Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Neurogastroenterol Motil ; 26(4): 556-70, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24460867

RESUMO

BACKGROUND: 5-Hydroxytryptamine (5-HT, serotonin) is an important regulator of colonic motility and secretion; yet the role of serotonergic neurons in the colon is controversial. METHODS: We used immunohistochemical techniques to examine their projections throughout the enteric nervous system and interstitial cells of Cajal (ICC) networks in the murine proximal to mid colon. KEY RESULTS: Serotonergic neurons, which were mainly calbindin positive, occurred only in myenteric ganglia (1 per 3 ganglia). They were larger than nNOS neurons but similar in size to Dogiel Type II (AH) neurons. 5-HT neurons, appeared to make numerous varicose contacts with each other, most nNOS neurons, Dogiel Type II/AH neurons and glial cells. 5-HT, calbindin and nNOS nerve fibers also formed a thin perimuscular nerve plexus that was associated with ganglia, which contained both nNOS positive and negative neurons, which lay directly upon the submucosal pacemaker ICC network. Neurons in perimuscular ganglia were surrounded by 5-HT varicosities. Submucous ganglia contained nNOS positive and negative neurons, and calbindin positive neurons, which also appeared richly supplied by serotonergic nerve varicosities. Serotonergic nerve fibers ran along submucosal arterioles, but not veins. Varicosities of serotonergic nerve fibers were closely associated with pacemaker ICC networks and with intramuscular ICC (ICC-IM). 5-HT2B receptors were found on a subpopulation of non-5-HT containing myenteric neurons and their varicosities, pacemaker ICC-MY and ICC-IM. CONCLUSIONS & INFERENCES: Myenteric serotonergic neurons, whose axons exhibit considerable divergence, regulate the entire enteric nervous system and are important in coordinating motility with secretion. They are not just interneurons, as regularly assumed, but possibly also motor neurons to ICC and blood vessels, and some may even be sensory neurons.


Assuntos
Colo/inervação , Plexo Mientérico/citologia , Neurônios Serotoninérgicos/citologia , Animais , Colo/metabolismo , Células Intersticiais de Cajal/citologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Receptor 5-HT2B de Serotonina/metabolismo
2.
Neurogastroenterol Motil ; 23(10): 949-e409, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21806740

RESUMO

BACKGROUND: Inflammatory responses can include recruitment of cells of hematopoietic origin to the tunica muscularis. These cells can secrete a variety of factors which can reset the gain of smooth muscle cells (SMC) and influence motor patterns. Histamine (H), a major mediator in inflammation, is released by mast cells and exerts diverse effects in SMC by binding to H receptors. The profiles of H receptor expression in animal models used to study inflammatory diseases are unknown. METHODS: Histamine receptor expression and electro-mechanical responses to H were tested in simian and murine colonic smooth muscle using qualitative and quantitative PCR, isometric force measurements, microelectrode recordings and patch clamp techniques. KEY RESULTS: H1, H2, and H4 receptor transcripts were expressed at similar levels in simian colonic tissue whereas only the H2 receptor transcript was detected in murine colonic tissue. Stimulation of simian colonic muscles with H caused depolarization and contraction in the presence of tetrodotoxin. Histamine activated non-selective cation channels in simian SMC. In contrast, H caused hyperpolarization and inhibited contractions of murine colon. The hyperpolarization was inhibited by the K(ATP) channel blocker, glibenclamide. Histamine-activated K(+) currents were inhibited by glibenclamide in murine colonic SMC. CONCLUSIONS & INFERENCES: Histamine receptor expression in simian SMC was similar to that reported in humans. However, H receptor profile and responses to H were considerably different in mice. Thus, monkey colon may be a more suitable model to study how inflammatory mediators affect the gain of smooth muscle excitability.


Assuntos
Colo/metabolismo , Histamina/metabolismo , Inflamação/metabolismo , Músculo Liso/metabolismo , Receptores Histamínicos/biossíntese , Animais , Colo/efeitos dos fármacos , Feminino , Histamina/farmacologia , Macaca fascicularis , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Contração Muscular/efeitos dos fármacos , Músculo Liso/efeitos dos fármacos , Técnicas de Patch-Clamp , Reação em Cadeia da Polimerase em Tempo Real
3.
Br J Pharmacol ; 163(3): 638-48, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21265824

RESUMO

BACKGROUND AND PURPOSE: The Rho-kinase pathway regulates agonist-induced contractions in several smooth muscles, including the intestine, urinary bladder and uterus, via dynamic changes in the Ca(2+) sensitivity of the contractile apparatus. However, there is evidence that Rho-kinase also modulates other cellular effectors such as ion channels. EXPERIMENTAL APPROACH: We examined the regulation of colonic smooth muscle excitability by Rho-kinase using conventional microelectrode recording, isometric force measurements and patch-clamp techniques. KEY RESULTS: The Rho-kinase inhibitors, Y-27632 and H-1152, decreased nerve-evoked on- and off-contractions elicited at a range of frequencies and durations. The Rho-kinase inhibitors decreased the spontaneous contractions and the responses to carbachol and substance P independently of neuronal inputs, suggesting Y-27632 acts directly on smooth muscle. The Rho-kinase inhibitors significantly reduced the depolarization in response to carbachol, an effect that cannot be due to regulation of Ca(2+) sensitization. Patch-clamp experiments showed that Rho-kinase inhibitors reduce GTPγS-activated non-selective cation currents. CONCLUSIONS AND IMPLICATIONS: The Rho-kinase inhibitors decreased contractions evoked by nerve stimulation, carbachol and substance P. These effects were not solely due to inhibition of the Ca(2+) sensitization pathway, as the Rho-kinase inhibitors also inhibited the non-selective cation conductances activated by excitatory transmitters. Thus, Rho-kinase may regulate smooth muscle excitability mechanisms by regulating non-selective cation channels as well as changing the Ca(2+) sensitivity of the contractile apparatus.


