Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 92
Filter
1.
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
2.
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
3.
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
4.
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
5.
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
6.
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
7.
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
8.
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
9.
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
10.
J Pharmacol Exp Ther ; 352(3): 509-18, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25550199

ABSTRACT

The effect of proinflammatory cytokines on the expression and activity of soluble guanylyl cyclase (sGC) and cGMP-phosphodiesterases (PDEs) was determined in intestinal longitudinal smooth muscle. In control muscle cells, cGMP levels are regulated via activation of sGC and PDE5; the activity of the latter is regulated via feedback phosphorylation by cGMP-dependent protein kinase. In muscle cells isolated from muscle strips cultured with interleukin-1ß (IL-1ß) or tumor necrosis factor α (TNF-α) or obtained from the colon of TNBS (2,4,6-trinitrobenzene sulfonic acid)-treated mice, expression of inducible nitric oxide synthase (iNOS) was induced and sGC was S-nitrosylated, resulting in attenuation of nitric oxide (NO)-induced sGC activity and cGMP formation. The effect of cytokines on sGC S-nitrosylation and activity was blocked by the iNOS inhibitor 1400W [N-([3-(aminomethyl)phenyl]methyl)ethanimidamide dihydrochloride]. The effect of cytokines on cGMP levels measured in the absence of IBMX (3-isobutyl-1-methylxanthine), however, was partly reversed by 1400W or PDE1 inhibitor vinpocetine and completely reversed by a combination of 1400W and vinpocetine. Expression of PDE1A was induced and was accompanied by an increase in PDE1A activity in muscle cells isolated from muscle strips cultured with IL-1ß or TNF-α or obtained from the colon of TNBS-treated mice; the effect of cytokines on PDE1 expression and activity was blocked by MG132 (benzyl N-[(2S)-4-methyl-1-[[(2S)-4-methyl-1-[[(2S)-4-methyl-1-oxopentan-2-yl]amino]-1-oxopentan-2-yl]amino]-1-oxopentan-2-yl]carbamate), an inhibitor of nuclear factor κB activity. NO-induced muscle relaxation was inhibited in longitudinal muscle cells isolated from muscle strips cultured with IL-1ß or TNF-α or obtained from the colon of TNBS-treated mice, and this inhibition was completely reversed by the combination of both 1400W and vinpocetine. Inhibition of smooth muscle relaxation during inflammation reflects the combined effects of decreased sGC activity via S-nitrosylation and increased cGMP hydrolysis via PDE1 expression.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 1/biosynthesis , Gene Expression Regulation, Enzymologic , Guanylate Cyclase/biosynthesis , Muscle Relaxation/physiology , Muscle, Smooth/enzymology , Nitric Oxide Synthase Type II/biosynthesis , Receptors, Cytoplasmic and Nuclear/biosynthesis , Animals , Cytokines/toxicity , Male , Mice , Mice, Inbred C57BL , Muscle Relaxation/drug effects , Muscle, Smooth/drug effects , Soluble Guanylyl Cyclase
11.
J Urol ; 193(5): 1676-83, 2015 May.
Article in English | MEDLINE | ID: mdl-25572034

ABSTRACT

PURPOSE: We examined the role of NMDAR in the regulation of bladder hypertrophy and function in a rat model of cyclophosphamide induced cystitis. MATERIALS AND METHODS: Cystitis was induced by intraperitoneal injection of cyclophosphamide (150 mg/kg body weight). NMDAR phosphorylation (activity) and signal transduction pathways were examined by direct measurement and by specific inhibitors in vivo. Bladder hypertrophy was measured by bladder weight/body weight and type I collagen expression. Bladder function was examined by metabolic recording, conscious cystometry and detrusor muscle strip contractility in response to carbachol. RESULTS: NMDAR activity measured by the phosphorylation level of the NMDAR1 (NR1) subunit was expressed in the spinal cord but not in the bladder at 48 hours of cystitis. NMDAR inhibition with dizocilpine (MK-801) reduced the cystitis induced increment of bladder weight and type I collagen up-regulation in the bladder. NMDAR regulated type I collagen up-regulation was mediated by the PI3K/Akt pathway. NMDAR inhibition also attenuated cystitis induced urinary frequency measured by metabolic cage and cystometry. Cystitis decreased the responsiveness of detrusor muscle strips to carbachol, which was reversed by MK-801 in vivo. Unlike MK-801 the NMDAR antagonist D-AP5, which could not block central NMDAR activity, had no effect on bladder hypertrophy, type I collagen up-regulation or Akt activation caused by cystitis in the bladder. CONCLUSIONS: Findings suggest that NMDAR activity has a role in cystitis induced bladder hypertrophy and overactivity. NMDAR mediated Akt activation may underlie the mechanism of bladder dysfunction.


