Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Biochim Biophys Acta ; 1828(4): 1214-21, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23305758

RESUMO

G-protein activated inwardly rectifying K(+) channels (GIRKs) of the heterotetrameric GIRK1/GIRK4 composition mediate I(K+ACh) in atrium and are regulated by cAMP dependent protein kinase (PKA). Phosphorylation of GIRK1/GIRK4 complexes promotes the activation of the channel by the G-protein Gßγ-dimer ("heterologous facilitation"). Previously we reported that 3 serines/threonines (S/Ts) within the GIRK1 subunit are phosphorylated by the catalytic subunit of PKA (PKA-cs) in-vitro and are responsible for the acute functional effects exerted by PKA on the homooligomeric GIRK1(F137S) (GIRK1(⁎)) channel. Here we report that homooligomeric GIRK4(WT) and GIRK4(S143T) (GIRK4(⁎)) channels are clearly regulated by PKA phosphorylation. Heterooligomeric channels of the GIRK1(S385CS401CT407C)/GIRK4(WT) composition, where the GIRK1 subunit is devoid of PKA mediated phosphorylation, exhibited reduced but still significant acute effects (reduction during agonist application was ≈49% compared to GIRK1(WT)/GIRK4(WT)). Site directed mutagenesis of truncated cytosolic regions of GIRK4 revealed four serines/threonines (S/Ts) that were heavily phosphorylated by PKA-cs in vitro. Two of them were found to be responsible for the acute effects exerted by PKA in vivo, since the effect of cAMP injection was reduced by ≈99% in homooligomeric GIRK4(⁎T199CS412C) channels. Coexpression of GIRK1(WT)/GIRK4(T199CS412C) reduced the acute effect by ≈65%. Only channels of the GIRK1(S385CS401CT407C)/GIRK4(T199CS412C) composition were practically devoid of PKA mediated effects (reduction by ≈97%), indicating that both subunits contribute to the heterologous facilitation of I(K+ACh).


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/química , Sequência de Aminoácidos , Animais , Dados de Sequência Molecular , Fosforilação , Multimerização Proteica , Subunidades Proteicas , Xenopus laevis
2.
Cardiovasc Res ; 93(1): 41-9, 2012 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-21960685

RESUMO

AIMS: High-density lipoprotein (HDL) is known for its cardioprotective properties independent from its cholesterol transport activity. These properties are mediated by activation of kinases such as protein kinase C (PKC). Connexin43 (Cx43) is a gap junction protein present in ventricular cardiomyocytes. PKC-dependent phosphorylation modifies Cx43 gap junction channel properties and is involved in cardioprotection. We hypothesized that cardioprotective properties of HDL may be mediated in part by affecting Cx43 gap junction channels. METHODS AND RESULTS: Neonatal rat cardiomyocytes were treated with HDL and Cx43 phosphorylation was evaluated by western blotting and immunofluorescence. We found that HDL promoted phosphorylation of Cx43 with a maximal induction at 5 min, which was inhibited by pre-treatment with various PKC inhibitors. Sphingosine-1-phosphate (S1P), a component of HDL, induced effects that were similar to those of HDL. These compounds significantly reduced diffusion of fluorescent dye among cardiomyocytes (∼50%) which could be prevented by PKC inhibition. As observed during optical recordings of transmembrane voltage, HDL and S1P depressed impulse conduction only minimally (<5%). Moreover, 5 min of HDL and S1P treatment at the onset of reperfusion significantly reduced infarct size (∼50%) in response to 30 min ischaemia in ex vivo experiments. CONCLUSION: Short-term treatment with HDL or S1P induces phosphorylation of Cx43 by a PKC-dependent pathway. HDL-induced phosphorylation of Cx43 reduced the diffusion of large tracer molecules between cells, whereas impulse conduction was maintained. Moreover, 5 min treatment with HDL confers cardioprotection against ischaemia/reperfusion injury. These results link Cx43 for the first time to the short-term cardioprotective effects of HDL.


