Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 149
Filter
Add more filters










Publication year range
1.
Am J Physiol Gastrointest Liver Physiol ; 280(3): G381-8, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11171620

ABSTRACT

The identity of G proteins mediating CCK-stimulated phospholipase D (PLD) activity was determined in intestinal smooth muscle cells. CCK-8 activated G(q/11), G(13), and G(12), and the monomeric G proteins Ras-homology protein (RhoA) and ADP ribosylation factor (ARF). Activation of RhoA, but not ARF, was mediated by G(13) and inhibited by Galpha(13) antibody. CCK-stimulated PLD activity was partly mediated by RhoA and could be inhibited to the same extent (47 +/- 2% to 53 +/- 6%) by 1) a dominant negative RhoA mutant, 2) RhoA antibody or Galpha(13) antibody, and 3) Clostridium botulinum C3 exoenzyme. PLD activity was also inhibited by ARF antibody, and the effect was additive to that of RhoA antibody or C3 exoenzyme. PLD activity was inhibited by calphostin C, bisindolylmaleimide I, and a selective protein kinase C (PKC)-alpha inhibitor; the inhibition was additive to that of ARF and RhoA antibodies and C3 exoenzyme. In contrast, activated G(12) was not coupled to RhoA or ARF, and Galpha(12) antibody augmented PLD activity. Thus agonist-stimulated PLD activity is mediated additively by G(13)-dependent RhoA and by ARF and PKC-alpha and is modulated by an inhibitory G(12)-dependent pathway.


Subject(s)
Botulinum Toxins , Heterotrimeric GTP-Binding Proteins/metabolism , Monomeric GTP-Binding Proteins/metabolism , Muscle, Smooth/metabolism , Phospholipase D/metabolism , ADP Ribose Transferases/pharmacology , ADP-Ribosylation Factors/antagonists & inhibitors , ADP-Ribosylation Factors/metabolism , ADP-Ribosylation Factors/pharmacology , Animals , Antibodies/pharmacology , Cells, Cultured , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , GTP-Binding Protein alpha Subunits, G12-G13 , In Vitro Techniques , Intestines , Muscle, Smooth/cytology , Phospholipase D/antagonists & inhibitors , Protein Kinase C/metabolism , Rabbits , Signal Transduction/physiology , Sincalide/metabolism , Sincalide/pharmacology , rhoA GTP-Binding Protein/antagonists & inhibitors , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/metabolism , rhoA GTP-Binding Protein/pharmacology
2.
Am J Physiol Cell Physiol ; 279(4): C925-34, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11003572

ABSTRACT

This study examined the ability of protein kinase C (PKC) to induce heterologous desensitization by targeting specific G proteins and limiting their ability to transduce signals in smooth muscle. Activation of PKC by pretreatment of intestinal smooth muscle cells with phorbol 12-myristate 13-acetate, cholecystokinin octapeptide, or the phosphatase 1 and phosphatase 2A inhibitor, calyculin A, selectively phosphorylated Galpha(i-1) and Galpha(i-2), but not Galpha(i-3) or Galpha(o), and blocked inhibition of adenylyl cyclase mediated by somatostatin receptors coupled to G(i-1) and opioid receptors coupled to G(i-2), but not by muscarinic M(2) and adenosine A(1) receptors coupled to G(i-3). Phosphorylation of Galpha(i-1) and Galpha(i-2) and blockade of cyclase inhibition were reversed by calphostin C and bisindolylmaleimide, and additively by selective inhibitors of PKCalpha and PKCepsilon. Blockade of inhibition was prevented by downregulation of PKC. Phosphorylation of Galpha-subunits by PKC also affected responses mediated by betagamma-subunits. Pretreatment of muscle cells with cANP-(4-23), a selective agonist of the natriuretic peptide clearance receptor, NPR-C, which activates phospholipase C (PLC)-beta3 via the betagamma-subunits of G(i-1) and G(i-2), inhibited the PLC-beta response to somatostatin and [D-Pen(2,5)]enkephalin. The inhibition was partly reversed by calphostin C. Short-term activation of PKC had no effect on receptor binding or effector enzyme (adenylyl cyclase or PLC-beta) activity. We conclude that selective phosphorylation of Galpha(i-1) and Galpha(i-2) by PKC partly accounts for heterologous desensitization of responses mediated by the alpha- and betagamma-subunits of both G proteins. The desensitization reflects a decrease in reassociation and thus availability of heterotrimeric G proteins.


Subject(s)
GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Muscle, Smooth/metabolism , Protein Kinase C/metabolism , Adenylyl Cyclase Inhibitors , Adenylyl Cyclases/metabolism , Animals , Blotting, Western , Cells, Cultured , Colforsin/pharmacology , Cyclic AMP/metabolism , Dose-Response Relationship, Drug , Enkephalin, D-Penicillamine (2,5)-/pharmacology , Enzyme Inhibitors/pharmacology , GTP-Binding Protein alpha Subunits, Gi-Go/agonists , GTP-Binding Protein alpha Subunits, Gi-Go/antagonists & inhibitors , Hormones/pharmacology , Isoenzymes/antagonists & inhibitors , Isoenzymes/metabolism , Muscle, Smooth/cytology , Muscle, Smooth/drug effects , Naphthalenes/pharmacology , Phospholipase C beta , Phosphoric Monoester Hydrolases/antagonists & inhibitors , Phosphorylation/drug effects , Precipitin Tests , Protein Isoforms/metabolism , Protein Kinase C/antagonists & inhibitors , Rabbits , Signal Transduction/drug effects , Sincalide/pharmacology , Tetradecanoylphorbol Acetate/pharmacology , Type C Phospholipases/metabolism
3.
J Pharmacol Exp Ther ; 294(3): 991-6, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10945851

