (4 days) induced a differential increase in the expression of nAChR subunit mRNA and protein in a dose- and time-dependent fashion. Mecamylamine, a non-selective antagonist of nAChRs, inhibited the nicotine-induced increase in mRNA expression of nAChRs. Exposing STC-1 cells to nicotine increased intracellular Ca2+ in a dose-dependent manner that was inhibited in the presence of mecamylamine or dihydro-ß-erythroidine, a α4ß2 nAChR antagonist. Brain-derived neurotrophic factor (BDNF) mRNA and protein were detected in STC-1 cells using RT-PCR, specific BDNF antibody, and enzyme-linked immunosorbent assay. Acute nicotine exposure (30 min) decreased the cellular content of BDNF in STC-1 cells. The nicotine-induced decrease in BDNF was inhibited in the presence of mecamylamine. We also detected α3 and ß4 mRNA in intestinal mucosal cells and α3 protein expression in intestinal enteroendocrine cells. We conclude that STC-1 cells and intestinal enteroendocrine cells express nAChRs. In STC-1 cells nAChR expression is modulated by exposure to nicotine in a dose- and time-dependent manner. Nicotine interacts with nAChRs and inhibits BDNF expression in STC-1 cells.


Subject(s)
Brain-Derived Neurotrophic Factor/genetics , Nicotine/metabolism , Receptors, G-Protein-Coupled/biosynthesis , Receptors, Nicotinic/biosynthesis , Animals , Brain-Derived Neurotrophic Factor/antagonists & inhibitors , Brain-Derived Neurotrophic Factor/biosynthesis , Calcium/metabolism , Cell Line , Dihydro-beta-Erythroidine/administration & dosage , Enterochromaffin Cells/metabolism , Enteroendocrine Cells/metabolism , Gene Expression Regulation/drug effects , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Mecamylamine/administration & dosage , Mecamylamine/metabolism , Mice , Nicotine/administration & dosage , Nicotine/antagonists & inhibitors , RNA, Messenger/biosynthesis , Receptors, G-Protein-Coupled/genetics , Receptors, Nicotinic/genetics
17.
Dig Dis Sci ; 61(7): 1925-40, 2016 07.
Article in English | MEDLINE | ID: mdl-26879904

ABSTRACT

BACKGROUND: The contractility of colonic smooth muscle is dysregulated due to immune/inflammatory responses in inflammatory bowel diseases. Inflammation in vitro induces up-regulation of regulator of G-protein signaling 4 (RGS4) expression in colonic smooth muscle cells. AIMS: To characterize the immune/inflammatory responses and RGS4 expression pattern in colonic smooth muscle after induction of colitis. METHODS: Colitis was induced in rabbits by intrarectal instillation of 2,4,6-trinitrobenzene sulfonic acid (TNBS). Innate/adaptive immune response RT-qPCR array was performed using colonic circular muscle strips. At 1-9 weeks after colonic intramuscular microinjection of lentivirus, the distal and proximal colons were collected, and muscle strips and dispersed muscle cells were prepared from circular muscle layer. Expression levels of RGS4 and NFκB signaling components were determined by Western blot analysis. The biological consequences of RGS4 knockdown were assessed by measurement of muscle contraction and phospholipase C (PLC)-ß activity in response to acetylcholine (ACh). RESULTS: Contraction in response to ACh was significantly inhibited in the inflamed colonic circular smooth muscle cells. RGS4, IL-1, IL-6, IL-8, CCL3, CD1D, and ITGB2 were significantly up-regulated, while IL-18, CXCR4, CD86, and C3 were significantly down-regulated in the inflamed muscle strips. RGS4 protein expression in the inflamed smooth muscles was dramatically increased. RGS4 stable knockdown in vivo augmented ACh-stimulated PLC-ß activity and contraction in colonic smooth muscle cells. CONCLUSION: Inflamed smooth muscle exhibits up-regulation of IL-1-related signaling components, Th1 cytokines and RGS4, and inhibition of contraction. Stable knockdown of endogenous RGS4 in colonic smooth muscle increases PLC-ß activity and contractile responses.