Assuntos
Conexina 43/metabolismo , Lipoproteínas HDL/metabolismo , Lipoproteínas HDL/farmacologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Animais , Cardiotônicos/metabolismo , Cardiotônicos/farmacologia , Comunicação Celular/efeitos dos fármacos , Morte Celular/efeitos dos fármacos , Células Cultivadas , Junções Comunicantes/efeitos dos fármacos , Junções Comunicantes/metabolismo , Técnicas In Vitro , Lisofosfolipídeos/metabolismo , Lisofosfolipídeos/farmacologia , Traumatismo por Reperfusão Miocárdica/tratamento farmacológico , Traumatismo por Reperfusão Miocárdica/patologia , Fosforilação , Proteína Quinase C/metabolismo , Ratos , Ratos Sprague-Dawley , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Esfingosina/farmacologia
3.
Circ Res ; 109(10): 1120-31, 2011 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-21921266

RESUMO

RATIONALE: Myofibroblasts typically appear in the myocardium after insults to the heart like mechanical overload and infarction. Apart from contributing to fibrotic remodeling, myofibroblasts induce arrhythmogenic slow conduction and ectopic activity in cardiomyocytes after establishment of heterocellular electrotonic coupling in vitro. So far, it is not known whether α-smooth muscle actin (α-SMA) containing stress fibers, the cytoskeletal components that set myofibroblasts apart from resident fibroblasts, are essential for myofibroblasts to develop arrhythmogenic interactions with cardiomyocytes. OBJECTIVE: We investigated whether pharmacological ablation of α-SMA containing stress fibers by actin-targeting drugs affects arrhythmogenic myofibroblast-cardiomyocyte cross-talk. METHODS AND RESULTS: Experiments were performed with patterned growth cell cultures of neonatal rat ventricular cardiomyocytes coated with cardiac myofibroblasts. The preparations exhibited slow conduction and ectopic activity under control conditions. Exposure to actin-targeting drugs (Cytochalasin D, Latrunculin B, Jasplakinolide) for 24 hours led to disruption of α-SMA containing stress fibers. In parallel, conduction velocities increased dose-dependently to values indistinguishable from cardiomyocyte-only preparations and ectopic activity measured continuously over 24 hours was completely suppressed. Mechanistically, antiarrhythmic effects were due to myofibroblast hyperpolarization (Cytochalasin D, Latrunculin B) and disruption of heterocellular gap junctional coupling (Jasplakinolide), which caused normalization of membrane polarization of adjacent cardiomyocytes. CONCLUSIONS: The results suggest that α-SMA containing stress fibers importantly contribute to myofibroblast arrhythmogeneicity. After ablation of this cytoskeletal component, cells lose their arrhythmic effects on cardiomyocytes, even if heterocellular electrotonic coupling is sustained. The findings identify α-SMA containing stress fibers as a potential future target of antiarrhythmic therapy in hearts undergoing structural remodeling.


Assuntos
Actinas/antagonistas & inibidores , Antiarrítmicos/farmacologia , Arritmias Cardíacas/tratamento farmacológico , Miócitos Cardíacos/efeitos dos fármacos , Miofibroblastos/efeitos dos fármacos , Fibras de Estresse/efeitos dos fármacos , Actinas/metabolismo , Potenciais de Ação , Animais , Animais Recém-Nascidos , Arritmias Cardíacas/metabolismo , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Comunicação Celular/efeitos dos fármacos , Forma Celular/efeitos dos fármacos , Células Cultivadas , Técnicas de Cocultura , Citocalasina D/farmacologia , Depsipeptídeos/farmacologia , Relação Dose-Resposta a Droga , Junções Comunicantes/efeitos dos fármacos , Junções Comunicantes/metabolismo , Miócitos Cardíacos/metabolismo , Miofibroblastos/metabolismo , Fenótipo , Ratos , Ratos Wistar , Fibras de Estresse/metabolismo , Tiazolidinas/farmacologia , Fatores de Tempo
4.
J Biol Chem ; 284(8): 5186-94, 2009 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-19116207