Assuntos
Membrana Celular/fisiologia , Colo/fisiologia , Músculo Liso/fisiologia , Quinases Associadas a rho/fisiologia , Amidas/farmacologia , Animais , Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio Tipo L/fisiologia , Carbacol/farmacologia , Colo/inervação , Guanosina 5'-O-(3-Tiotrifosfato)/farmacologia , Técnicas In Vitro , Potenciais da Membrana , Camundongos , Contração Muscular , Músculo Liso/inervação , Técnicas de Patch-Clamp , Piridinas/farmacologia , Quinases Associadas a rho/antagonistas & inibidores
4.
Neurogastroenterol Motil ; 22(2): 186-95, e64, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19735476

RESUMO

BACKGROUND: Altered calcium mobilization has been implicated in the development of colonic dysmotility in inflammatory bowel disease. The aim of this study was to investigate the mechanisms by which disrupted intracellular Ca(2+) signalling contributes to the impaired contractility of colon circular smooth muscles. METHODS: Acute colitis was induced in C57Bl/6 mice with dextran sulphate sodium (DSS) in the drinking water for 5 days. KEY RESULTS: Spontaneous and acetylcholine-evoked contractions, caffeine-evoked hyperpolarization, and SERCA2 and phospholamban expression were reduced compared with controls. Tetrodotoxin did not restore control levels of contractile activity. The amplitudes, but not the frequency, of intracellular Ca(2+) waves were increased compared with controls. Caffeine abolished intracellular Ca(2+) waves in control smooth muscle cells, but not in smooth muscle cells from DSS-treated mice. CaM kinase II activity and cytosolic levels of HDAC4 were increased, and I kappaB alpha levels were decreased in distal colon smooth muscles from DSS-treated mice. CONCLUSIONS & INFERENCES: These results suggest that disruptions in intracellular Ca(2+) mobilization due to down-regulation of SERCA2 and phospholamban expression lead to increased CaM kinase II activity and cytosolic HDAC4 that may contribute to the dysmotility of colonic smooth muscles in colitis by enhancing NF-kappaB activity.


Assuntos
Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Colite/fisiopatologia , Colo/metabolismo , Motilidade Gastrointestinal/fisiologia , Contração Muscular/fisiologia , Músculo Liso/metabolismo , Acetilcolina/farmacologia , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Animais , Western Blotting , Cafeína/farmacologia , Sinalização do Cálcio/efeitos dos fármacos , Sinalização do Cálcio/fisiologia , Proteínas de Ligação ao Cálcio/metabolismo , Colite/induzido quimicamente , Colite/metabolismo , Colo/efeitos dos fármacos , Colo/fisiopatologia , Sulfato de Dextrana/toxicidade , Regulação para Baixo , Eletrofisiologia , Motilidade Gastrointestinal/efeitos dos fármacos , Masculino , Camundongos , Contração Muscular/efeitos dos fármacos , Músculo Liso/efeitos dos fármacos , Músculo Liso/fisiopatologia , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/metabolismo , Fatores de Tempo
5.
Biophys J ; 96(12): 4834-52, 2009 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-19527643

RESUMO

Spontaneously rhythmic pacemaker activity produced by interstitial cells of Cajal (ICC) is the result of the entrainment of unitary potential depolarizations generated at intracellular sites termed pacemaker units. In this study, we present a mathematical modeling framework that quantitatively represents the transmembrane ion flows and intracellular Ca2+ dynamics from a single ICC operating over the physiological membrane potential range. The mathematical model presented here extends our recently developed biophysically based pacemaker unit modeling framework by including mechanisms necessary for coordinating unitary potential events, such as a T-Type Ca2+ current, Vm-dependent K+ currents, and global Ca2+ diffusion. Model simulations produce spontaneously rhythmic slow wave depolarizations with an amplitude of 65 mV at a frequency of 17.4 cpm. Our model predicts that activity at the spatial scale of the pacemaker unit is fundamental for ICC slow wave generation, and Ca2+ influx from activation of the T-Type Ca2+ current is required for unitary potential entrainment. These results suggest that intracellular Ca2+ levels, particularly in the region local to the mitochondria and endoplasmic reticulum, significantly influence pacing frequency and synchronization of pacemaker unit discharge. Moreover, numerical investigations show that our ICC model is capable of qualitatively replicating a wide range of experimental observations.


Assuntos
Fenômenos Biofísicos , Potenciais da Membrana , Animais , Transporte Biológico , Cálcio/metabolismo , Membrana Celular , Fenômenos Eletrofisiológicos , Ativação do Canal Iônico , Modelos Biológicos , Técnicas de Patch-Clamp , Antro Pilórico/citologia
6.
Br J Pharmacol ; 153(6): 1259-71, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18204472

RESUMO

BACKGROUND AND PURPOSE: During the bladder filling phase, the volume of the urinary bladder increases dramatically, with only minimal increases in intravesical pressure. To accomplish this, the smooth muscle of the bladder wall must remain relaxed during bladder filling. However, the mechanisms responsible for the stabilization of bladder excitability during stretch are unclear. We hypothesized that stretch-dependent K(+) (TREK) channels in bladder smooth muscle cells may inhibit contraction in response to stretch. EXPERIMENTAL APPROACHES: Bladder tissues from mouse, guinea pig and monkey were used for molecular, patch clamp, mechanical, electrical, Ca(2+) imaging and cystometric responses to methionine and its derivatives, which are putative blockers of stretch-dependent K(+) (SDK) channels. KEY RESULTS: SDK channels are functionally expressed in bladder myocytes. The single channel conductance of SDK channels is 89pS in symmetrical K(+) conditions and is blocked by L-methionine. Expressed TREK-1 currents are also inhibited by L-methioninol. All three types of bladder smooth muscle cells from mouse, guinea pig and monkey expressed TREK-1 genes. L-methionine, methioninol and methionine methyl ester but not D-methionine increased contractility in concentration-dependent manner. Methioninol further increased contractility and depolarized the membrane in the presence of blockers of Ca(2+)-activated K(+) conductance. L-methionine induced Ca(2+) waves that spread long distances through the tissue under stretched conditions and were associated with strong contractions. In cystometric assays, methioninol injection increased bladder excitability mimicking overactive bladder activity. CONCLUSIONS AND IMPLICATIONS: Methioninol-sensitive K(+) (SDK, TREK-1) channels appear to be important to prevent spread of excitation through the syncitium during bladder filling.