Subject(s)
Cystitis/drug therapy , Cystitis/physiopathology , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Urinary Bladder/pathology , Urinary Bladder/physiopathology , Animals , Cyclophosphamide/administration & dosage , Cystitis/chemically induced , Hypertrophy/drug therapy , Male , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/physiology , Urinary Bladder/drug effects
12.
Am J Physiol Cell Physiol ; 307(4): C402-11, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-24944202

ABSTRACT

This study identified a distinctive pattern of expression and activity of adenylyl cyclase (AC) and phosphodiesterase (PDE) isoforms in mouse colonic longitudinal smooth muscle cells and determined the changes in their expression and/or activity in response to proinflammatory cytokines (IL-1ß and TNF-α) in vitro and 2,4,6 trinitrobenzene sulphonic acid (TNBS)-induced colonic inflammation in vivo. AC5/6 and PDE4D5, expressed in circular muscle cells, were also expressed in longitudinal smooth muscle. cAMP formation was tightly regulated via feedback phosphorylation of AC5/6 and PDE4D5 by PKA. Inhibition of PKA activity by myristoylated PKI blocked phosphorylation of AC5/6 and PDE4D5 and enhanced cAMP formation. TNBS treatment in vivo and IL-1ß and TNF-α in vitro induced inducible nitric oxide synthase (iNOS) expression, stimulated ERK1/2 activity, caused iNOS-mediated S-nitrosylation and inhibition of AC5/6, and induced phosphorylation of PDE4D5 and stimulated its activity. The resultant decrease in AC5/6 activity and increase in PDE4D5 activity decreased cAMP formation and smooth muscle relaxation. S-nitrosylation and inhibition of AC5/6 activity were reversed by the iNOS inhibitor 1400W, whereas phosphorylation and activation of PDE4D5 were reversed by the phosphatidylinositol 3-kinase inhibitor LY294002 and the ERK1/2 inhibitor PD98059. The effects of IL-1ß or TNF-α on forskolin-stimulated cAMP formation and smooth muscle relaxation reflected inhibition of AC5/6 activity and activation of PDE4D5 and were partly reversed by 1400W or PD98059 and completely reversed by a combination of the two inhibitors. The changes in the cAMP/PKA signaling and smooth muscle relaxation contribute to colonic dysmotility during inflammation.


Subject(s)
Adenylyl Cyclases/metabolism , Colitis/enzymology , Colon/enzymology , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Cytokines/metabolism , Gastrointestinal Motility , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Muscle, Smooth/enzymology , Nitric Oxide Synthase Type II/metabolism , Adenylyl Cyclase Inhibitors , Animals , Colitis/chemically induced , Colitis/immunology , Colitis/physiopathology , Colon/drug effects , Colon/immunology , Colon/physiopathology , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Disease Models, Animal , Down-Regulation , Enzyme Activation , Enzyme Inhibitors/pharmacology , Gastrointestinal Motility/drug effects , Inflammation Mediators/metabolism , Interleukin-1beta/metabolism , Male , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Muscle Relaxation , Muscle, Smooth/drug effects , Muscle, Smooth/immunology , Muscle, Smooth/physiopathology , Nitric Oxide Synthase Type II/antagonists & inhibitors , Phosphorylation , Signal Transduction , Trinitrobenzenesulfonic Acid , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation
13.
Am J Physiol Cell Physiol ; 306(12): C1129-41, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24740538