Subject(s)
Colon/drug effects , Inflammation/chemically induced , Muscle Contraction/immunology , Muscle, Smooth/pathology , Trinitrobenzenesulfonic Acid/toxicity , Animals , Cytokines/genetics , Cytokines/metabolism , Gene Expression Regulation/immunology , Inflammation/pathology , Rabbits , Up-Regulation
18.
J Biol Chem ; 291(13): 6626-40, 2016 Mar 25.
Article in English | MEDLINE | ID: mdl-26757816

ABSTRACT

The physiological role of the TGR5 receptor in the pancreas is not fully understood. We previously showed that activation of TGR5 in pancreatic ß cells by bile acids induces insulin secretion. Glucagon released from pancreatic α cells and glucagon-like peptide 1 (GLP-1) released from intestinal L cells regulate insulin secretion. Both glucagon and GLP-1 are derived from alternate splicing of a common precursor, proglucagon by PC2 and PC1, respectively. We investigated whether TGR5 activation in pancreatic α cells enhances hyperglycemia-induced PC1 expression thereby releasing GLP-1, which in turn increases ß cell mass and function in a paracrine manner. TGR5 activation augmented a hyperglycemia-induced switch from glucagon to GLP-1 synthesis in human and mouse islet α cells by GS/cAMP/PKA/cAMP-response element-binding protein-dependent activation of PC1. Furthermore, TGR5-induced GLP-1 release from α cells was via an Epac-mediated PKA-independent mechanism. Administration of the TGR5 agonist, INT-777, to db/db mice attenuated the increase in body weight and improved glucose tolerance and insulin sensitivity. INT-777 augmented PC1 expression in α cells and stimulated GLP-1 release from islets of db/db mice compared with control. INT-777 also increased pancreatic ß cell proliferation and insulin synthesis. The effect of TGR5-mediated GLP-1 from α cells on insulin release from islets could be blocked by GLP-1 receptor antagonist. These results suggest that TGR5 activation mediates cross-talk between α and ß cells by switching from glucagon to GLP-1 to restore ß cell mass and function under hyperglycemic conditions. Thus, INT-777-mediated TGR5 activation could be leveraged as a novel way to treat type 2 diabetes mellitus.


Subject(s)
Cholic Acids/pharmacology , Diabetes Mellitus, Experimental/genetics , Glucagon-Like Peptide 1/metabolism , Glucose/metabolism , Paracrine Communication/genetics , Receptors, G-Protein-Coupled/agonists , Animals , Benzene Derivatives/pharmacology , Benzenesulfonates/pharmacology , Cell Line , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Estrenes/pharmacology , Gene Expression Regulation , Glucagon-Like Peptide 1/biosynthesis , Glucagon-Like Peptide 1/genetics , Glucagon-Secreting Cells/drug effects , Glucagon-Secreting Cells/metabolism , Glucagon-Secreting Cells/pathology , Homeostasis/drug effects , Humans , Insulin Resistance , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Proprotein Convertase 1/genetics , Proprotein Convertase 1/metabolism , Proprotein Convertase 2/genetics , Proprotein Convertase 2/metabolism , Pyrrolidinones/pharmacology , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Sulfones/pharmacology
19.
J Immunol ; 194(7): 3422-31, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25740948

ABSTRACT

Increased TGF-ß1 and TGF-ß1-dependent Collagen I production in intestinal mesenchymal cells result in fibrosis in patients with Montreal B2 fibrostenotic Crohn's disease. Numerous cytokines, including IL-6, are produced by activated mesenchymal cells themselves and activate STAT3. The aim of the current study was to determine the mechanisms by which STAT-3 activation might result in intestinal fibrosis. Cytokine levels were measured by ELISA. STAT3 and suppressor of cytokine signaling 3 protein levels were measured by immunoblot, STAT3-TGFB1 DNA-binding activity by chromatin immunoprecipitation, and TGFB1 transcriptional activity by luciferase reporter assay. TGF-ß1 (TGFB1), Collagen1α1, and connective tissue growth factor (CTGF) gene expression was measured by quantitative RT-PCR. The role of STAT3 activation was determined using STAT3 inhibitor, Stattic, and by transfection of STAT3 mutants. Autocrine production of cytokines was increased in muscle cells of B2 phenotype patients from strictures and normal intestine in the same patient and compared with other Crohn's phenotypes, ulcerative colitis, and non-Crohn's patients. A unique pattern of STAT3 phosphorylation emerged: high STAT3(S727) and low STAT3(Y705) in strictures and the opposite in unaffected intestine. TGFB1 transcriptional activity was regulated by phospho-STAT3(S727) and was decreased by Stattic or dominant-negative STAT3(S727A). TGF-ß1, COL1A1, and CTGF expression was inhibited by Stattic or dominant-negative STAT3(S727A). Treatment of normal muscle cells with IL-6 or expression of constitutively active STAT3(S727E) phenocopied muscle cells from strictured intestine. Neutralization of autocrine IL-6 reversed STAT3 phosphorylation and normalized expression of TGF-ß1 in strictured intestinal muscle. The ability of Stattic to improve development of fibrosis was confirmed in mice with 2,4,6-trinitrobenzenesulfonic acid-induced colitis. We observed a unique phospho-STAT3(S727) response in patients with Montreal B2 Crohn's disease, particularly in response to IL-6 leading to increased TGF-ß1, collagen, and CTGF production in ileal strictures.