Assuntos
Metionina/farmacologia , Músculo Liso/efeitos dos fármacos , Bloqueadores dos Canais de Potássio/farmacologia , Canais de Potássio de Domínios Poros em Tandem/efeitos dos fármacos , Animais , Cálcio/metabolismo , Relação Dose-Resposta a Droga , Feminino , Expressão Gênica , Cobaias , Macaca fascicularis , Masculino , Metionina/administração & dosagem , Metionina/análogos & derivados , Camundongos , Contração Muscular/efeitos dos fármacos , Músculo Liso/citologia , Técnicas de Patch-Clamp , Canais de Potássio de Domínios Poros em Tandem/metabolismo , Especificidade da Espécie , Bexiga Urinária/citologia , Bexiga Urinária/efeitos dos fármacos , Bexiga Urinária/metabolismo
7.
Neurogastroenterol Motil ; 16 Suppl 1: 100-5, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15066013

RESUMO

Considerable work has led many to conclude that interstitial cells of Cajal (ICC) are the pacemaker cells of the gastrointestinal (GI) tract. These cells form electrically coupled networks within the pacemaker regions of the GI tract, and ICC are electrically coupled to smooth muscle cells. ICC express unique ion channels that periodically produce inward (pacemaker) currents. Recent work has suggested that the inward current is produced by a calcium (Ca2+)-regulated, nonselective cation conductance. Channels responsible for this conductance oscillate in open probability in response to the periodic drop in intracellular Ca2+ concentration during the slow wave cycle. Pacemaker activity generates slow waves that are propagated actively through ICC networks. Depolarization coordinates the pacemaker activity through the ICC network by activating a dihydropyridine-resistant Ca2+ conductance. Entry of small amounts of Ca2+ into ICC entrains spontaneous pacemaker activity and produces cell-to-cell propagation of slow waves. This review discusses the mechanisms and conductances involved in generation and propagation of electrical slow waves in ICC.


Assuntos
Sistema Digestório/inervação , Motilidade Gastrointestinal/fisiologia , Músculo Liso/fisiologia , Neurônios/fisiologia , Transmissão Sináptica/fisiologia , Animais , Relógios Biológicos/fisiologia , Humanos , Canais Iônicos/fisiologia , Potenciais da Membrana/fisiologia
8.
Circ Res ; 94(5): 626-33, 2004 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-14739158

RESUMO

This study investigated regulation of L-type calcium channels (Cav1.2b) by acetylcholine (ACh) in rabbit portal vein myocytes. Whole-cell currents were recorded using 5 mmol/L barium as charge carrier. ACh (10 micromol/L) increased peak currents by 40%. This effect was not reversed by the selective muscarinic M3 receptor antagonist 4-DAMP (100 nmol/L) but was blocked by the M2 receptor antagonist methoctramine (5 micromol/L). The classical and novel protein kinase C (PKC) antagonist calphostin C (50 nmol/L) abolished ACh responses, whereas the classical PKC antagonist Gö6976 (200 nmol/L) had no effect. ACh responses were also abolished by the phosphatidylinositol 3-kinase (PI3K) inhibitor LY294002 (20 micromol/L), by the c-Src inhibitor PP2 (10 micromol/L) (but not the inactive analogue PP3), and by dialyzing cells with an antibody to the G-protein subunit Gbetagamma. Cells dialyzed with c-Src had significantly greater currents than control cells. Current enhancement persisted in the presence of LY294002, suggesting that c-Src is downstream of PI3K. Phorbol 12,13-dibutyrate (PDBu, 0.1 micromol/L) increased currents by 74%. This effect was abolished by calphostin C and reduced by Gö6976. The PDBu response was also reduced by PP2, and the PP2-insensitive component was blocked by Gö6976. In summary, these data suggest that ACh enhances Cav1.2b currents via M2 receptors that couple sequentially to Gbetagamma, PI3K, a novel PKC, and c-Src. PDBu stimulates the novel PKC/c-Src pathway along with a second pathway that is independent of c-Src and involves a classical PKC.


Assuntos
Canais de Cálcio Tipo L/fisiologia , Isoenzimas/fisiologia , Agonistas Muscarínicos/farmacologia , Miócitos de Músculo Liso/efeitos dos fármacos , Fosfatidilinositol 3-Quinases/fisiologia , Proteína Quinase C/fisiologia , Proteínas Proto-Oncogênicas pp60(c-src)/fisiologia , Receptor Muscarínico M2/fisiologia , Transdução de Sinais/fisiologia , Acetilcolina/farmacologia , Animais , Bário/metabolismo , Carbazóis/farmacologia , Células Cultivadas/efeitos dos fármacos , Células Cultivadas/fisiologia , Cromonas/farmacologia , Classe Ib de Fosfatidilinositol 3-Quinase , Diaminas/farmacologia , Inibidores Enzimáticos/farmacologia , Subunidades beta da Proteína de Ligação ao GTP/antagonistas & inibidores , Subunidades beta da Proteína de Ligação ao GTP/fisiologia , Subunidades gama da Proteína de Ligação ao GTP/antagonistas & inibidores , Subunidades gama da Proteína de Ligação ao GTP/fisiologia , Indóis/farmacologia , Ativação do Canal Iônico/efeitos dos fármacos , Ativação do Canal Iônico/fisiologia , Transporte de Íons/efeitos dos fármacos , Isoenzimas/antagonistas & inibidores , Masculino , Morfolinas/farmacologia , Antagonistas Muscarínicos/farmacologia , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/fisiologia , Naftalenos/farmacologia , Técnicas de Patch-Clamp , Dibutirato de 12,13-Forbol/farmacologia , Inibidores de Fosfoinositídeo-3 Quinase , Piperidinas/farmacologia , Veia Porta/citologia , Proteína Quinase C/antagonistas & inibidores , Proteínas Proto-Oncogênicas pp60(c-src)/antagonistas & inibidores , Pirimidinas/farmacologia , Coelhos , Receptor Muscarínico M2/agonistas , Receptor Muscarínico M2/antagonistas & inibidores , Receptor Muscarínico M3/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos
9.
Am J Physiol Cell Physiol ; 281(6): C2020-8, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11698261