ABSTRACT

The signaling pathways mediating sustained contraction of mouse colonic longitudinal smooth muscle and the mechanisms involved in hypercontractility of this muscle layer in response to cytokines and TNBS-induced colitis have not been fully explored. In control longitudinal smooth muscle cells, ACh acting via m3 receptors activated sequentially Gα12, RhoGEF (LARG), and the RhoA/Rho kinase pathway. There was abundant expression of MYPT1, minimal expression of CPI-17, and a notable absence of a PKC/CPI-17 pathway. LARG expression was increased in longitudinal muscle cells isolated from muscle strips cultured for 24 h with IL-1ß or TNF-α or obtained from the colon of TNBS-treated mice. The increase in LARG expression was accompanied by a significant increase in ACh-stimulated Rho kinase and ZIP kinase activities, and sustained muscle contraction. The increase in LARG expression, Rho kinase and ZIP kinase activities, and sustained muscle contraction was abolished in cells pretreated with the Jun kinase inhibitor, SP600125. Expression of the MLCP activator, telokin, and MLCP activity were also decreased in longitudinal muscle cells from TNBS-treated mice or from strips treated with IL-1ß or TNF-α. In contrast, previous studies had shown that sustained contraction in circular smooth muscle is mediated by sequential activation of Gα13, p115RhoGEF, and dual RhoA-dependent pathways involving phosphorylation of MYPT1 and CPI-17. In colonic circular smooth muscle cells isolated from TNBS-treated mice or from strips treated with IL-1ß or TNF-α, CPI-17 expression and sustained muscle contraction were decreased. The disparate changes in the two muscle layers contribute to intestinal dysmotility during inflammation.


Subject(s)
Colitis/metabolism , Inflammation/genetics , JNK Mitogen-Activated Protein Kinases/metabolism , Muscle, Smooth/metabolism , Rho Guanine Nucleotide Exchange Factors/biosynthesis , Animals , Colitis/chemically induced , Colitis/pathology , Colon/metabolism , Death-Associated Protein Kinases/metabolism , Gene Expression Regulation/drug effects , Inflammation/metabolism , Inflammation/pathology , Mice , Muscle Contraction/genetics , Muscle, Smooth/pathology , Myosin-Light-Chain Kinase/biosynthesis , Organ Culture Techniques , Peptide Fragments/biosynthesis , Phosphorylation/drug effects , Rho Guanine Nucleotide Exchange Factors/metabolism , Signal Transduction/genetics , Trinitrobenzenesulfonic Acid/toxicity
14.
Am J Physiol Gastrointest Liver Physiol ; 306(6): G535-45, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24407588

ABSTRACT

We examined whether CB1 receptors in smooth muscle conform to the signaling pattern observed with other Gi-coupled receptors that stimulate contraction via two Gßγ-dependent pathways (PLC-ß3 and phosphatidylinositol 3-kinase/integrin-linked kinase). Here we show that the anticipated Gßγ-dependent signaling was abrogated. Except for inhibition of adenylyl cyclase via Gαi, signaling resulted from Gßγ-independent phosphorylation of CB1 receptors by GRK5, recruitment of ß-arrestin1/2, and activation of ERK1/2 and Src kinase. Neither uncoupling of CB1 receptors from Gi by pertussis toxin (PTx) or Gi minigene nor expression of a Gßγ-scavenging peptide had any effect on ERK1/2 activity. The latter was abolished in muscle cells expressing ß-arrestin1/2 siRNA. CB1 receptor internalization and both ERK1/2 and Src kinase activities were abolished in cells expressing kinase-deficient GRK5(K215R). Activation of ERK1/2 and Src kinase endowed CB1 receptors with the ability to inhibit concurrent contractile activity. We identified a consensus sequence (102KSPSKLSP109) for phosphorylation of RGS4 by ERK1/2 and showed that expression of a RGS4 mutant lacking Ser103/Ser108 blocked the ability of anandamide to inhibit acetylcholine-mediated phosphoinositide hydrolysis or enhance Gαq:RGS4 association and inactivation of Gαq. Activation of Src kinase by anandamide enhanced both myosin phosphatase RhoA-interacting protein (M-RIP):RhoA and M-RIP:MYPT1 association and inhibited Rho kinase activity, leading to increase of myosin light chain (MLC) phosphatase activity and inhibition of sustained muscle contraction. Thus, unlike other Gi-coupled receptors in smooth muscle, CB1 receptors did not engage Gßγ but signaled via GRK5/ß-arrestin activation of ERK1/2 and Src kinase: ERK1/2 accelerated inactivation of Gαq by RGS4, and Src kinase enhanced MLC phosphatase activity, leading to inhibition of ACh-stimulated contraction.