RESUMO

Ryanodine receptors (RyR) are Ca(2+) channels that mediate Ca(2+) release from intracellular stores in response to diverse intracellular signals. In RINm5F insulinoma cells, caffeine, and 4-chloro-m-cresol (4CmC), agonists of RyR, stimulated Ca(2+) entry that was independent of store-operated Ca(2+) entry, and blocked by prior incubation with a concentration of ryanodine that inactivates RyR. Patch-clamp recording identified small numbers of large-conductance (gamma(K) = 169 pS) cation channels that were activated by caffeine, 4CmC or low concentrations of ryanodine. Similar channels were detected in rat pancreatic beta-cells. In RINm5F cells, the channels were blocked by cytosolic, but not extracellular, ruthenium red. Subcellular fractionation showed that type 3 IP(3) receptors (IP(3)R3) were expressed predominantly in endoplasmic reticulum, whereas RyR2 were present also in plasma membrane fractions. Using RNAi selectively to reduce expression of RyR1, RyR2, or IP(3)R3, we showed that RyR2 mediates both the Ca(2+) entry and the plasma membrane currents evoked by agonists of RyR. We conclude that small numbers of RyR2 are selectively expressed in the plasma membrane of RINm5F pancreatic beta-cells, where they mediate Ca(2+) entry.


Assuntos
Cálcio/metabolismo , Membrana Celular/metabolismo , Células Secretoras de Insulina/metabolismo , Canal de Liberação de Cálcio do Receptor de Rianodina/biossíntese , Animais , Cafeína/farmacocinética , Agonistas dos Canais de Cálcio/farmacologia , Linhagem Celular Tumoral , Estimulantes do Sistema Nervoso Central/farmacologia , Cresóis/farmacologia , Relação Dose-Resposta a Droga , Fungicidas Industriais/farmacologia , Receptores de Inositol 1,4,5-Trifosfato/agonistas , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Masculino , Ratos , Ratos Wistar , Rianodina/farmacologia
5.
J Recept Signal Transduct Res ; 27(5-6): 369-82, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-18097938

RESUMO

Four isoforms of GIRK channels (GIRK1-4) have been described in humans. In addition, several splice variants of more or less unknown function have been identified from several tissues and species. In our study, we investigated the structure and function of a new variant of GIRK1 that has been isolated from rat brain. Because of wide similarities with a previously described variant, we also named it GIRK1d. This variant lacks a region corresponding to exon 2 of full-length GIRK1, leading to a truncated GIRK1 that lacks the main part of the C-terminus. To study GIRK1d we used the Xenopus laevis expression system, the two-electrode voltage clamp method, and confocal laser scan microscopy. We found that our GIRK1d variant preferentially binds GIRK2 or GIRK4 over GIRK1. Furthermore, it largely reduces conductances mediated by GIRK1/2 or GIRK1/4 hetero-multimeric channels when coexpressed and nearly totally abolishes currents when replacing GIRK1 in hetero-multimeric channels.


Assuntos
Química Encefálica , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/genética , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/metabolismo , Animais , Técnicas de Cultura de Células , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/isolamento & purificação , Oócitos/metabolismo , Técnicas de Patch-Clamp , Splicing de RNA , Ratos , Proteínas Recombinantes de Fusão/metabolismo , Xenopus laevis/metabolismo
6.
J Recept Signal Transduct Res ; 27(2-3): 113-24, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17613724

RESUMO

A large variety of cation transport systems are involved in the regulation of calcium homeostasis in endothelial cells. The focus of the present study is to determine the contribution of nonselective cation channels from the TRP (transient receptor potential) family to cellular calcium homeostasis of porcine aortic endothelial cells (PAEC). One member of the TRPV (vanniloid) subfamily, TRPV4, has previously been shown to be involved in cation transport induced by a large variety of stimulations including osmolarity, temperature, mechanical stress, and phosphorylation. Here, we demonstrate the existence of several TRP proteins, including TRPV4, in PAEC using RT-PCR. To test whether this channel is functional, we performed FURA-2 calcium measurements and whole-cell patch-clamp experiments. We observed the induction of large calcium signals following mechanical stress, altered extracellular temperature, and the selective TRPV4 activator 4-alpha -PDD. These effects were diminished in the presence of the TRPV4 inhibitor miconazole, suggesting the involvement of this channel in mediating endothelial calcium signals. The large amounts of transported calcium and the short signaling ways suggest a potentially important role of this channel in many physiological processes.