ABSTRACT

This study examined the source of Ca(2+) mobilized by phorbol esters and its requirement for phorbol-induced contraction of smooth muscle cells isolated from the circular and longitudinal layers of guinea pig intestine. Phorbol-12-myristate-13-acetate caused rapid, sustained, concentration-dependent muscle contraction and increase in cystolic free [Ca(2+)](i) in muscle cells from both layers. Maximal contraction was similar to that elicited by receptor-linked agonists, whereas maximal [Ca(2+)](i) was 50% less. The increase in [Ca(2+)](i) was mediated by Ca(2+) release in circular, and Ca(2+) influx in longitudinal muscle cells; only the latter was abolished by methoxyverapamil and in Ca(2+)-free medium. [Ca(2+)](i) was essential for contraction in both cell types: contraction in longitudinal muscle cells was abolished by methoxyverapamil and in Ca(2+)-free medium; contraction in circular muscle cells was abolished only after depletion of Ca(2+) stores. Contraction was abolished by the protein kinase C (PKC) inhibitor calphostin C (1 microM), but was not affected by the myosin light chain kinase inhibitor KT5926 (1 microM), suggesting that activation of myosin light chain kinase was suppressed by phorbol-12-myristate-13-acetate or via PKC. Phorbol-induced contraction of permeabilized circular and longitudinal muscle cells was abolished by pretreatment with a common antibody to Ca(2+)-dependent PKC-alpha,beta,gamma, but was not affected by pretreatment with a specific PKC-epsilon antibody. This study demonstrates the ability of phorbol esters to mobilize Ca(2+) from different sources in different smooth muscle cell types and establishes the requirement of Ca(2+) for phorbol-induced contraction; the latter is exclusively mediated by Ca(2+)-dependent PKC isozymes.


Subject(s)
Calcium/metabolism , Intestinal Mucosa/metabolism , Intestines/physiology , Muscle, Smooth/metabolism , Muscle, Smooth/physiology , Tetradecanoylphorbol Acetate/pharmacology , Animals , Guinea Pigs , In Vitro Techniques , Intestines/cytology , Isoenzymes/metabolism , Muscle Contraction , Muscle, Smooth/cytology , Protein Kinase C/metabolism
4.
Am J Physiol Gastrointest Liver Physiol ; 279(1): G201-10, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10898764

ABSTRACT

The role of protein kinase C (PKC) in sustained contraction was examined in intestinal circular and longitudinal muscle cells. Initial contraction induced by agonists (CCK-8 and neuromedin C) was abolished by 1) inhibitors of Ca(2+) mobilization (neomycin and dimethyleicosadienoic acid), 2) calmidazolium, and 3) myosin light chain (MLC) kinase (MLCK) inhibitor KT-5926. In contrast, sustained contraction was not affected by these inhibitors but was abolished by 1) the PKC inhibitors chelerythrine and calphostin C, 2) PKC-epsilon antibody, and 3) a pseudosubstrate PKC-epsilon inhibitor. GDPbetaS abolished both initial and sustained contraction, whereas a Galpha(q/11) antibody inhibited only initial contraction, implying that sustained contraction was dependent on activation of a distinct G protein. Sustained contraction induced by epidermal growth factor was inhibited by calphostin C, PKC-alpha,beta,gamma antibody, and a pseudosubstrate PKC-alpha inhibitor. Ca(2+) (0.4 microM) induced an initial contraction in permeabilized muscle cells that was blocked by calmodulin and MLCK inhibitors and a sustained contraction that was blocked by calphostin C and a PKC-alpha,beta,gamma antibody. Thus initial contraction induced by Ca(2+), agonists, and growth factors is mediated by MLCK, whereas sustained contraction is mediated by specific Ca(2+)-dependent and -independent PKC isozymes. G protein-coupled receptors are linked to PKC activation via distinct G proteins.


Subject(s)
Calcium/metabolism , Carbazoles , Epidermal Growth Factor/pharmacology , Indoles , Isoenzymes/metabolism , Muscle Contraction/drug effects , Muscle, Smooth/enzymology , Protein Kinase C/metabolism , Sincalide/pharmacology , Alkaloids/pharmacology , Animals , Antibodies/pharmacology , Benzophenanthridines , Bombesin/pharmacology , Enzyme Inhibitors/pharmacology , Fatty Acids, Unsaturated/pharmacology , Guinea Pigs , Imidazoles/pharmacology , In Vitro Techniques , Intestines/cytology , Isoenzymes/antagonists & inhibitors , Isoenzymes/immunology , Muscle, Smooth/cytology , Myosin-Light-Chain Kinase/antagonists & inhibitors , Naphthalenes/pharmacology , Neomycin/pharmacology , Peptide Fragments/pharmacology , Phenanthridines/pharmacology , Phosphodiesterase Inhibitors/pharmacology , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/immunology , Protein Kinase C beta , Protein Kinase C-alpha , Protein Kinase C-epsilon , Protein Synthesis Inhibitors/pharmacology
5.
Am J Physiol Gastrointest Liver Physiol ; 278(6): G974-80, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10859228

ABSTRACT

Single-transmembrane natriuretic peptide clearance receptor (NPR-C), which is devoid of a cytoplasmic guanylyl cyclase domain, interacts with pertussis toxin (PTx)-sensitive G proteins to activate endothelial nitric oxide synthase (eNOS) expressed in gastrointestinal smooth muscle cells. We examined the ability of NPR-C to activate other effector enzymes in eNOS-deficient tenia coli smooth muscle cells; these cells expressed NPR-C and NPR-B but not NPR-A. Atrial natriuretic peptide (ANP), the selective NPR-C ligand cANP-(4-23), and vasoactive intestinal peptide (VIP) inhibited (125)I-ANP and (125)I-VIP binding to muscle membranes in a pattern indicating high-affinity binding to NPR-C. Interaction of VIP with NPR-C was confirmed by its ability to inhibit (125)I-ANP binding to membranes of NPR-C-transfected COS-1 cells. In tenia muscle cells, all ligands selectively activated G(i-1) and G(i-2); VIP also activated G(s) via VIP(2) receptors. All ligands stimulated phosphoinositide hydrolysis, which was inhibited by ANP-(1-11), PTx, and antibodies to phospholipase C-beta3 (PLC-beta3) and Gbeta. cANP-(4-23) contracted tenia muscle cells; contraction was blocked by U-73122 and PTx and by antibodies to PLC-beta3 and Gbeta in intact and permeabilized muscle cells, respectively. VIP and ANP contracted muscle cells only after inhibition of cAMP- and cGMP-dependent protein kinases. ANP and cANP-(4-23) inhibited forskolin-stimulated cAMP in a PTx-sensitive fashion. We conclude that NPR-C is coupled to activation of PLC-beta3 via betagamma-subunits of G(i-1) and G(i-2) and to inhibition of adenylyl cyclase via alpha-subunits.