Subject(s)
Arrestins/metabolism , G-Protein-Coupled Receptor Kinase 5/metabolism , MAP Kinase Signaling System/physiology , Muscle, Smooth/physiology , Receptor, Cannabinoid, CB1/physiology , src-Family Kinases/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Arachidonic Acids/pharmacology , Endocannabinoids/pharmacology , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Muscle, Smooth/drug effects , Polyunsaturated Alkamides/pharmacology , Rabbits , Receptor, Cannabinoid, CB1/biosynthesis , Receptor, Cannabinoid, CB1/drug effects , beta-Arrestins , rho-Associated Kinases/metabolism
15.
Am J Physiol Gastrointest Liver Physiol ; 306(9): G802-10, 2014 May 01.
Article in English | MEDLINE | ID: mdl-24578342

ABSTRACT

Others and we have characterized several Gßγ-dependent effectors in smooth muscle, including G protein-coupled receptor kinase 2 (GRK2), PLCß3, and phosphatidylinositol (PI) 3-kinase-γ, and have identified various signaling targets downstream of PI 3-kinase-γ, including cSrc, integrin-linked kinase, and Rac1-Cdc42/p21-activated kinase/p38 MAP kinase. This study identified a novel mechanism whereby Gßγ acting via PI 3-kinase-γ and cSrc exerts an inhibitory influence on Gαi activity. The Gi2-coupled δ-opioid receptor agonist d-penicillamine (2,5)-enkephalin (DPDPE) activated cSrc, stimulated tyrosine phosphorylation of Gαi2, and induced regulator of G protein signaling 12 (RGS12) association; all three events were blocked by PI 3-kinase (LY294002) and cSrc (PP2) inhibitors and by expression of the COOH-terminal sequence of GRK2-(495-689), a Gßγ-scavenging peptide. Inhibition of forskolin-stimulated cAMP and muscle relaxation by DPDPE was augmented by PP2, LY294002, and a selective PI 3-kinase-γ inhibitor, AS-605420. Expression of tyrosine-deficient (Y69F, Y231F, or Y321F) Gαi2 mutant or knockdown of RGS12 blocked Gαi2 phosphorylation and Gαi2-RGS12 association and caused greater inhibition of cAMP. Parallel studies using somatostatin, cyclopentyl adenosine, or ACh to activate, respectively, Gi1-coupled somatostatin (sstr3) receptors, and Gi3-coupled adenosine A1 or muscarinic m2 receptors elicited cSrc activation, Gαi1 or Gαi3 phosphorylation, Gαi1-RGS12 or Gαi3-RGS12 association, and inhibition of cAMP. Inhibition of cAMP and muscle relaxation was greatly increased by AS-605240 and PP2. The results demonstrate that Gßγ-dependent tyrosine phosphorylation of Gαi1/2/3 by cSrc facilitated recruitment of RGS12, a Gαi-specific RGS protein with a unique phosphotyrosine-binding domain, resulting in rapid deactivation of Gαi and facilitation of smooth muscle relaxation.


Subject(s)
GTP-Binding Protein alpha Subunit, Gi2/metabolism , GTP-Binding Protein beta Subunits/metabolism , GTP-Binding Protein gamma Subunits/metabolism , Muscle Relaxation , Myocytes, Smooth Muscle/enzymology , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins pp60(c-src)/metabolism , RGS Proteins/metabolism , Stomach/enzymology , Adenosine A1 Receptor Agonists/pharmacology , Analgesics, Opioid/pharmacology , Animals , Cells, Cultured , Cyclic AMP/metabolism , Enzyme Activation , Feedback, Physiological , GTP-Binding Protein alpha Subunit, Gi2/genetics , Muscarinic Agonists/pharmacology , Muscle Relaxation/drug effects , Mutation , Myocytes, Smooth Muscle/drug effects , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins pp60(c-src)/antagonists & inhibitors , RGS Proteins/genetics , RNA Interference , Rabbits , Receptor, Adenosine A1/drug effects , Receptor, Adenosine A1/metabolism , Receptor, Muscarinic M2/agonists , Receptor, Muscarinic M2/metabolism , Receptors, Opioid, delta/agonists , Receptors, Opioid, delta/metabolism , Receptors, Somatostatin/agonists , Receptors, Somatostatin/metabolism , Signal Transduction , Stomach/cytology , Stomach/drug effects , Transfection , Tyrosine
16.
Am J Physiol Gastrointest Liver Physiol ; 306(10): G839-48, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24699329