Assuntos
Sinalização do Cálcio , Cálcio/metabolismo , Células Endoteliais/metabolismo , Homeostase , Suínos/metabolismo , Canais de Cátion TRPV/metabolismo , Animais , Aorta/citologia , Aorta/efeitos dos fármacos , Aorta/metabolismo , Sinalização do Cálcio/efeitos dos fármacos , Células Cultivadas , Células Endoteliais/efeitos dos fármacos , Homeostase/efeitos dos fármacos , Miconazol/farmacologia , Ésteres de Forbol/farmacologia , Temperatura
7.
Am J Physiol Cell Physiol ; 293(2): C783-9, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17522141

RESUMO

The blocking efficacy of 4,9-anhydro-TTX (4,9-ah-TTX) and TTX on several isoforms of voltage-dependent sodium channels, expressed in Xenopus laevis oocytes, was tested (Na(v1.2), Na(v1.3), Na(v1.4), Na(v1.5), Na(v1.6), Na(v1.7), and Na(v1.8)). Generally, TTX was 40-231 times more effective, when compared with 4,9-ah-TTX, on a given isoform. An exception was Na(v1.6), where 4,9-ah-TTX in nanomole per liter concentrations sufficed to result in substantial block, indicating that 4,9-ah-TTX acts specifically at this peculiar isoform. The IC(50) values for TTX/4,9-ah-TTX were as follows (in nmol/l): 7.8 +/- 1.3/1,260 +/- 121 (Na(v1.2)), 2.8 +/- 2.3/341 +/- 36 (Na(v1.3)), 4.5 +/- 1.0/988 +/- 62 (Na(v1.4)), 1,970 +/- 565/78,500 +/- 11,600 (Na(v1.5)), 3.8 +/- 1.5/7.8 +/- 2.3 (Na(v1.6)), 5.5 +/- 1.4/1,270 +/- 251 (Na(v1.7)), and 1,330 +/- 459/>30,000 (Na(v1.8)). Analysis of approximal half-maximal doses of both compounds revealed minor effects on voltage-dependent activation only, whereas steady-state inactivation was shifted to more negative potentials by both TTX and 4,9-ah-TTX in the case of the Na(v1.6) subunit, but not in the case of other TTX-sensitive ones. TTX shifted steady-state inactivation also to more negative potentials in case of the TTX-insensitive Na(v1.5) subunit, where it also exerted profound effects on the time course of recovery from inactivation. Isoform-specific interaction of toxins with ion channels is frequently observed in the case of proteinaceous toxins. Although the sensitivity of Na(v1.1) to 4,9-ah-TTX is not known, here we report evidence on a highly isoform-specific TTX analog that may well turn out to be an invaluable tool in research for the identification of Na(v1.6)-mediated function, but also for therapeutic intervention.


Assuntos
Proteínas do Tecido Nervoso/efeitos dos fármacos , Bloqueadores dos Canais de Sódio/farmacologia , Canais de Sódio/efeitos dos fármacos , Sódio/metabolismo , Tetrodotoxina/análogos & derivados , Animais , Relação Dose-Resposta a Droga , Humanos , Cinética , Potenciais da Membrana/efeitos dos fármacos , Camundongos , Microinjeções , Proteínas Musculares/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.2 , Canal de Sódio Disparado por Voltagem NAV1.3 , Canal de Sódio Disparado por Voltagem NAV1.5 , Canal de Sódio Disparado por Voltagem NAV1.6 , Canal de Sódio Disparado por Voltagem NAV1.7 , Canal de Sódio Disparado por Voltagem NAV1.8 , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Oócitos , Ratos , Bloqueadores dos Canais de Sódio/metabolismo , Canais de Sódio/genética , Canais de Sódio/metabolismo , Tetrodotoxina/metabolismo , Tetrodotoxina/farmacologia , Xenopus laevis
8.
J Biol Chem ; 281(19): 13588-13595, 2006 May 12.
Artigo em Inglês | MEDLINE | ID: mdl-16537542