RESUMO

A rapidly inactivating K(+) current (A-type current) participates in the regulation of colonic muscle excitability. We found 19-pS K(+) channels in cell-attached patches of murine colonic myocytes that activated and inactivated with kinetics similar to the A-type current. The A-type current in colonic myocytes is regulated by Ca(2+)/calmodulin-dependent protein kinase II. Therefore, we studied regulation of the 19-pS K(+) channels by Ca(2+)-dependent phosphorylation/dephosphorylation. The rates of inactivation of ensemble-averaged currents resulting from 19-pS K(+) channels were increased by the calmodulin antagonist W-7. Inhibitors of calcineurin, cyclosporin A and FK-506, slowed the inactivation of the 19-pS K(+) channels. Okadaic acid, an inhibitor of the calcineurin/inhibitor-1/protein phosphatase 1 cascade, also slowed inactivation of the 19-pS K(+) channels. Polymerase chain reaction detected transcripts encoding calcineurin A in isolated colonic smooth muscle cells, and immunohistochemical studies demonstrated specific expression of calcineurin A-like immunoreactivity in colonic muscle tissues and in colonic myocytes. These data, when considered with previous findings, suggest that Ca(2+)-dependent phosphorylation/dephosphorylation regulates the A-type current in murine colonic smooth muscle cells.


Assuntos
Cálcio/metabolismo , Colo/citologia , Músculo Liso/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Animais , Calcineurina/química , Calcineurina/metabolismo , Proteínas Quinases Dependentes de Cálcio-Calmodulina/antagonistas & inibidores , Colo/metabolismo , Ciclosporina/farmacologia , Inibidores Enzimáticos/farmacologia , Imunossupressores/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Músculo Liso/citologia , Músculo Liso/efeitos dos fármacos , Ácido Okadáico/farmacologia , Técnicas de Patch-Clamp , Fosforilação , Proteína Fosfatase 1 , Sulfonamidas/farmacologia , Tacrolimo/farmacologia
10.
J Biol Chem ; 276(47): 44338-46, 2001 Nov 23.
Artigo em Inglês | MEDLINE | ID: mdl-11560940

RESUMO

Potassium channels activated by membrane stretch may contribute to maintenance of relaxation of smooth muscle cells in visceral hollow organs. Previous work has identified K(+) channels in murine colon that are activated by stretch and further regulated by NO-dependent mechanisms. We have screened murine gastrointestinal, vascular, bladder, and uterine smooth muscles for the expression of TREK and TRAAK mRNA. Although TREK-1 was expressed in many of these smooth muscles, TREK-2 was expressed only in murine antrum and pulmonary artery. TRAAK was not expressed in any smooth muscle cells tested. Whole cell currents from TREK-1 expressed in mammalian COS cells were activated by stretch, and single channel recordings showed that the stretch-dependent conductance was due to 90 pS channels. Sodium nitroprusside (10(-6) or 10(-5) m) and 8-Br-cGMP (10(-4) or 10(-3) m) increased TREK-1 currents in perforated whole cell and single channel recordings. Mutation of the PKG consensus sequence at serine 351 blocked the stimulatory effects of sodium nitroprusside and 8-Br-cGMP on open probability without affecting the inhibitory effects of 8-Br-cAMP. TREK-1 encodes a component of the stretch-activated K(+) conductance in smooth muscles and may contribute to nitrergic inhibition of gastrointestinal muscles.


Assuntos
Proteínas Quinases Dependentes de GMP Cíclico/fisiologia , Músculo Liso/fisiologia , Óxido Nítrico/fisiologia , Canais de Potássio de Domínios Poros em Tandem , Canais de Potássio/fisiologia , Transmissão Sináptica/fisiologia , Animais , Sequência de Bases , Células COS , GMP Cíclico/farmacologia , Primers do DNA , Camundongos , Camundongos Endogâmicos BALB C , Músculo Liso/efeitos dos fármacos , Músculo Liso/inervação , Mutagênese Sítio-Dirigida , Nitroprussiato/farmacologia , Técnicas de Patch-Clamp , Reação em Cadeia da Polimerase Via Transcriptase Reversa
11.
Am J Physiol Gastrointest Liver Physiol ; 281(4): G964-73, 2001 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11557517

RESUMO

Small-conductance Ca2+-activated K+ (SK) channels are important participants in inhibitory neurotransmission in gastrointestinal smooth muscles. Three isoforms of an SK channel family were cloned from murine proximal colon smooth muscle. The transcripts encoding these subunits (SK1, SK2, and SK3) were detected in murine proximal colon and other peripheral tissues. The mRNA of each subunit was expressed at different levels in murine and canine colonic smooth muscles. The mRNA quantitative ratio of SK transcriptional expression in murine proximal colon is SK2 > SK3 > SK1; transcriptional expression of SK isoforms in canine proximal colon is minimal. SK3 immunohistochemical localization in murine small intestine (jejunum) and proximal colon showed immunoreactivity in circular and longitudinal muscularis. In transversely sectioned muscularis, staining was localized at the cell membrane in smooth muscle cells. Immunoreactivity was more intense in myenteric ganglia between longitudinal and circular muscularis and neuronal processes in circular and longitudinal muscularis. Transient expression of mSK1, mSK2, and mSK3 in COS cells resulted in Ca2+-activated voltage-independent channels. mSK1 is less sensitive to apamin compared with SK2 and showed intracellular Ca2+ sensitivity (10(-8) to 10(-6) M) in asymmetrical K+ (5/140 mM K+) gradients. Our results suggest that SK channel expression varies in colonic myocytes from different species and may contribute differentially to inhibitory junction potentials.