ABSTRACT

Substance P (SP) is commonly coexpressed with ACh in enteric motor neurons, and, according to the classical paradigm, both these neurotransmitters excite smooth muscle via parallel pathways. We hypothesized that, in addition, SP was responsible for maintaining the muscular responsiveness to ACh. We tested this hypothesis by using botulinum toxin (BoNT/A), a known blocker of vesicular release of neurotransmitters including ACh and neuropeptides. BoNT/A was injected into rat pyloric sphincter in different doses; as control we used boiled BoNT/A. At the desired time point, pylorus was dissected out and pyloric contractility was measured ex vivo in an organ bath and by measuring phosphorylation of myosin light chain 20 (MLC20). BoNT/A (10 IU) significantly reduced the response of pyloric muscle to exogenous ACh, an effect that was accompanied by reduced MLC20 phosphorylation in the muscle. Both effects were reversed by exogenous SP. CP-96345, a NK1 receptor antagonist, blocked the ability of exogenous SP to reverse the cholinergic hyporesponsiveness as well as the reduction in MLC20 phosphorylation induced by BoNT/A. In conclusion, we have identified a novel role for SP as a coneurotransmitter that appears to be important for the maintenance of muscular responsiveness to the principal excitatory neurotransmitter, ACh. These results also provide new insight into the effects of botulinum toxin on the enteric nervous system and gastrointestinal smooth muscle.


Subject(s)
Botulinum Toxins/pharmacology , Muscle Contraction/drug effects , Muscle, Smooth/drug effects , Substance P/physiology , Acetylcholine/pharmacology , Animals , Biphenyl Compounds/pharmacology , Electric Stimulation , In Vitro Techniques , Male , Muscle, Smooth/physiology , Myosin Light Chains/metabolism , Pyloric Antrum/drug effects , Pyloric Antrum/physiology , Pylorus/drug effects , Rats , Receptors, Neurokinin-1/drug effects , Substance P/pharmacology
17.
Am J Physiol Gastrointest Liver Physiol ; 307(11): G1100-7, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25324508

ABSTRACT

Intraluminal nutrients in the gut affect the peristaltic reflex, although the mechanism is not well defined. Recent evidence supports the presence of taste receptors and their signaling components in enteroendocrine cells, although their function is unclear. This study aimed to determine if nutrients modify colonic motility through activation of taste receptors. Colonic sections were immunostained for the umami taste receptor T1R1/T1R3, which mediates the response to umami ligands, such as monosodium glutamate (MSG), in taste cells. Ascending contraction, descending relaxation, and calcitonin gene-related peptide release were measured in three-chamber flat-sheet preparations of rat colon in response to MSG alone or with inosine 5'-monophosphate (IMP). Velocity of artificial fecal pellet propulsion was measured by video recording in guinea pig distal colon. T1R1/T1R3 receptors were present in enteroendocrine cells of colonic sections from human, rat, mouse, and guinea pig. MSG initiated ascending contraction and descending relaxation components of the peristaltic reflex and calcitonin gene-related peptide release in flat-sheet preparations. IMP augmented the MSG-induced effects, suggesting activation of T1R1/T1R3 receptors. In T1R1(-/-) mice, mucosal stroking, but not MSG, elicited a peristaltic reflex. Intraluminal perfusion of MSG enhanced the velocity of artificial fecal pellet propulsion, which was also augmented by IMP. Propulsion was also increased by l-cysteine, but not l-tryptophan, supporting a role of T1R1/T1R3 receptors. We conclude that T1R1/T1R3 activation by luminal MSG or l-cysteine elicits a peristaltic reflex and CGRP release and increases the velocity of pellet propulsion in distal colon. This mechanism may explain how nutrients regulate colonic propulsion.