RESUMO

Canonical transient receptor potential proteins (TRPC) have been proposed to form homo- or heteromeric cation channels in a variety of tissues, including the vascular endothelium. Assembly of TRPC multimers is incompletely understood. In particular, heteromeric assembly of distantly related TRPC isoforms is still a controversial issue. Because we have previously suggested TRPC proteins as the basis of the redox-activated cation conductance of porcine aortic endothelial cells (PAECs), we set out to analyze the TRPC subunit composition of endogenous endothelial TRPC channels and report here on a redox-sensitive TRPC3-TRPC4 channel complex. The ability of TRPC3 and TRPC4 proteins to associate and to form a cation-conducting pore complex was supported by four lines of evidence as follows: 1) Co-immunoprecipitation experiments in PAECs and in HEK293 cells demonstrated the association of TRPC3 and TRPC4 in the same complex. 2) Fluorescence resonance energy transfer analysis demonstrated TRPC3-TRPC4 association, involving close proximity between the N terminus of TRPC4 and the C terminus of TRPC3 subunits. 3) Co-expression of TRPC3 and TRPC4 in HEK293 cells generated a channel that displayed distinct biophysical and regulatory properties. 4) Expression of dominant-negative TRPC4 proteins suppressed TRPC3-related channel activity in the HEK293 expression system and in native endothelial cells. Specifically, an extracellularly hemagglutinin (HA)-tagged TRPC4 mutant, which is sensitive to blockage by anti-HA-antibody, was found to transfer anti-HA sensitivity to both TRPC3-related currents in the HEK293 expression system and the redox-sensitive cation conductance of PAECs. We propose TRPC3 and TRPC4 as subunits of native endothelial cation channels that are governed by the cellular redox state.


Assuntos
Células Endoteliais/metabolismo , Canais de Cátion TRPC/química , Canais de Cátion TRPC/metabolismo , Animais , Linhagem Celular , Regulação da Expressão Gênica , Humanos , Potenciais da Membrana , Camundongos , Oxirredução , Estresse Oxidativo , Ligação Proteica , Estrutura Quaternária de Proteína , Suínos , Canais de Cátion TRPC/genética
9.
Biochem J ; 396(1): 147-55, 2006 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-16448384

RESUMO

TRPC3 (canonical transient receptor potential protein 3) has been suggested to be a component of cation channel complexes that are targeted to cholesterol-rich lipid membrane microdomains. In the present study, we investigated the potential role of membrane cholesterol as a regulator of cellular TRPC3 conductances. Functional experiments demonstrated that cholesterol loading activates a non-selective cation conductance and a Ca2+ entry pathway in TRPC3-overexpressing cells but not in wild-type HEK-293 (human embryonic kidney 293) cells. The cholesterol-induced membrane conductance exhibited a current-to-voltage relationship similar to that observed upon PLC (phospholipase C)-dependent activation of TRPC3 channels. Nonetheless, the cholesterol-activated conductance lacked negative modulation by extracellular Ca2+, a typical feature of agonist-activated TRPC3 currents. Involvement of TRPC3 in the cholesterol-dependent membrane conductance was further corroborated by a novel dominant-negative strategy for selective blockade of TRPC3 channel activity. Expression of a TRPC3 mutant, which contained a haemagglutinin epitope tag in the second extracellular loop, conferred antibody sensitivity to both the classical PLC-activated as well as the cholesterol-activated conductance in TRPC3-expressing cells. Moreover, cholesterol loading as well as PLC stimulation was found to increase surface expression of TRPC3. Promotion of TRPC3 membrane expression by cholesterol was persistent over 30 min, while PLC-mediated enhancement of plasma membrane expression of TRPC3 was transient in nature. We suggest the cholesterol content of the plasma membrane as a determinant of cellular TRPC3 activity and provide evidence for cholesterol dependence of TRPC3 surface expression.