Subject(s)
Collagen Type I/genetics , Crohn Disease/genetics , Crohn Disease/metabolism , Gene Expression Regulation , Muscles/metabolism , STAT3 Transcription Factor/metabolism , Transforming Growth Factor beta1/metabolism , Adolescent , Adult , Aged , Animals , Colitis/chemically induced , Colitis/genetics , Colitis/metabolism , Colitis/pathology , Collagen Type I/metabolism , Collagen Type I, alpha 1 Chain , Connective Tissue Growth Factor/genetics , Connective Tissue Growth Factor/metabolism , Crohn Disease/pathology , Cytokines/metabolism , Disease Models, Animal , Female , Fibrosis , Gene Expression , Genes, Reporter , Humans , Intestinal Mucosa/metabolism , Intestines/pathology , Male , Mice , Middle Aged , Muscle, Smooth/metabolism , Mutation , Phosphorylation , Promoter Regions, Genetic , Protein Binding , STAT3 Transcription Factor/genetics , Transcription, Genetic , Transforming Growth Factor beta1/genetics , Young Adult
20.
Am J Physiol Cell Physiol ; 308(6): C485-95, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25567809

ABSTRACT

Inhibitory neurotransmitters, chiefly nitric oxide and vasoactive intestinal peptide, increase cyclic nucleotide levels and inhibit muscle contraction via inhibition of myosin light chain (MLC) kinase and activation of MLC phosphatase (MLCP). H2S produced as an endogenous signaling molecule synthesized mainly from l-cysteine via cystathionine-γ-lyase (CSE) and cystathionine-ß-synthase (CBS) regulates muscle contraction. The aim of this study was to analyze the expression of CSE and H2S function in the regulation of MLCP activity, 20-kDa regulatory light chain of myosin II (MLC20) phosphorylation, and contraction in isolated gastric smooth muscle cells. Both mRNA expression and protein expression of CSE, but not CBS, were detected in smooth muscle cells of rabbit, human, and mouse stomach. l-cysteine, an activator of CSE, and NaHS, a donor of H2S, inhibited carbachol-induced Rho kinase and PKC activity, Rho kinase-sensitive phosphorylation of MYPT1, PKC-sensitive phosphorylation of CPI-17, and MLC20 phosphorylation and sustained muscle contraction. The inhibitory effects of l-cysteine, but not NaHS, were blocked upon suppression of CSE expression by siRNA or inhibition of its activity by dl-propargylglycine (PPG) suggesting that the effect of l-cysteine is mediated via activation of CSE. Glibenclamide, an inhibitor of KATP channels, had no effect on the inhibition of contraction by H2S. Both l-cysteine and NaHS had no effect on basal cAMP and cGMP levels but augmented forskolin-induced cAMP and SNP-induced cGMP formation. We conclude that both endogenous and exogenous H2S inhibit muscle contraction, and the mechanism involves inhibition of Rho kinase and PKC activities and stimulation of MLCP activity leading to MLC20 dephosphorylation and inhibition of muscle contraction.


Subject(s)
Hydrogen Sulfide/metabolism , Muscle Contraction , Myocytes, Smooth Muscle/metabolism , Signal Transduction , Stomach/enzymology , rhoA GTP-Binding Protein/metabolism , Animals , Cells, Cultured , Cyclic AMP/metabolism , Cyclic GMP/metabolism , Cystathionine gamma-Lyase/antagonists & inhibitors , Cystathionine gamma-Lyase/genetics , Cystathionine gamma-Lyase/metabolism , Dose-Response Relationship, Drug , Humans , Hydrogen Sulfide/pharmacology , Mice, Inbred C57BL , Muscle Contraction/drug effects , Muscle Proteins/metabolism , Myocytes, Smooth Muscle/drug effects , Myosin Light Chains/metabolism , Myosin-Light-Chain Phosphatase/metabolism , Nitric Oxide Donors/pharmacology , Phosphoproteins/metabolism , Phosphorylation , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , Protein Kinase Inhibitors/pharmacology , RNA Interference , Rabbits , Signal Transduction/drug effects , Stomach/cytology , Stomach/drug effects , Transfection , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...