Assuntos
Cálcio/metabolismo , Colo/fisiologia , Músculo Liso/fisiologia , Canais de Potássio/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Células COS , Clonagem Molecular , Cães , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Técnicas de Patch-Clamp , Canais de Potássio/química , Canais de Potássio/genética , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Alinhamento de Sequência , Transmissão Sináptica/fisiologia , Distribuição Tecidual
12.
Am J Physiol Cell Physiol ; 281(3): C857-64, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11502563

RESUMO

We investigated the regulation of ATP-sensitive K(+) (K(ATP)) currents in murine colonic myocytes with patch-clamp techniques. Pinacidil (10(-5) M) activated inward currents in the presence of high external K(+) (90 mM) at a holding potential of -80 mV in dialyzed cells. Glibenclamide (10(-5) M) suppressed pinacidil-activated current. Phorbol 12,13-dibutyrate (PDBu; 2 x 10(-7) M) inhibited pinacidil-activated current. 4-alpha-Phorbol ester (5 x 10(-7) M), an inactive form of PDBu, had no effect on pinacidil-activated current. In cell-attached patches, the open probability of K(ATP) channels was increased by pinacidil, and PDBu suppressed openings of K(ATP) channels. When cells were pretreated with chelerythrine (10(-6) M) or calphostin C (10(-7) M), inhibition of the pinacidil-activated whole cell currents by PDBu was significantly reduced. In cells studied with the perforated patch technique, PDBu also inhibited pinacidil-activated current, and this inhibition was reduced by chelerythrine (10(-6) M). Acetylcholine (ACh; 10(-5) M) inhibited pinacidil-activated currents, and preincubation of cells with calphostin C (10(-7) M) decreased the effect of ACh. Cells dialyzed with protein kinase C epsilon-isoform (PKCepsilon) antibody had normal responses to pinacidil, but the effects of PDBu and ACh on K(ATP) were blocked in these cells. Immunofluorescence and Western blots showed expression of PKCepsilon in intact muscles and isolated smooth muscle cells of the murine proximal colon. These data suggest that PKC regulates K(ATP) in colonic muscle cells and that the effects of ACh on K(ATP) are largely mediated by PKC. PKCepsilon appears to be the major isozyme that regulates K(ATP) in murine colonic myocytes.


Assuntos
Trifosfato de Adenosina/metabolismo , Colo/fisiologia , Isoenzimas/metabolismo , Músculo Liso/fisiologia , Canais de Potássio/fisiologia , Proteína Quinase C/metabolismo , Acetilcolina/farmacologia , Alcaloides , Animais , Benzofenantridinas , Colo/efeitos dos fármacos , Colo/enzimologia , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Feminino , Glibureto/farmacologia , Técnicas In Vitro , Cinética , Masculino , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Camundongos , Camundongos Endogâmicos BALB C , Músculo Liso/efeitos dos fármacos , Músculo Liso/enzimologia , Naftalenos/farmacologia , Técnicas de Patch-Clamp , Fenantridinas/farmacologia , Dibutirato de 12,13-Forbol/farmacologia , Pinacidil/farmacologia , Canais de Potássio/efeitos dos fármacos , Proteína Quinase C-épsilon
13.
J Physiol ; 533(Pt 2): 341-55, 2001 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-11389196

RESUMO

1. Two components of voltage-gated, inward currents were observed from murine colonic myocytes. One component had properties of L-type Ca(2+) currents and was inhibited by nicardipine (5 x 10(-7) M). A second component did not 'run down' during dialysis and was resistant to nicardipine (up to 10(-6) M). The nicardipine-insensitive current was activated by small depolarizations above the holding potential and reversed near 0 mV. 2. This low-voltage-activated current (I(LVA)) was resolved with step depolarizations positive to -60 mV, and the current rapidly inactivated upon sustained depolarization. The voltage of half-inactivation was -65 mV. Inactivation and activation time constants at -45 mV were 86 and 15 ms, respectively. The half-recovery time from inactivation was 98 ms at -45 mV. I(LVA) peaked at -40 mV and the current reversed at 0 mV. 3. I(LVA) was inhibited by Ni(2+) (IC(50) = 1.4 x 10(-5) M), mibefradil (10(-6) to 10(-5) M), and extracellular Ba(2+). Replacement of extracellular Na(+) with N-methyl-D-glucamine inhibited I(LVA) and shifted the reversal potential to -7 mV. Increasing extracellular Ca(2+) (5 x 10(-3) M) increased the amplitude of I(LVA) and shifted the reversal potential to +22 mV. I(LVA) was also blocked by extracellular Cs(+) (10(-4) M) and Gd(3+) (10(-6) M). 4. Warming increased the rates of activation and deactivation without affecting the amplitude of the peak current. 5. We conclude that the second component of voltage-dependent inward current in murine colonic myocytes is not a 'T-type' Ca(2+) current but rather a novel, voltage-gated non-selective cation current. Activation of this current could be important in the recovery of membrane potential following inhibitory junction potentials in gastrointestinal smooth muscle or in mediating responses to agonists.