Subject(s)
GTP-Binding Protein alpha Subunits, Gi-Go/physiology , Guanylate Cyclase , Receptors, Atrial Natriuretic Factor/physiology , Signal Transduction/physiology , Adenylyl Cyclase Inhibitors , Animals , Atrial Natriuretic Factor/metabolism , Cell Membrane/metabolism , Cells, Cultured , Colon/cytology , Colon/metabolism , Enzyme Activation , GTP-Binding Proteins/physiology , Guinea Pigs , Isoenzymes/metabolism , Muscle, Smooth/cytology , Muscle, Smooth/metabolism , Natriuretic Agents/pharmacology , Phospholipase C beta , Receptors, Atrial Natriuretic Factor/agonists , Receptors, Atrial Natriuretic Factor/metabolism , Type C Phospholipases/metabolism , Vasoactive Intestinal Peptide/metabolism
6.
J Biol Chem ; 275(39): 30211-9, 2000 Sep 29.
Article in English | MEDLINE | ID: mdl-10862762

ABSTRACT

We examined the notion that sequestration of G protein subunits by binding to caveolin impedes G protein reassociation and leads to transient, G protein-specific desensitization of response in dispersed smooth muscle cells. Cholecystokinin octapeptide (CCK-8) and substance P (SP) were used to activate G(q/11), cyclopentyl adenosine (CPA) was used to activate G(i3), and acetylcholine (ACh) was used to activate both G(q/11) and G(i3) via m3 and m2 receptors, respectively. CCK-8 and SP increased only Galpha(q/11), and CPA increased only Galpha(i3) in caveolin immunoprecipitates; caveolin and other G proteins were not increased. ACh increased both Galpha(q/11) and Galpha(i3) in a time- and concentration-dependent fashion: only Galpha(q/11) was increased in the presence of an m2 antagonist, and only Galpha(i3) was increased in the presence of an m3 antagonist. To determine whether transient G protein binding to caveolin affected subsequent responses mediated by the same G protein, PLC-beta activity was measured in cells stimulated sequentially with two different agonists that activate either the same or a different G protein. After treatment of the cells with ACh and an m2 antagonist, the phospholipase C-beta (PLC-beta) response to CCK-8 and SP, but not CPA, was decreased; conversely, after treatment of the cells with ACh and an m3 antagonist, the PLC-beta response to CPA, but not CCK-8 or SP, was decreased. Similarly, after treatment with CCK-8 or SP, the PLC-beta response mediated by G(q/11) only was decreased, whereas after treatment with CPA, the PLC-beta response mediated by G(i3) only was decreased. A caveolin-binding Galpha(q/11) fragment blocked the binding of activated Galpha(q/11) but not Galpha(i3) to caveolin-3 and prevented desensitization of the PLC-beta response mediated only by other G(q/11)-coupled receptors. A caveolin-binding Galpha(i3) fragment had the reverse effect. Thus, transient binding of receptor-activated G protein subunits to caveolin impedes reassociation of the heterotrimeric species and leads to desensitization of response mediated by other receptors coupled to the same G protein.


Subject(s)
Caveolins/metabolism , Heterotrimeric GTP-Binding Proteins/metabolism , Muscle, Smooth/physiology , Acetylcholine/pharmacology , Adenosine/analogs & derivatives , Animals , Caveolin 1 , Caveolins/isolation & purification , Cell Compartmentation , Intestines/cytology , Muscle, Smooth/cytology , Protein Binding , Rabbits , Receptors, Cell Surface/metabolism , Receptors, Muscarinic/metabolism , Signal Transduction , Sincalide/pharmacology , Subcellular Fractions/chemistry , Substance P/pharmacology
7.
J Biol Chem ; 274(25): 17587-92, 1999 Jun 18.
Article in English | MEDLINE | ID: mdl-10364194

ABSTRACT

We have shown recently that the 37-amino acid intracellular domain of the single-transmembrane, natriuretic peptide clearance receptor, NPR-C, which is devoid of kinase and guanylyl cyclase activities, activates selectively Gi1 and Gi2 in gastric and tenia coli smooth muscle. In this study, we have used synthetic peptide fragments of the N-terminal, C-terminal, and middle regions of the cytoplasmic domain of NPR-C to identify the G protein-activating sequence. A 17-amino acid peptide of the middle region (Arg469-Arg485), denoted Peptide 4, which possesses two N-terminal arginine residues and a C-terminal B-B-X-X-B motif (where B and X are basic and non-basic residues, respectively) bound selectively to Gi1 and Gi2, activated phospholipase C-beta3 via the betagamma subunits, inhibited adenylyl cyclase, and induced smooth muscle contraction, in similar fashion to the selective NPR-C ligand, cANP4-23. A similar sequence (Peptide 3), but with a partial C-terminal motif, had minimal activity. Sequences which possessed either the N-terminal basic residues (Peptide 1) or the C-terminal B-B-X-X-B motif (Peptide 2) were inactive. Peptide 2, however, inhibited G protein activation and cellular responses mediated by the stimulatory Peptide 4 and by cANP4-23, suggesting that the B-B-X-X-B motif mediated binding but not activation of G protein, thus causing Peptide 2 to act as a competitive inhibitor of G protein activation.