Assuntos
Colesterol/fisiologia , Transporte de Íons/efeitos dos fármacos , Lipídeos de Membrana/fisiologia , Canais de Cátion TRPC/fisiologia , Sinalização do Cálcio , Carbacol/farmacologia , Cátions/metabolismo , Linhagem Celular , Epitopos/genética , Marcação de Genes , Genes Dominantes , Genes Reporter , Humanos , Rim/citologia , Mutagênese Insercional , Técnicas de Patch-Clamp , Proteínas Recombinantes de Fusão/fisiologia , Canais de Cátion TRPC/genética , Transfecção , beta-Ciclodextrinas/farmacologia
10.
Naunyn Schmiedebergs Arch Pharmacol ; 371(4): 251-6, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15889239

RESUMO

Transient receptor potential (TRP) proteins have been recognized as sensors for a wide variety of external and internal signals involved in maintenance of cellular homeostasis and control of physiological functions. Evidence of a striking versatility in terms of signal integration and transduction has been reported for members of the canonical (or classical) TRP subfamily (TRPCs). TRPC species are cation channel subunits and emerge as multifunctional signal transduction molecules that are able to function as components of divergent signalplexes. Results obtained in heterologous expression systems suggest TRPC3 as a paradigm of multifunctional signal transduction by a cation channel protein. TRPC3 serves cellular Ca(2+) signaling by multiple mechanisms and may control a variety of distinct physiological functions. In this review, we summarize current knowledge on the properties and possible signaling partners of TRPC3, and discuss the role of TRPC3 channel proteins in cellular signaling networks.


Assuntos
Sinalização do Cálcio/fisiologia , Ativação do Canal Iônico/fisiologia , Canais de Cátion TRPC/fisiologia , Animais , Linhagem Celular , Humanos , Canais de Cátion TRPC/metabolismo
11.
Novartis Found Symp ; 258: 222-30; discussion 231-5, 263-6, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15104185

RESUMO

Increasing evidence suggests a pivotal role of reactive oxygen species (ROS) as well as reactive nitrogen species (RNS) in human pathophysiology. A typical target of ROS/RNS signalling is Ca2+ channels which mediate both long-term as well as acute cellular responses to oxidative stress. We have previously reported that cation channels related to the Drosophila transient receptor potential gene product (TRPC proteins) are likely to serve as redox sensors in the vascular endothelium, and demonstrated that TRPC3 expression is a determinant of the nitric oxide sensitivity of store-operated Ca2+ signalling. Experiments with TRPC species overexpressed in HEK293 cells confirmed that TRPC3 and TRPC4 are able to form redox sensitive cation channels. A key mechanism involved in redox activation of TRPC3 appears to be ROS-induced promotion of protein tyrosine phosphorylation and stimulation of phospholipase C activity. In addition, oxidative stress-induced disruption of caveolin 1-rich lipid raft domains, which interfere with functional TRPC channels, is likely to contribute to redox modulation of TRP proteins and to oxidative stress-induced changes in cellular Ca2+ signalling. Taken together, our data suggest TRPC species serve as a link between cellular redox state and Ca2+ homeostasis. Thus, modulation of these cellular redox sensors may offer unique opportunities for therapeutic interventions.


Assuntos
Sinalização do Cálcio/fisiologia , Cálcio/fisiologia , Canais Iônicos/metabolismo , Estresse Oxidativo , Caveolina 1 , Caveolinas/metabolismo , Células Cultivadas , Colesterol/metabolismo , Células Endoteliais/metabolismo , Humanos , Canais Iônicos/química , Canais Iônicos/genética , Oxirredução , Peróxidos/farmacologia , Sódio/metabolismo , Canais de Cátion TRPC , Fosfolipases Tipo C/farmacologia
12.
J Biol Chem ; 279(14): 13696-704, 2004 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-14736881