Subject(s)
Colon/drug effects , Gastrointestinal Motility/drug effects , Peristalsis/drug effects , Receptors, G-Protein-Coupled/drug effects , Taste/drug effects , Animals , Calcitonin Gene-Related Peptide/metabolism , Cysteine/pharmacology , Female , Food Additives , Guinea Pigs , Humans , Male , Mice , Mice, Inbred C57BL , Rats, Sprague-Dawley , Sodium Glutamate/pharmacology , Tryptophan/pharmacology
18.
J Pharmacol Exp Ther ; 350(1): 89-98, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24769544

ABSTRACT

Recent studies have identified AMP-activated kinase (AMPK) as a target of Ca(2+)/calmodulin-dependent kinase kinase (CaMKKß) and a negative regulator of myosin light-chain (MLC) kinase (MLCK). The present study examined whether a change in expression or activity of AMPK is responsible for hypercontractility of intestinal longitudinal muscle during inflammation or in response to proinflammatory cytokines. In mouse colonic longitudinal muscle cells, acetylcholine (ACh) stimulated AMPK and MLCK phosphorylation and activity and induced MLC20 phosphorylation and muscle contraction. Blockade of CaMKKß with STO609 (7-oxo-7H-benzimidazo[2,1-a]benz[de]isoquinoline-3-carboxylic acid acetate) inhibited AMPK and MLCK phosphorylation and augmented MLCK activity, MLC20 phosphorylation, and smooth muscle cell contraction. In muscle cells isolated from the colon of TNBS (2,4,6-trinitrobenzenesulfonic acid)-treated mice or from strips treated with interleukin-1ß or tumor necrosis factor-α, nuclear factor κB was activated as indicated by an increase in p65 phosphorylation and IκBα degradation, and AMPK was phosphorylated at a cAMP-dependent protein kinase (PKA)-specific site (Ser(485)) that is distinct from the stimulatory CaMKKß site (Thr(172)), resulting in attenuation of ACh-stimulated AMPK activity and augmentation of MLCK activity and muscle cell contraction. Inhibition of nuclear factor-κB activity with MG-132 (carbobenzoxy-L-leucyl-L-leucyl-L-leucinal Z-LLL-CHO) or PKA activity with myristoylated PKA inhibitor 14-22 amide blocked phosphorylation of AMPK at Ser(485) and restored MLCK activity and muscle cell contraction to control levels. The results imply that PKA released from IκBα complex phosphorylated AMPK at a PKA-specific site and inhibited its activity, thereby relieving the inhibitory effect of AMPK on MLCK and increasing MLCK activity and muscle cell contraction. We conclude that hypercontractility of intestinal longitudinal muscle induced by inflammation or proinflammatory cytokines is mediated by nuclear factor κB/PKA-dependent inhibition of AMPK and activation of MLCK.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Colon/metabolism , Cytokines/pharmacology , Muscle, Smooth/metabolism , Myosin-Light-Chain Kinase/metabolism , Acetylcholine/pharmacology , Animals , Benzimidazoles/pharmacology , Calcium-Calmodulin-Dependent Protein Kinase Kinase/antagonists & inhibitors , Carrier Proteins/pharmacology , Colitis/chemically induced , Colitis/metabolism , Colon/drug effects , Drug Interactions , Interleukin-1beta/pharmacology , Leupeptins/pharmacology , Male , Mice , Muscle Contraction/drug effects , Muscle, Smooth/drug effects , Myosin Light Chains/metabolism , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Naphthalimides/pharmacology , Peptide Fragments/pharmacology , Phosphorylation/drug effects , Signal Transduction/drug effects , Trinitrobenzenesulfonic Acid/pharmacology , Tumor Necrosis Factor-alpha/pharmacology
19.
Am J Physiol Cell Physiol ; 305(3): C334-47, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23784544

ABSTRACT

Caveolae act as scaffolding proteins for several G protein-coupled receptor signaling molecules to regulate their activity. Caveolin-1, the predominant isoform in smooth muscle, drives the formation of caveolae. The precise role of caveolin-1 and caveolae as scaffolds for G protein-coupled receptor signaling and contraction in gastrointestinal muscle is unclear. Thus the aim of this study was to examine the role of caveolin-1 in the regulation of Gq- and Gi-coupled receptor signaling. RT-PCR, Western blot, and radioligand-binding studies demonstrated the selective expression of M2 and M3 receptors in gastric smooth muscle cells. Carbachol (CCh) stimulated phosphatidylinositol (PI) hydrolysis, Rho kinase and zipper-interacting protein (ZIP) kinase activity, induced myosin phosphatase 1 (MYPT1) phosphorylation (at Thr(696)) and 20-kDa myosin light chain (MLC20) phosphorylation (at Ser(19)) and muscle contraction, and inhibited cAMP formation. Stimulation of PI hydrolysis, Rho kinase, and ZIP kinase activity, phosphorylation of MYPT1 and MLC20, and muscle contraction in response to CCh were attenuated by methyl ß-cyclodextrin (MßCD) or caveolin-1 small interfering RNA (siRNA). Similar inhibition of PI hydrolysis, Rho kinase, and ZIP kinase activity and muscle contraction in response to CCh and gastric emptying in vivo was obtained in caveolin-1-knockout mice compared with wild-type mice. Agonist-induced internalization of M2, but not M3, receptors was blocked by MßCD or caveolin-1 siRNA. Stimulation of PI hydrolysis, Rho kinase, and ZIP kinase activities in response to other Gq-coupled receptor agonists such as histamine and substance P was also attenuated by MßCD or caveolin-1 siRNA. Taken together, these results suggest that caveolin-1 facilitates signaling by Gq-coupled receptors and contributes to enhanced smooth muscle function.