Assuntos
Canais de Cálcio Tipo L/fisiologia , Colo/citologia , Ativação do Canal Iônico/fisiologia , Músculo Liso/fisiologia , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Animais , Bário/farmacologia , Cálcio/farmacocinética , Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio Tipo T/genética , Césio/farmacologia , Gadolínio/farmacologia , Expressão Gênica , Ativação do Canal Iônico/efeitos dos fármacos , Mibefradil/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Músculo Liso/citologia , Músculo Liso/inervação , Inibição Neural/fisiologia , Nicardipino/farmacologia , Níquel/farmacologia , Técnicas de Patch-Clamp , Sódio/farmacologia , Temperatura
14.
J Physiol ; 533(Pt 1): 155-63, 2001 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-11351024

RESUMO

Gastrointestinal muscles are able to maintain negative resting membrane potentials in spite of stretch. We investigated whether stretch-dependent K+ channels might contribute to myogenic regulation of smooth muscle cells from the mouse colon. Negative pressure applied to on-cell membrane patches activated K+ channels that were voltage independent and had a slope conductance of 95 pS in symmetrical K+ gradients. The effects of negative pressure on open probability were graded as a function of pressure and reversible when atmospheric pressure was restored. Cell elongation activated K+ channels with the same properties as those activated by negative pressure, suggesting that the channels were stretch-dependent K+ (SDK) channels. Channels with the same properties were maximally activated by patch excision, suggesting that either an intracellular messenger or interactions with the cytoskeleton regulate open probability. Internal 4-aminopyridine, Ca2+ (10(-8) to 10(-6) M), and tetraethylammonium (internal or external) were without effect on SDK channels. Nitric oxide donors (and cell-permeant cGMP analogues) activated SDK channels, suggesting that these channels may mediate a portion of the enteric inhibitory neural response in colonic muscles. In summary, SDK channels are an important conductance expressed by colonic muscle cells. SDK channels may stabilize membrane potential during dynamic changes in cell length and mediate responses to enteric neurotransmitters.


Assuntos
Colo/citologia , Colo/fisiologia , Músculo Liso/citologia , Músculo Liso/fisiologia , Canais de Potássio/fisiologia , 8-Bromo Monofosfato de Adenosina Cíclica/farmacologia , Animais , Feminino , Ativação do Canal Iônico/efeitos dos fármacos , Ativação do Canal Iônico/fisiologia , Masculino , Potenciais da Membrana/fisiologia , Camundongos , Camundongos Endogâmicos BALB C , Nitroprussiato/farmacologia , Técnicas de Patch-Clamp , Estresse Mecânico , Vasodilatadores/farmacologia
15.
J Physiol ; 527 Pt 1: 149-62, 2000 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-10944178

RESUMO

1. Electrical rhythmicity (slow waves) in gastrointestinal muscles (GI) is generated by interstitial cells of Cajal (ICC). Cultured ICC from the murine small intestine were studied with the patch-clamp technique to characterize regulation of pacemaker currents by cyclic nucleotides. Cyclic nucleotide agonists were also tested on intact strips of murine small intestine. 2. Nitric oxide donors slowed the frequency of pacemaker currents in a concentration-dependent manner. These effects depended on cGMP formation and were reduced by 1H-[1,2, 4]oxadiazolo[4,3-a]quinoxalin-1-one (ODQ). The effects of nitric oxide donors were mimicked by membrane-permeable analogues of cGMP. The specific cGMP phosphodiesterase inhibitor zaprinast reduced the frequency of spontaneous pacemaker currents. 3. The cGMP-dependent effects on pacemaker currents were not affected by okadaic acid or KT-5823, an inhibitor of protein kinase G. 4. Forskolin, but not dideoxy forskolin, reduced the frequency of spontaneous pacemaker activity and activated a sustained outward current. The latter was likely to be due to ATP-dependent K+ channels because it was blocked by glibenclamide. 5. The effects of forskolin were not mimicked by membrane-permeable cAMP analogues. A membrane-permeable inhibitor of protein kinase A, myristoylated PKA inhibitor, and the adenylyl cyclase inhibitor SQ-22536, had no effect on responses to forskolin. 6. Responses of intact muscles to cGMP and cAMP agonists were similar to the responses of pacemaker cells. Changes in resting membrane potential and slow wave amplitude, however, were noted in intact jejunal muscles that were not observed in ICC. Differences in responses may have been due to the effects of cyclic nucleotide agonists on smooth muscle cells that would sum with responses of ICC in intact jejunal muscle strips. 7. A cGMP-dependent mechanism regulates slow wave frequency, but this occurs through direct action of cGMP not via protein phosphorylation. Regulation of pacemaker currents by cAMP-dependent mechanisms was not observed.


Assuntos
GMP Cíclico/fisiologia , Condutividade Elétrica , Intestino Delgado/fisiologia , Nucleotídeos Cíclicos/fisiologia , Inibidores de Adenilil Ciclases , Animais , Células Cultivadas , Colforsina/farmacologia , AMP Cíclico/análogos & derivados , AMP Cíclico/metabolismo , AMP Cíclico/fisiologia , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , GMP Cíclico/análogos & derivados , GMP Cíclico/antagonistas & inibidores , Proteínas Quinases Dependentes de GMP Cíclico/antagonistas & inibidores , Motilidade Gastrointestinal , Guanilato Ciclase/antagonistas & inibidores , Intestino Delgado/citologia , Intestino Delgado/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos BALB C , Óxido Nítrico/metabolismo , Nitroprussiato/farmacologia , Nucleotídeos Cíclicos/agonistas , Nucleotídeos Cíclicos/antagonistas & inibidores , Técnicas de Patch-Clamp , Inibidores de Fosfodiesterase/farmacologia
16.
J Physiol ; 525 Pt 2: 355-61, 2000 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-10835039

RESUMO

Pacemaker cells, known as interstitial cells of Cajal (ICC), generate electrical rhythmicity in the gastrointestinal tract. Pacemaker currents in ICC result from the activation of a voltage-independent, non-selective cation conductance, but the timing mechanism responsible for periodic activation of the pacemaker current is unknown. Previous studies suggest that pacemaking in ICC is dependent upon metabolic activity 1y1yand1 Ca2+ release from intracellular stores. We tested the hypothesis that mitochondrial Ca2+ handling may underlie the dependence of gastrointestinal pacemaking on oxidative metabolism. Pacemaker currents occurred spontaneously in cultured ICC and were associated with mitochondrial Ca2+ transients. Inhibition of the electrochemical gradient across the inner mitochondrial membrane blocked Ca2+ uptake and pacemaker currents in cultured ICC and blocked slow wave activity in intact gastrointestinal muscles from mouse, dog and guinea-pig. Pacemaker currents and rhythmic mitochondrial Ca2+ uptake in ICC were also blocked by inhibitors of IP3-dependent release of Ca2+ from the endoplasmic reticulum and by inhibitors of endoplasmic reticulum Ca2+ reuptake. Our data suggest that integrated Ca2+ handling by endoplasmic reticulum and mitochondria is a prerequisite of electrical pacemaking in the gastrointestinal tract.


Assuntos
Cálcio/metabolismo , Sistema Digestório/citologia , Sistema Digestório/inervação , Retículo Endoplasmático/metabolismo , Mitocôndrias/metabolismo , Animais , Relógios Biológicos , Células Cultivadas , Sistema Digestório/metabolismo , Cães , Transporte de Íons , Camundongos , Modelos Biológicos , Músculo Liso/citologia , Músculo Liso/inervação , Músculo Liso/metabolismo
17.
J Physiol ; 524 Pt 2: 331-7, 2000 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-10766915

RESUMO

1. Ca2+ regulates the activity of small conductance Ca2+-activated K+ (SK) channels via calmodulin-dependent binding. We investigated whether other forms of Ca2+-dependent regulation might control the open probability of SK channels. 2. Under whole-cell patch-clamp conditions, spontaneous openings of SK channels can be resolved as charybdotoxin-insensitive spontaneous transient outward currents (STOCs). The Ca2+-calmodulin-dependent (CaM) protein kinase II inhibitor KN-93 reduced the occurrence of charybdotoxin-insensitive STOCs. 3. The charybdotoxin-insensitive STOCs are related to spontaneous, local release of Ca2+. KN-93 did not affect spontaneous Ca2+-release events. 4. KN-93 and W-7, a calmodulin inhibitor, decreased the open probability of SK channels in on-cell patches but not in excised patches. 5. Application of autothiophosphorlated CaM kinase II to the cytoplasmic surface of excised patches increased the open probalibity of SK channels. Boiled CaM kinase II had no effect. 6. We conclude that CaM kinase II regulates SK channels in murine coloni myocytes. This mechanism provides a secondary means of regulation, increasing the impact of a given Ca2+ transient on SK channel open probability.


Assuntos
Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Colo/metabolismo , Canais de Potássio Cálcio-Ativados , Canais de Potássio/fisiologia , Animais , Benzilaminas/farmacologia , Proteínas Quinases Dependentes de Cálcio-Calmodulina/antagonistas & inibidores , Células Cultivadas , Charibdotoxina/farmacologia , Colo/citologia , Colo/fisiologia , Eletrofisiologia , Inibidores Enzimáticos/farmacologia , Processamento de Imagem Assistida por Computador , Potenciais da Membrana/fisiologia , Camundongos , Camundongos Endogâmicos BALB C , Microscopia Confocal , Músculo Liso/citologia , Músculo Liso/metabolismo , Técnicas de Patch-Clamp , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/fisiologia , Canais de Potássio Ativados por Cálcio de Condutância Baixa , Sulfonamidas/farmacologia
18.
Am J Physiol Cell Physiol ; 278(2): C352-62, 2000 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10666031

RESUMO

Spontaneous transient outward currents (STOCs) were recorded from smooth muscle cells of the guinea pig taenia coli using the whole cell patch-clamp technique. STOCs were resolved at potentials positive to -50 mV. Treating cells with caffeine (1 mM) caused a burst of outward currents followed by inhibition of STOCs. Replacing extracellular Ca(2+) with equimolar Mn(2+) caused STOCs to "run down. " Iberiotoxin (200 nM) or charybdotoxin (ChTX; 200 nM) inhibited large-amplitude STOCs, but small-amplitude "mini-STOCs" remained in the presence of these drugs. Mini-STOCs were reduced by apamin (500 nM), an inhibitor of small-conductance Ca(2+)-activated K(+) channels (SK channels). Application of ATP or 2-methylthioadenosine 5'-triphosphate (2-MeS-ATP) increased the frequency of STOCs. The effects of 2-MeS-ATP persisted in the presence of charybdotoxin but were blocked by combination of ChTX (200 nM) and apamin (500 nM). 2-MeS-ATP did not increase STOCs in the presence of pyridoxal phosphate 6-azophenyl-2',4'-disulfonic acid, a P(2) receptor blocker. Similarly, pretreatment of cells with U-73122 (1 microM), an inhibitor of phospholipase C (PLC), abolished the effects of 2-MeS-ATP. Xestospongin C, an inositol 1,4,5-trisphosphate (IP(3)) receptor blocker, attenuated STOCs, but these events were not affected by ryanodine. The data suggest that purinergic activation through P(2Y) receptors results in localized Ca(2+) release via PLC- and IP(3)-dependent mechanisms. Release of Ca(2+) is coupled to STOCs, which are composed of currents mediated by large-conductance Ca(2+)-activated K(+) channels and SK channels. The latter are thought to mediate hyperpolarization and relaxation responses of gastrointestinal muscles to inhibitory purinergic stimulation.