RESUMO

TRPC3 has been suggested as a key component of phospholipase C-dependent Ca(2+) signaling. Here we investigated the role of TRPC3-mediated Na(+) entry as a determinant of plasmalemmal Na(+)/Ca(2+) exchange. Ca(2+) signals generated by TRPC3 overexpression in HEK293 cells were found to be dependent on extracellular Na(+), in that carbachol-stimulated Ca(2+) entry into TRPC3 expressing cells was significantly suppressed when extracellular Na(+) was reduced to 5 mm. Moreover, KB-R9743 (5 microm) an inhibitor of the Na(+)/Ca(2+) exchanger (NCX) strongly suppressed TRPC3-mediated Ca(2+) entry but not TRPC3-mediated Na(+) currents. NCX1 immunoreactivity was detectable in HEK293 as well as in TRPC3-overexpressing HEK293 cells, and reduction of extracellular Na(+) after Na(+) loading with monensin resulted in significant rises in intracellular free Ca(2+) (Ca(2+)(i)) of HEK293 cells. Similar rises in Ca(2+)(i) were recorded in TRPC3-overexpressing cells upon the reduction of extracellular Na(+) subsequent to stimulation with carbachol. These increases in Ca(2+)(i) were associated with outward membrane currents at positive potentials and inhibited by KB-R7943 (5 microm), chelation of extracellular Ca(2+), or dominant negative suppression of TRPC3 channel function. This suggests that Ca(2+) entry into TRPC3-expressing cells involves reversed mode Na(+)/Ca(2+) exchange. Cell fractionation experiments demonstrated co-localization of TRPC3 and NCX1 in low density membrane fractions, and co-immunoprecipitation experiments provided evidence for association of TRPC3 and NCX1. Glutathione S-transferase pull-down experiments revealed that NCX1 interacts with the cytosolic C terminus of TRPC3. We suggest functional and physical interaction of nonselective TRPC cation channels with NCX proteins as a novel principle of TRPC-mediated Ca(2+) signaling.


Assuntos
Sinalização do Cálcio/fisiologia , Canais Iônicos/metabolismo , Trocador de Sódio e Cálcio/metabolismo , Sódio/metabolismo , Tioureia/análogos & derivados , Antiarrítmicos/farmacologia , Cálcio/metabolismo , Linhagem Celular , Membrana Celular/metabolismo , Homeostase , Humanos , Rim/citologia , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Trocador de Sódio e Cálcio/antagonistas & inibidores , Canais de Cátion TRPC , Tioureia/farmacologia
13.
Circ Res ; 92(8): 888-96, 2003 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-12663491

RESUMO

This study was designed to investigate the role of voltage-independent and voltage-dependent Ca2+ channels in the Ca2+ signaling associated with intracellular alkalinization in A7r5 vascular smooth muscle cells. Extracellular administration of ammonium chloride (20 mmol/L) resulted in elevation of intracellular pH and activation of a sustained Ca2+ entry that was inhibited by 2-amino-ethoxydiphenyl borate (2-APB, 200 micromol/L) but not by verapamil (10 micro;mol/L). Alkalosis-induced Ca2+ entry was mediated by a voltage-independent cation conductance that allowed permeation of Ca2+ (PCa/PNa approximately 6), and was associated with inhibition of L-type Ca2+ currents. Alkalosis-induced inhibition of L-type Ca2+ currents was dependent on the presence of extracellular Ca2+ and was prevented by expression of a dominant-negative mutant of calmodulin. In the absence of extracellular Ca2+, with Ba2+ or Na+ as charge carrier, intracellular alkalosis failed to inhibit but potentiated L-type Ca2+ channel currents. Inhibition of Ca2+ currents through voltage-independent cation channels by 2-APB prevented alkalosis-induced inhibition of L-type Ca2+ currents. Similarly, 2-APB prevented vasopressin-induced activation of nonselective cation channels and inhibition of L-type Ca2+ currents. We suggest the existence of a pH-controlled Ca2+ entry pathway that governs the activity of smooth muscle L-type Ca2+ channels due to control of Ca2+/calmodulin-dependent negative feedback regulation. This Ca2+ entry pathway exhibits striking similarity with the pathway activated by stimulation of phospholipase-C-coupled receptors, and may involve a similar type of cation channel. We demonstrate for the first time the tight functional coupling between these voltage-independent Ca2+ channels and classical voltage-gated L-type Ca2+ channels.


Assuntos
Canais de Cálcio Tipo L/fisiologia , Canais de Cálcio/fisiologia , Músculo Liso Vascular/fisiologia , Cloreto de Amônio/farmacologia , Animais , Arginina Vasopressina/farmacologia , Bário/farmacologia , Compostos de Boro/farmacologia , Cálcio/metabolismo , Cálcio/farmacologia , Calmodulina/genética , Calmodulina/metabolismo , Linhagem Celular , Humanos , Concentração de Íons de Hidrogênio , Potenciais da Membrana/efeitos dos fármacos , Músculo Liso Vascular/citologia , Músculo Liso Vascular/efeitos dos fármacos , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...