Subject(s)
GTP-Binding Proteins/metabolism , Guanylate Cyclase/chemistry , Receptors, Atrial Natriuretic Factor/chemistry , Adenylyl Cyclases/metabolism , Amino Acid Sequence , Animals , Cells, Cultured , Colforsin/pharmacology , Cyclic AMP/metabolism , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Guanosine Triphosphate/metabolism , Guanylate Cyclase/metabolism , Guinea Pigs , Isoenzymes/metabolism , Molecular Sequence Data , Muscle Contraction/drug effects , Muscle, Smooth/drug effects , Muscle, Smooth/metabolism , Peptide Fragments/pharmacology , Phospholipase C beta , Protein Binding , Receptors, Atrial Natriuretic Factor/metabolism , Type C Phospholipases/metabolism
8.
Am J Physiol ; 275(6): C1409-16, 1998 12.
Article in English | MEDLINE | ID: mdl-9843699

ABSTRACT

In gastrointestinal smooth muscle, the neuropeptides vasoactive intestinal peptide (VIP) and pituitary adenylate cyclase-activating polypeptide (PACAP) induce relaxation by interacting with VIP2/PACAP3 receptors coupled via Gs to adenylyl cyclase and with distinct receptors coupled via Gi1 and/or Gi2 to a smooth muscle endothelial nitric oxide synthase (eNOS). The present study identifies the receptor as the single-transmembrane natriuretic peptide clearance receptor (NPR-C). RT-PCR and Northern analysis demonstrated expression of the natriuretic peptide receptors NPR-C and NPR-B but not NPR-A in rabbit gastric muscle cells. In binding studies using 125I-labeled atrial natriuretic peptide (125I-ANP) and 125I-VIP as radioligands, VIP, ANP, and the selective NPR-C ligand cANP(4-23) bound with high affinity to NPR-C. ANP, cANP-(4-23), and VIP initiated identical signaling cascades consisting of Ca2+ influx, activation of eNOS via Gi1 and Gi2, stimulation of cGMP formation, and muscle relaxation. NOS activity and cGMP formation were abolished (93 +/- 3 to 96 +/- 2% inhibition) by nifedipine, pertussis toxin, the NOS inhibitor, NG-nitro-L-arginine, and the antagonists ANP-(1-11) and VIP-(10-28). NOS activity stimulated by all three ligands in muscle membranes was additively inhibited by Gi1 and Gi2 antibodies (82 +/- 2 to 84 +/- 1%). In reconstitution studies, VIP, cANP-(4-23), and guanosine 5'-O-(3-thiotriphosphate) stimulated NOS activity in membranes of COS-1 cells cotransfected with NPR-C and eNOS. The results establish a unique mechanism for G protein-dependent activation of a constitutive NOS expressed in gastrointestinal smooth muscle involving interaction of the relaxant neuropeptides VIP and PACAP with a single-transmembrane natriuretic peptide receptor, NPR-C.


Subject(s)
GTP-Binding Proteins/physiology , Guanylate Cyclase/physiology , Muscle, Smooth/enzymology , Receptors, Atrial Natriuretic Factor/physiology , Animals , Atrial Natriuretic Factor/metabolism , Atrial Natriuretic Factor/pharmacology , COS Cells , Cattle , Gastric Mucosa/metabolism , Muscle, Smooth/cytology , Peptide Fragments/pharmacology , Rabbits , Signal Transduction/physiology , Stomach/cytology , Vasoactive Intestinal Peptide/metabolism , Vasoactive Intestinal Peptide/physiology
9.
J Biol Chem ; 273(51): 34519-26, 1998 Dec 18.
Article in English | MEDLINE | ID: mdl-9852121

ABSTRACT

Both cAMP- and cGMP-dependent protein kinases inhibit agonist-stimulated phospholipase C-beta (PLC-beta) activity and inositol 1,4,5-trisphosphate-dependent Ca2+ release in vascular and visceral smooth muscle. In smooth muscle of the intestinal longitudinal layer, however, the initial steps in Ca2+ mobilization involve activation of cytosolic PLA2 (cPLA2) and arachidonic acid (AA)-dependent stimulation of Ca2+ influx. The present study examined whether cAMP- and cGMP-dependent protein kinases are capable of regulating these processes also. Agents that activated cAMP-dependent protein kinase (5, 6-dichloro-1-beta-D-ribofuranosylbenzimidazole 3',5'-cyclic monophosphothioate (Sp-isomer) and isoproterenol), cGMP-dependent protein kinase (8-(4-chlorophenylthio)-guanosine 3',5'-cyclic monophosphate and Na nitroprusside), or both kinases (vasoactive intestinal peptide and isoproterenol >1 microM) induced phosphorylation of cPLA2 and inhibition of agonist-stimulated cPLA2 activity. Phosphorylation and inhibition of cPLA2 activity by cAMP- and cGMP-dependent protein kinases were blocked by the corresponding selective inhibitors (cAMP-dependent protein kinase, N-[2(p-bromocinnamylamino)ethyl]-5-isoquinoline-sulfonamide hydrochloride (H-89) and myristoylated protein kinase inhibitor () amide; cGMP-dependent protein kinase, (8R,9S, 11S)-(-)-9-methoxy-carbamyl-8-methyl-2,3,9,10-tetrahydro-8, 11-epoxy-1H,8H,11H,-2,7b,11a-trizadizobenzo(a,g)cycloocta(c, d, e)-trinden-1-one (KT-5823)). In contrast, AA-stimulated Ca2+ influx was inhibited by agents that activated cGMP-dependent protein kinase only; the inhibition was selectively blocked by KT-5823. The study provides the first evidence of inhibitory phosphorylation of cPLA2 in vivo by cAMP- and cGMP-dependent protein kinases. Inhibition of cPLA2 activity and AA-induced Ca2+ influx partly account for the ability of cAMP-dependent protein kinase and/or cGMP-dependent protein kinase to cause relaxation. Their importance resides in their location at the inception of the Ca2+ signaling cascade.


Subject(s)
Calcium/metabolism , Carbazoles , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic GMP-Dependent Protein Kinases/metabolism , Indoles , Muscle, Smooth/physiology , Phospholipases A/metabolism , Signal Transduction/physiology , Sulfonamides , Alkaloids/pharmacology , Animals , Arachidonic Acid/pharmacology , Cyclic GMP/analogs & derivatives , Cyclic GMP/pharmacology , Enzyme Activation , Enzyme Inhibitors/pharmacology , In Vitro Techniques , Intestines , Isoproterenol/pharmacology , Isoquinolines/pharmacology , Muscle Contraction/drug effects , Muscle, Smooth/drug effects , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/physiology , Nitroprusside/pharmacology , Phospholipases A2 , Phosphorylation , Rabbits , Thionucleotides/pharmacology , Vasoactive Intestinal Peptide/pharmacology
10.
Regul Pept ; 77(1-3): 127-34, 1998 Oct 16.
Article in English | MEDLINE | ID: mdl-9809806

ABSTRACT

In both functional and radioligand binding studies of gastric smooth muscle from rabbit and guinea pig, vasoactive intestinal peptide (VIP) and pituitary adenylate cyclase-activating polypeptide (PACAP) show equal potency indicating that the receptor type is either a VIP1/PACAP2 or a VIP2/PACAP3 receptor. We have characterized the VIP/PACAP receptor expressed in freshly dispersed and cultured gastric and tenia coli smooth muscle cells of rabbit and guinea pig by reverse transcriptase-polymerase chain reaction (RT-PCR), Northern analysis, and cloning of the first extracellular domain. Specific primers based on cDNA sequences for rat VIP1/PACAP2, VIP2/PACAP3 and PACAP1 receptors were designed spanning the first extracellular domain. A 275 base pair product corresponding to VIP2/PACAP3 receptor was amplified by RT-PCR in muscle cells from both species. No RT-PCR product was obtained with primers for VIP1/PACAP2 and PACAP1 receptors. The deduced amino acid sequences showed 90% similarity in rabbit and 77% in guinea pig to the sequence in rat. The location of the aspartate, tryptophan and glycine residues and all six N-terminal cysteines required for VIP binding were conserved. The sequence in guinea pig tenia coli differed from that in guinea pig stomach by two amino acid residues, Phe40 and Phe41. Northern analysis revealed a single 3.9 kilobase (kb) mRNA corresponding to VIP2/PACAP3 receptors in rabbit and a 2.1 kb mRNA in guinea pig gastric and tenia coli muscle cells. We conclude that only VIP2/PACAP3 receptors are expressed in smooth muscle cells of rabbit and guinea pig. The two amino acid difference in the sequence obtained from guinea pig tenia coli may reflect the distinct binding and functional properties of this tissue.


Subject(s)
Colon/metabolism , Gastric Mucosa/metabolism , Gene Expression Regulation/genetics , Muscle, Smooth, Vascular/metabolism , Receptors, Pituitary Hormone/genetics , Receptors, Vasoactive Intestinal Peptide/genetics , Amino Acid Sequence , Animals , Blotting, Southern , Cells, Cultured , Cloning, Molecular , Guinea Pigs , Molecular Sequence Data , RNA, Messenger/genetics , Rabbits , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide , Sequence Analysis, DNA , Sequence Homology, Amino Acid
11.
Am J Physiol ; 275(2): G342-51, 1998 08.
Article in English | MEDLINE | ID: mdl-9688662

ABSTRACT

The aim of this study was to identify the nitric oxide synthase (NOS) isoform expressed in freshly dispersed rabbit gastric smooth muscle cells and in cultured rabbit gastric, human intestinal, and guinea pig taenia coli smooth muscle cells. RT-PCR products of the predicted size (354 bp) were obtained with endothelial NOS (eNOS)-specific primers, but not neuronal NOS (nNOS)- or inducible NOS (iNOS)-specific primers, in all smooth muscle preparations except guinea pig taenia coli. Control RT-PCR studies showed absence of the endothelial markers, platelet endothelial cell adhesion molecule-1 (PECAM-1) and vascular endothelial growth factor receptor (VEGFR), and the interstitial cell marker, c-kit, from cultures of smooth muscle cells. Cloning and sequence analysis showed that the predicted amino acid sequence (117 residues) in rabbit and human smooth muscle cells differed by only one residue from that of human eNOS. Northern blot analysis, using the PCR-generated and cloned eNOS cDNA from rabbits and humans as probes, demonstrated the expression of eNOS mRNA (4.4 kb) in both species. eNOS, but not nNOS or iNOS, transcripts were localized by in situ RT-PCR in single, freshly dispersed rabbit gastric smooth muscle cells; expression was evident in the majority of cells in each preparation. We conclude that eNOS is selectively expressed in rabbit gastric and human intestinal smooth muscle cells. The results confirm functional evidence for the existence of a constitutive NOS in smooth muscle cells of the gut in different species, except for guinea pig taenia coli.


Subject(s)
Intestine, Small/enzymology , Muscle, Smooth/enzymology , Nitric Oxide Synthase/genetics , Stomach/enzymology , Amino Acid Sequence , Animals , Base Sequence , Cells, Cultured , Cloning, Molecular , Colon/cytology , Colon/enzymology , DNA Primers , Endothelium, Vascular/enzymology , Humans , Intestine, Small/cytology , Jejunum/cytology , Jejunum/enzymology , Molecular Sequence Data , Muscle, Smooth/cytology , Nitric Oxide Synthase/biosynthesis , Nitric Oxide Synthase/chemistry , Nitric Oxide Synthase Type III , Platelet Endothelial Cell Adhesion Molecule-1/biosynthesis , Platelet Endothelial Cell Adhesion Molecule-1/genetics , Polymerase Chain Reaction , Rabbits , Receptor Protein-Tyrosine Kinases/biosynthesis , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Growth Factor/biosynthesis , Receptors, Growth Factor/genetics , Receptors, Vascular Endothelial Growth Factor , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Sequence Alignment , Sequence Homology, Amino Acid , Stomach/cytology , Transcription, Genetic
12.
Mol Pharmacol ; 54(1): 122-8, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9658197

ABSTRACT

The characteristics of inhibitory regulation of adenylyl cyclase V/VI by Ca2+ and G proteins were examined in dispersed gastric smooth muscle cells. The mechanisms were evoked separately, sequentially, or concurrently using ligand-gated and G protein-coupled receptor agonists and receptor-independent probes (e. g, thapsigargin). During the initial phase of agonist stimulation, alpha,beta-methylene-ATP, UTP, and ATP inhibited forskolin-stimulated cAMP formation in a concentration-dependent fashion. Inhibition by alpha,beta-methylene-ATP, which activates ligand-gated P2X receptors, was abolished by zero Ca2+, whereas inhibition by UTP, which activates P2Y2 receptors coupled to Gq/11 and Gi3, was not affected by zero Ca2+ but was abolished by pertussis toxin (PTX). Inhibition by ATP, which activates both P2X and P2Y2 receptors, was not affected by zero Ca2+ alone; but after inhibition mediated by Galphai3 was blocked with PTX, inhibition by Ca2+ influx was unmasked and was abolished by zero Ca2+. Inhibition by cholecystokinin-8 was observed only during the phase of capacitative Ca2+ influx and was blocked by zero Ca2+. Inhibition by UTP during this phase was not affected by zero Ca2+ alone; but after inhibition mediated by Galphai3 was blocked with PTX, inhibition by Ca2+ influx was unmasked and was abolished by zero Ca2+. Inhibition of adenylyl cyclase V/VI activity in smooth muscle can be mediated independently by inhibitory G proteins and Ca2+ influx but is exclusively mediated by inhibitory G proteins when both mechanisms are triggered.


Subject(s)
Adenylyl Cyclases/metabolism , Calcium/metabolism , GTP-Binding Proteins/antagonists & inhibitors , Muscle, Smooth/enzymology , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/pharmacology , Animals , Colforsin/pharmacology , Cyclic AMP/metabolism , Dose-Response Relationship, Drug , GTP-Binding Proteins/drug effects , GTP-Binding Proteins/metabolism , Rabbits , Stomach/enzymology
13.
Am J Physiol ; 274(5): C1199-205, 1998 05.
Article in English | MEDLINE | ID: mdl-9612206

ABSTRACT

Recent studies on the role of nitric oxide (NO) in gastrointestinal smooth muscle have raised the possibility that NO-stimulated cGMP could, in the absence of cGMP-dependent protein kinase (PKG) activity, act as a Ca(2+)-mobilizing messenger [K. S. Murthy, K.-M. Zhang, J.-G. f1p4 J. T. Grider, and G. M. Makhlouf. Am. J. Physiol. 265 (Gastrointest. Liver Physiol. 28): G660-G671, 1993]. This notion was examined in dispersed gastric smooth muscle cells with 8-bromo-cGMP (8-BrcGMP) and with NO and vasoactive intestinal peptide (VIP), which stimulate endogenous cGMP. In muscle cells treated with cAMP-dependent protein kinase (PKA) and PKG inhibitors (H-89 and KT-5823), 8-BrcGMP (10 microM), NO (1 microM), and VIP (1 microM) stimulated 45Ca2+ release (21 +/- 3 to 30 +/- 1% decrease in 45Ca2+ cell content); Ca2+ release stimulated by 8-BrcGMP was concentration dependent with an EC50 of 0.4 +/- 0.1 microM and a threshold of 10 nM. 8-BrcGMP and NO increased cytosolic free Ca2+ concentration ([Ca2+]i) and induced contraction; both responses were abolished after Ca2+ stores were depleted with thapsigargin. With VIP, which normally increases [Ca2+]i by stimulating Ca2+ influx, treatment with PKA and PKG inhibitors caused a further increase in [Ca2+]i that reverted to control levels in cells pretreated with thapsigargin. Neither Ca2+ release nor contraction induced by cGMP and NO in permeabilized muscle cells was affected by heparin or ruthenium red. Ca2+ release induced by maximally effective concentrations of cGMP and inositol 1,4,5-trisphosphate (IP3) was additive, independent of which agent was applied first. We conclude that, in the absence of PKA and PKG activity, cGMP stimulates Ca2+ release from an IP3-insensitive store and that its effect is additive to that of IP3.


Subject(s)
Calcium/metabolism , Cyclic GMP/physiology , Inositol 1,4,5-Trisphosphate/pharmacology , Muscle, Smooth/drug effects , Muscle, Smooth/metabolism , Animals , Cyclic GMP/analogs & derivatives , Cyclic GMP/pharmacology , Cytosol/metabolism , Muscle Contraction/physiology , Muscle, Smooth/cytology , Nitric Oxide/pharmacology , Nitric Oxide Synthase/drug effects , Nitric Oxide Synthase/metabolism , Osmolar Concentration , Rabbits , Vasoactive Intestinal Peptide/pharmacology
14.
J Biol Chem ; 273(8): 4695-704, 1998 Feb 20.
Article in English | MEDLINE | ID: mdl-9468531

ABSTRACT

P2 receptor subtypes and their signaling mechanisms were characterized in dispersed smooth muscle cells. UTP and ATP stimulated inositol 1,4,5-triphosphate formation, Ca2+ release, and contraction that were abolished by U-73122 and guanosine 5'-O-(3-thio)diphosphate, and partly inhibited (50-60%) by pertussis toxin (PTX). ATP analogs (adenosine 5'-(alpha, beta-methylene)triphosphate, adenosine 5'-(beta, gamma-methylene)triphosphate, and 2-methylthio-ATP) stimulated Ca2+ influx and contraction that were abolished by nifedipine and in Ca2+-free medium. Micromolar concentrations of ATP stimulated both Ca2+ influx and Ca2+ release. ATP and UTP activated Gq/11 and Gi3 in gastric and aortic smooth muscle and heart membranes, Gq/11 and Gi1 and/or Gi2 in liver membranes, and Go and Gi1-3 in brain membranes. Phosphoinositide hydrolysis stimulated by ATP and UTP was mediated concurrently by Galphaq/11-dependent activation of phospholipase (PL) C-beta1 and Gbetagammai3-dependent activation of PLC-beta3. Phosphoinositide hydrolysis was partially inhibited by PTX or by antibodies to Galphaq/11, Gbeta, PLC-beta1, or PLC-beta3, and completely inhibited by the following combinations (PLC-beta1 and PLC-beta3 antibodies; Galphaq/11 and Gbeta antibodies; PLC-beta1 and Gbeta antibodies; PTX with either PLC-beta1 or Galphaq/11 antibody). The pattern of responses implied that P2Y2 receptors in visceral, and probably vascular, smooth muscle are coupled to PLC-beta1 via Galphaq/11 and to PLC-beta3 via Gbetagammai3. These receptors co-exist with ligand-gated P2X1 receptors activated by ATP analogs and high levels of ATP.


Subject(s)
GTP-Binding Proteins/metabolism , Isoenzymes/metabolism , Muscle, Smooth/metabolism , Receptors, Purinergic P2/metabolism , Type C Phospholipases/metabolism , Adenosine Monophosphate/analogs & derivatives , Adenosine Monophosphate/pharmacology , Adenosine Triphosphate/pharmacology , Amino Acid Sequence , Animals , Calcium/metabolism , GTP-Binding Proteins/chemistry , Muscle Contraction/drug effects , Muscle, Smooth/physiology , Phospholipase C beta , Purinergic P2 Receptor Agonists , Rabbits , Uridine Triphosphate/pharmacology
15.
Cell Biochem Biophys ; 28(1): 31-44, 1998.
Article in English | MEDLINE | ID: mdl-9386891

ABSTRACT

Ca2+ mobilization in muscle cells from the circular muscle layer of the mammalian intestine is mediated by IP3-dependent Ca2+ release. Ca2+ mobilization in muscle from the adjacent longitudinal muscle layer involves a distinct, phosphoinositide-independent pathway. Receptors for contractile agonists in longitudinal muscle cells are coupled via a pertussis toxin-sensitive G protein to activation of PLA2 and formation of arachidonic acid (AA). The latter activates Cl- channels resulting in depolarization of the plasma membrane and opening of voltage-sensitive Ca2+ channels. Ca2+ influx via these channels induces Ca2+ release by activating sarcoplasmic ryanodine receptor/Ca2+ channels. The increase in [Ca2+]i activates membrane-bound ADP ribosyl cyclase, and the resultant formation of cADPR enhances Ca(2+)-induced Ca2+ release.


Subject(s)
Adenosine Diphosphate Ribose/analogs & derivatives , Calcium/physiology , Intestines/physiology , Muscle, Smooth/physiology , Signal Transduction , Adenosine Diphosphate Ribose/physiology , Animals , Calcium/metabolism , Calcium Channels/physiology , Cyclic ADP-Ribose , Enzyme Activation , Humans , Phosphatidylinositols/metabolism , Phospholipases A/metabolism , Phospholipases A2
16.
J Biol Chem ; 272(34): 21317-24, 1997 Aug 22.
Article in English | MEDLINE | ID: mdl-9261144

ABSTRACT

Muscarinic m2 and m4 receptors couple preferentially to inhibition of adenylyl cyclase, whereas m1, m3, and m5 receptors couple preferentially to activation of phospholipase C-beta and in some cells to stimulation of cAMP. Smooth muscle cells were shown to express adenylyl cyclases types V and/or VI. Acetylcholine (ACh) stimulated the binding of [35S]GTPgammaS.Galpha complexes in smooth muscle membranes to Galphaq/11 and Galphai3 antibody. Binding to Galphaq/11 antibody was inhibited by the m3 receptor antagonist, 4-DAMP, and binding to Galphai3 antibody was inhibited by the m2 receptor antagonist, N,N'-bis[6[[(2-methoxyphenyl)methyl]amino]hexyl]-1,8-octanediamine tetrahydrochloride (methoctramine). The decrease in basal cAMP (35 +/- 5%) induced by ACh in dispersed muscle cells was accentuated by 4-DAMP or Gbeta antibody (55 +/- 8 to 63 +/- 6%). In contrast, methoctramine, pertussis toxin (PTx), or Galphai3 antibody converted the decrease in cAMP to increase above basal level (+28 +/- 5 to +32 +/- 6%); the increase in cAMP was abolished by 4-DAMP or Gbeta antibody. In muscle cells where only m3 receptors were preserved by selective receptor protection, ACh caused only an increase in cAMP that was abolished by 4-DAMP. Conversely, in muscle cells where only m2 receptors were preserved, ACh caused an accentuated decrease in cAMP that was abolished by methoctramine or PTx. In conclusion, m2 receptors in smooth muscle couple to inhibition of adenylyl cyclases V/VI via Galphai3, and m3 receptors couple to activation of the enzymes via Gbetagammaq/11.


Subject(s)
Adenylyl Cyclases/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , GTP-Binding Proteins/metabolism , Receptors, Muscarinic/metabolism , Acetylcholine/pharmacology , Adenylyl Cyclases/classification , Animals , Cells, Cultured , Cyclic AMP/biosynthesis , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Immunologic Techniques , Muscle, Smooth , Rabbits , Receptor, Muscarinic M2 , Receptor, Muscarinic M3 , Scopolamine/metabolism , Signal Transduction , Stomach
17.
Am J Physiol ; 272(6 Pt 1): G1391-9, 1997 Jun.
Article in English | MEDLINE | ID: mdl-9227474

ABSTRACT

Pituitary adenylate cyclase-activating peptide (PACAP) receptors and their signaling pathways were characterized in dispersed rabbit gastric muscle cells. 125I-PACAP-27 and 125I-vasoactive intestinal peptide (VIP) binding to muscle cells were inhibited equally by PACAP and VIP (mean inhibitory concentration 0.8 to 1.3 nM) and desensitized to the same extent (70-80%) by exposure to either peptide. PACAP, like VIP, increased cytosolic free Ca2+ and the formation of L-[3H]citrulline, NO-3/NO-2, guanosine 3',5'-cyclic monophosphate (cGMP), and adenosine 3'5'-cyclic monophosphate (cAMP) and induced relaxation (mean effective concentration 1.8 +/- 0.1 nM) that was partly inhibited by NG-nitro-L-arginine (L-NNA), VIP-(10-28), and PACAP 6-38. L-[3H]citrulline and cGMP formation were blocked by nifedipine, L-NNA, and pertussis toxin (PTx), implying activation of a G protein-coupled, Ca(2+)-calmodulin-dependent nitric oxide (NO) synthase. PACAP-induced relaxation was inhibited to the same extent (46-49%) by nifedipine, L-NNA, PTx, and the protein kinase G inhibitor KT-5823; the inhibition reflected the component of relaxation mediated by the NO-cGMP pathway. The residual relaxation was abolished by the protein kinase A inhibitor H-89. The pattern of inhibition of all responses was identical to that observed with VIP. Desensitization with VIP or PACAP abolished cAMP formation but had no effect on L-[3H]citrulline and cGMP formation induced by either peptide. Receptor protection with VIP or PACAP preserved fully all responses (L-[3H]citrulline, cGMP, and cAMP formation and relaxation) to either peptide. The complete cross-competition, cross-desensitization, cross-antagonism, and cross-protection of receptors by either VIP or PACAP are consistent with interaction of both peptides with the same receptors; the receptors consist of two classes, each coupled to a distinct signaling pathway.


Subject(s)
Carbazoles , Indoles , Muscle, Smooth/physiology , Neuropeptides/pharmacology , Receptors, Pituitary Hormone/physiology , Signal Transduction , Stomach/physiology , Sulfonamides , Adenylate Cyclase Toxin , Alkaloids/pharmacology , Aminoquinolines/pharmacology , Animals , Calcium/metabolism , Cells, Cultured , Citrulline/metabolism , Cyclic AMP/pharmacology , Cyclic GMP/metabolism , Cyclic GMP/pharmacology , Cytosol/metabolism , Enzyme Inhibitors/pharmacology , GTP-Binding Proteins/metabolism , Guanylate Cyclase/antagonists & inhibitors , Isoquinolines/pharmacology , Kinetics , Muscle Relaxation , Muscle, Smooth/drug effects , Neuropeptides/metabolism , Neurotransmitter Agents/metabolism , Nifedipine/pharmacology , Nitric Oxide Synthase/metabolism , Nitroarginine/pharmacology , Pertussis Toxin , Pituitary Adenylate Cyclase-Activating Polypeptide , Protein Kinase Inhibitors , Rabbits , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide , Stomach/drug effects , Vasoactive Intestinal Peptide/metabolism , Vasoactive Intestinal Peptide/pharmacology , Virulence Factors, Bordetella/pharmacology
19.
Cell Signal ; 9(3-4): 269-76, 1997.
Article in English | MEDLINE | ID: mdl-9218127

ABSTRACT

Signal transduction in gastric and intestinal smooth muscle is mediated by receptors coupled via distinct G proteins to various effector enzymes, including PI-specific PLC-beta 1 and PLC-beta 3, and phosphatidylcholine (PC)-specific PLC, PLD and PLA2. Activation of these enzymes is different in circular and longitudinal muscle cells, generating Ca(2+)-mobilizing (IP3, AA, cADPR) and other (DAG) messengers responsible for the initial and sustained phases of contraction, respectively. IP3-dependent Ca2+ release occurs only in circular muscle. Ca2+ mobilization in longitudinal muscle involves a cascade initiated by agonist-induced transient activation of PLA2 and formation of AA, AA-dependent depolarization of the plasma membrane and opening of voltage-sensitive Ca2+ channels. The influx of Ca2+ induces Ca2+ release by activating sarcoplasmic ryanodine receptor/Ca2+ channel and stimulates cADPR formation which enhances Ca(2+)-induced Ca2+ release. The initial [Ca2+]i transient in both muscle cell types results in Ca2+/calmodulin-dependent activation of MLC kinase, phosphorylation of MLC20 and interaction of actin and myosin. The sustained phase is mediated by a Ca(2+)-independent isoform of PKC, PKC-epsilon DAG for this process is generated by PLC- and PLD-mediated hydrolysis of PC. Relaxation is mediated by cAMP-and/or cGMP-dependent protein kinase which inhibit the initial [Ca2+]i transient and reduce the sensitivity of MLC kinase to [Ca2+]i. Relaxation induced by the main neurotransmitters, VIP and PACAP, involves two cascades, one of which reflects activation of adenylyl cyclase. A distinct cascade involves G-protein-dependent stimulation of Ca2+ influx leading to Ca2+/calmodulin-dependent activation of a constitutive eNOS in muscle cells; the generation of NO activates soluble guanylyl cyclase. The resultant activation of PKA and PKG is jointly responsible for muscle relaxation.


Subject(s)
GTP-Binding Proteins/metabolism , Gastric Mucosa/metabolism , Intestinal Mucosa/metabolism , Muscle, Smooth/metabolism , Signal Transduction , Animals , Humans , Muscle Contraction , Muscle Relaxation
SELECTION OF CITATIONS
SEARCH DETAIL
...