Assuntos
Cálcio/farmacologia , Colo/metabolismo , Músculo Liso/metabolismo , Canais de Potássio Cálcio-Ativados , Receptores Purinérgicos P2/metabolismo , Éster Metílico do Ácido 3-Piridinacarboxílico, 1,4-Di-Hidro-2,6-Dimetil-5-Nitro-4-(2-(Trifluormetil)fenil)/farmacologia , Trifosfato de Adenosina/análogos & derivados , Trifosfato de Adenosina/farmacologia , Animais , Apamina/farmacologia , Cafeína/farmacologia , Agonistas dos Canais de Cálcio/farmacologia , Canais de Cálcio/fisiologia , Estimulantes do Sistema Nervoso Central/farmacologia , Charibdotoxina/farmacologia , Colo/química , Colo/citologia , Estrenos/farmacologia , Feminino , Cobaias , Técnicas In Vitro , Receptores de Inositol 1,4,5-Trifosfato , Ativação do Canal Iônico/efeitos dos fármacos , Ativação do Canal Iônico/fisiologia , Canais de Potássio Ativados por Cálcio de Condutância Alta , Compostos Macrocíclicos , Masculino , Fibras Musculares Esqueléticas/química , Fibras Musculares Esqueléticas/metabolismo , Músculo Liso/química , Músculo Liso/citologia , Oxazóis/farmacologia , Técnicas de Patch-Clamp , Inibidores de Fosfodiesterase/farmacologia , Canais de Potássio/fisiologia , Pirrolidinonas/farmacologia , Receptores Citoplasmáticos e Nucleares/fisiologia , Rianodina/farmacologia , Tionucleotídeos/farmacologia
19.
Neurogastroenterol Motil ; 11(5): 311-38, 1999 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-10520164

RESUMO

Interstitial cells of Cajal (ICC) are the pacemakers in gastrointestinal (GI) muscles, and these cells also mediate or transduce inputs from the enteric nervous system. Different classes of ICC are involved in pacemaking and neurotransmission. ICC express specific ionic conductances that make them unique in their ability to generate and propagate slow waves in GI muscles or transduce neural inputs. Much of what we know about the function of ICC comes from developmental studies that were made possible by the discoveries that ICC express c-kit and proper development of ICC depends upon signalling via the Kit receptor pathway. Manipulating Kit signalling with reagents to block the receptor or downstream signalling pathways or by using mutant mice in which Kit or its ligand, stem cell factor, are defective has allowed novel studies into the specific functions of the different classes of ICC in several regions of the GI tract. Kit is also a surface antigen that can be used to conveniently label ICC in GI muscles. Immunohistochemical studies using Kit antibodies have expanded our knowledge about the ICC phenotype, the structure of ICC networks, the interactions of ICC with other cells in the gut wall, and the loss of ICC in some clinical disorders. Preparations made devoid of ICC have also allowed analysis of the consequences of losing specific classes of ICC on GI motility. This review describes recent advances in our knowledge about the development and plasticity of ICC and how developmental studies have contributed to our understanding of the functions of ICC. We have reviewed the clinical literature and discussed how loss or defects in ICC affect GI motor function.


Assuntos
Sistema Nervoso Entérico , Motilidade Gastrointestinal/fisiologia , Plasticidade Neuronal/fisiologia , Animais , Sistema Nervoso Entérico/citologia , Sistema Nervoso Entérico/crescimento & desenvolvimento , Sistema Nervoso Entérico/fisiologia
20.
J Physiol ; 518(Pt 1): 247-56, 1999 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-10373706

RESUMO

1. The membrane potential of gastrointestinal smooth muscles determines the open probability of ion channels involved in rhythmic electrical activity. The role of Ba2+-sensitive K+ conductances in the maintenance of membrane potential was examined in canine proximal colon circular muscle. 2. Application of Ba2+ (1-100 microM) to strips of tunica muscularis produced depolarization of cells along the submucosal surface of the circular muscle layer. Significantly higher concentrations of Ba2+ were needed to depolarize preparations from which the submucosal and myenteric pacemaker regions were removed. 3. Elevation of extracellular [K+]o (from 5.9 to 12 mM) brought membrane potentials closer to EK (the Nernst potential for K+ ions), suggesting activation of a K+ conductance. This occurred at potentials much more negative than the activation range for delayed rectifier channels (Kv). 4. Forskolin (1 microM) caused hyperpolarization and a leftward shift in the dose-response relationship for Ba2+, suggesting that forskolin may activate a Ba2+-sensitive conductance. 5. Patch-clamp recordings from interstitial cells of Cajal (ICC) revealed the presence of a Ba2+-sensitive inward rectifier potassium conductance. Far less of this conductance was present in smooth muscle cells. 6. Kir2.1 was expressed in the circular muscle layer of the canine proximal colon, duodenum, jejunum and ileum. Kir2.1 mRNA was expressed in greater abundance along the submucosal surface of the circular muscle layer in the colon. 7. These results demonstrate that ICC express a Ba2+-sensitive conductance (possibly encoded by Kir2.1). This conductance contributes to the generation and maintenance of negative membrane potentials between slow waves.


Assuntos
Colo/fisiologia , Músculo Liso/fisiologia , Canais de Potássio Corretores do Fluxo de Internalização , Canais de Potássio/fisiologia , Animais , Bário/farmacologia , Colforsina/farmacologia , Colo/efeitos dos fármacos , Cães , Estimulação Elétrica , Eletrofisiologia , Feminino , Técnicas In Vitro , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/fisiologia , Masculino , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Músculo Liso/efeitos dos fármacos , Técnicas de Patch-Clamp , Potássio/farmacologia , Canais de Potássio/efeitos dos fármacos , Canais de Potássio/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...