Subject(s)
Caveolin 1/metabolism , Muscle Contraction/drug effects , Muscle, Smooth/metabolism , Receptor, Muscarinic M2/metabolism , Receptor, Muscarinic M3/metabolism , Animals , Carbachol/pharmacology , Caveolae/metabolism , Caveolin 1/genetics , Cells, Cultured , Cholinergic Agonists/pharmacology , Cyclic AMP/biosynthesis , Gastric Emptying , Gastrointestinal Tract/metabolism , MAP Kinase Kinase Kinases/metabolism , Mice , Mice, Knockout , Myosin-Light-Chain Phosphatase/metabolism , Phosphatidylinositols/metabolism , Phosphorylation , RNA Interference , RNA, Small Interfering , Rabbits , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , rho-Associated Kinases/metabolism
20.
Am J Physiol Gastrointest Liver Physiol ; 305(4): G314-24, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23764893

ABSTRACT

Previous studies have identified differences in the expression of proteins that regulate myosin light chain phosphorylation and contraction in tonic and phasic smooth muscle. cGMP plays a critical role in smooth muscle relaxation and is important for optimal function of phasic and tonic smooth muscle. The intracellular cGMP levels are regulated by its hydrolysis via phosphodiesterase 5 (PDE5) and efflux via novel multidrug resistance protein 5 (MRP5). In the present study we tested the hypothesis that the differences in the phasic and tonic behavior of smooth muscles may be related to differences in mechanisms that terminate cGMP signaling. Expression of PDE5 and MRP5 was significantly (more than 2-fold) higher in fundus compared with antrum. The NO donor S-nitrosoglutathione (GSNO) caused an increase in PDE5 activity and intra- and extracellular cGMP levels in both fundus and antrum. Stimulation of PDE5 activity and increase in extracellular cGMP were significantly higher in fundus, whereas increase in intracellular cGMP was significantly higher in antrum. GSNO-induced increase in extracellular cGMP was blocked in dispersed cells by the cyclic nucleotide export blocker probenecid and in cultured muscle cells by depletion of ATP or suppression of MRP5 by siRNA, providing evidence that cGMP efflux was mediated by ATP-dependent export via MRP5. Consistent with the higher expression and activity levels of PDE5 and MRP5, GSNO-induced PKG activity and muscle relaxation were significantly lower in muscle cells from fundus compared with antrum. Thus higher expression of PDE5 and MRP5 in muscle cells from fundus correlates with tonic phenotype of muscle.


Subject(s)
Cyclic GMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 5/metabolism , Gastric Fundus/enzymology , Multidrug Resistance-Associated Proteins/metabolism , Muscle Contraction , Muscle, Smooth/enzymology , Myocytes, Smooth Muscle/enzymology , Pyloric Antrum/enzymology , Adenosine Triphosphate/metabolism , Animals , Cells, Cultured , Cyclic GMP-Dependent Protein Kinases/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 5/genetics , Dose-Response Relationship, Drug , Gastric Fundus/cytology , Gastric Fundus/drug effects , Hydrolysis , Multidrug Resistance-Associated Proteins/genetics , Muscle Contraction/drug effects , Muscle Relaxation , Muscle, Smooth/cytology , Muscle, Smooth/drug effects , Myocytes, Smooth Muscle/drug effects , Nitric Oxide/metabolism , Nitric Oxide Donors/pharmacology , Phenotype , Pyloric Antrum/cytology , Pyloric Antrum/drug effects , RNA Interference , RNA, Messenger/metabolism , Rabbits , Second Messenger Systems , Time Factors , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL