Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Eur J Pharmacol ; 944: 175562, 2023 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-36736940

RESUMO

Phosphodiesterases (PDE) type 3 and 4 promote vasoconstriction by hydrolysing cAMP. In experimental heart failure (HF), PDE3 makes PDE4 redundant in aorta, but it is not known if this occurs in resistance vessels, such as mesenteric artery. As PDE2 is increased in the failing myocardium, its possible role in the vasculature also needs to be addressed. Here, the function of PDE2, PDE3 and PDE4 in rat mesenteric arteries was characterized in experimental HF. Mesenteric arteries were isolated from rats sacrificed 22 weeks after surgical stenosis of the ascending aorta (HF), or Sham surgery. PDE inhibitors were used to probe isoenzyme contributions in enzymatic and isometric tension assays. PDE2 and PDE4 activities, but not PDE3 activity, facilitate contraction produced by the thromboxane analogue U46619 in Sham arteries, while in HF all three isoenzymes contribute to this response. NO synthase inhibition by L-NAME abolished the action of the PDE2 inhibitor. L-NAME eliminated the contribution of PDE4 in HF, but unmasked a contribution for PDE3 in Sham. PDE3 and PDE4 activities attenuated relaxant response to ß-adrenergic stimulation in Sham and HF. PDE2 did not participate in cAMP or cGMP-mediated relaxant responses. PDE3 and PDE4 cAMP-hydrolysing activities were smaller in HF mesenteric arteries, while PDE2 activity was scarce in both groups. Endothelial cells and arterial myocytes displayed PDE2 immunolabelling. We highlight that, by contrast with previous observations in aorta, PDE4 participates equally as PDE3 in contracting mesenteric artery in HF. PDE2 activity emerges as a promoter of contractile response that is preserved in HF.


Assuntos
Nucleotídeo Cíclico Fosfodiesterase do Tipo 4 , Insuficiência Cardíaca , Ratos , Animais , Ratos Wistar , Células Endoteliais , NG-Nitroarginina Metil Éster , Nucleotídeo Cíclico Fosfodiesterase do Tipo 3 , Artérias Mesentéricas , 3',5'-AMP Cíclico Fosfodiesterases
2.
Eur Respir J ; 58(5)2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-33926975

RESUMO

INTRODUCTION: A reduction in pulmonary artery relaxation is a key event in the pathogenesis of pulmonary arterial hypertension (PAH). Cystic fibrosis transmembrane conductance regulator (CFTR) dysfunction in airway epithelial cells plays a central role in cystic fibrosis; CFTR is also expressed in pulmonary arteries and has been shown to control endothelium-independent relaxation. AIM AND OBJECTIVES: We aimed to delineate the role of CFTR in PAH pathogenesis through observational and interventional experiments in human tissues and animal models. METHODS AND RESULTS: Reverse-transcriptase quantitative PCR, confocal imaging and electron microscopy showed that CFTR expression was reduced in pulmonary arteries from patients with idiopathic PAH (iPAH) and in rats with monocrotaline-induced pulmonary hypertension (PH). Moreover, using myography on human, pig and rat pulmonary arteries, we demonstrated that CFTR activation induces pulmonary artery relaxation. CFTR-mediated pulmonary artery relaxation was reduced in pulmonary arteries from iPAH patients and rats with monocrotaline- or chronic hypoxia-induced PH. Long-term in vivo CFTR inhibition in rats significantly increased right ventricular systolic pressure, which was related to exaggerated pulmonary vascular cell proliferation in situ and vessel neomuscularisation. Pathologic assessment of lungs from patients with severe cystic fibrosis (F508del-CFTR) revealed severe pulmonary artery remodelling with intimal fibrosis and medial hypertrophy. Lungs from homozygous F508delCftr rats exhibited pulmonary vessel neomuscularisation. The elevations in right ventricular systolic pressure and end diastolic pressure in monocrotaline-exposed rats with chronic CFTR inhibition were more prominent than those in vehicle-exposed rats. CONCLUSIONS: CFTR expression is strongly decreased in pulmonary artery smooth muscle and endothelial cells in human and animal models of PH. CFTR inhibition increases vascular cell proliferation and strongly reduces pulmonary artery relaxation.


Assuntos
Regulador de Condutância Transmembrana em Fibrose Cística , Hipertensão Arterial Pulmonar , Animais , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Células Endoteliais , Humanos , Monocrotalina , Hipertensão Arterial Pulmonar/genética , Hipertensão Arterial Pulmonar/patologia , Ratos , Suínos
3.
Pharmacol Ther ; 209: 107499, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32068004

RESUMO

Numerous mediators and drugs regulate blood flow or arterial pressure by acting on vascular tone, involving cyclic nucleotide intracellular pathways. These signals lead to regulation of several cellular effectors, including ion channels that tune cell membrane potential, Ca2+ influx and vascular tone. The characterization of these vasocontrictive or vasodilating mechanisms has grown in complexity due to i) the variety of ion channels that are expressed in both vascular endothelial and smooth muscle cells, ii) the heterogeneity of responses among the various vascular beds, and iii) the number of molecular mechanisms involved in cyclic nucleotide signalling in health and disease. This review synthesizes key data from literature that highlight ion channels as physiologically relevant effectors of cyclic nucleotide pathways in the vasculature, including the characterization of the molecular mechanisms involved. In smooth muscle cells, cation influx or chloride efflux through ion channels are associated with vasoconstriction, whereas K+ efflux repolarizes the cell membrane potential and mediates vasodilatation. Both categories of ion currents are under the influence of cAMP and cGMP pathways. Evidence that some ion channels are influenced by CN signalling in endothelial cells will also be presented. Emphasis will also be put on recent data touching a variety of determinants such as phosphodiesterases, EPAC and kinase anchoring, that complicate or even challenge former paradigms.


Assuntos
Artérias/fisiologia , Canais Iônicos/metabolismo , Músculo Liso Vascular/fisiologia , Nucleotídeos Cíclicos/metabolismo , Vasoconstrição/fisiologia , Vasodilatação/fisiologia , Animais , Endotélio Vascular/fisiologia , Humanos , Transdução de Sinais/fisiologia
4.
Circ Res ; 125(7): 678-695, 2019 09 13.
Artigo em Inglês | MEDLINE | ID: mdl-31347976

RESUMO

RATIONALE: Pulmonary arterial hypertension is a severe lethal cardiopulmonary disease. Loss of function mutations in KCNK3 (potassium channel subfamily K member 3) gene, which encodes an outward rectifier K+ channel, have been identified in pulmonary arterial hypertension patients. OBJECTIVE: We have demonstrated that KCNK3 dysfunction is common to heritable and nonheritable pulmonary arterial hypertension and to experimental pulmonary hypertension (PH). Finally, KCNK3 is not functional in mouse pulmonary vasculature. METHODS AND RESULTS: Using CRISPR/Cas9 technology, we generated a 94 bp out of frame deletion in exon 1 of Kcnk3 gene and characterized these rats at the electrophysiological, echocardiographic, hemodynamic, morphological, cellular, and molecular levels to decipher the cellular mechanisms associated with loss of KCNK3. Using patch-clamp technique, we validated our transgenic strategy by demonstrating the absence of KCNK3 current in freshly isolated pulmonary arterial smooth muscle cells from Kcnk3-mutated rats. At 4 months of age, echocardiographic parameters revealed shortening of the pulmonary artery acceleration time associated with elevation of the right ventricular systolic pressure. Kcnk3-mutated rats developed more severe PH than wild-type rats after monocrotaline exposure or chronic hypoxia exposure. Kcnk3-mutation induced a lung distal neomuscularization and perivascular extracellular matrix activation. Lungs of Kcnk3-mutated rats were characterized by overactivation of ERK1/2 (extracellular signal-regulated kinase1-/2), AKT (protein kinase B), SRC, and overexpression of HIF1-α (hypoxia-inducible factor-1 α), survivin, and VWF (Von Willebrand factor). Linked with plasma membrane depolarization, reduced endothelial-NOS expression and desensitization of endothelial-derived hyperpolarizing factor, Kcnk3-mutated rats presented predisposition to vasoconstriction of pulmonary arteries and a severe loss of sildenafil-induced pulmonary arteries relaxation. Moreover, we showed strong alteration of right ventricular cardiomyocyte excitability. Finally, Kcnk3-mutated rats developed age-dependent PH associated with low serum-albumin concentration. CONCLUSIONS: We established the first Kcnk3-mutated rat model of PH. Our results confirm that KCNK3 loss of function is a key event in pulmonary arterial hypertension pathogenesis. This model presents new opportunities for understanding the initiating mechanisms of PH and testing biologically relevant therapeutic molecules in the context of PH.


Assuntos
Modelos Animais de Doenças , Hipertensão Pulmonar/genética , Mutação com Perda de Função , Proteínas do Tecido Nervoso/genética , Canais de Potássio de Domínios Poros em Tandem/genética , Potenciais de Ação , Animais , Pressão Sanguínea , Feminino , Hipertensão Pulmonar/metabolismo , Hipertensão Pulmonar/patologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Pulmão/metabolismo , Pulmão/patologia , Masculino , Proteína Quinase 1 Ativada por Mitógeno/genética , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/fisiologia , Proteínas do Tecido Nervoso/metabolismo , Óxido Nítrico Sintase Tipo III/genética , Óxido Nítrico Sintase Tipo III/metabolismo , Canais de Potássio de Domínios Poros em Tandem/metabolismo , Ratos , Ratos Sprague-Dawley , Survivina/genética , Survivina/metabolismo , Vasoconstrição , Fator de von Willebrand/genética , Fator de von Willebrand/metabolismo
5.
Br J Pharmacol ; 176(11): 1780-1792, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30825186

RESUMO

BACKGROUND AND PURPOSE: Up-regulation of phosphodiesterases (PDEs) is associated with several vascular diseases, and better understanding of the roles of each PDE isoform in controlling subcellular pools of cyclic nucleotides in vascular cells is needed. We investigated the respective role of PDE1, PDE5, and PDE9 in controlling intracellular cAMP and/or cGMP concentrations ([cAMP]i , [cGMP]i ) in cultured rat aortic smooth muscle cells (RASMCs). EXPERIMENTAL APPROACH: We used selective inhibitors of PDE1 (PF-04471141), PDE5 (sildenafil), and PDE9 (PF-04447943) to measure cAMP- and cGMP-PDE activities with a radioenzymatic assay, in RASMC extracts. Real-time [cAMP]i and [cGMP]i were recorded by Förster resonance energy transfer-imaging in single living cells, and cell proliferation was assessed in FBS-stimulated cells. KEY RESULTS: PDE1, PDE5, and PDE9 represented the major cGMP-hydrolyzing activity in RASMCs. Basal PDE1 exerted a functional role in degrading in situ the cGMP produced in response to activation of particulate GC by C-type natriuretic peptide. In high intracellular Ca2+ concentrations, PDE1 also regulated the NO/soluble GC-dependent cGMP response, as well as the ß-adrenoceptor-mediated cAMP response. PDE5 exerted a major role in degrading cGMP produced by NO and the natriuretic peptides. PDE9 only regulated the NO-induced [cGMP]i increase. All three PDEs contributed differently to regulate cell proliferation under basal conditions and upon cGMP-elevating stimuli. CONCLUSIONS AND IMPLICATIONS: Our data emphasize the distinct roles of PDE1, PDE5, and PDE9 in local regulation of [cAMP]i and [cGMP]i , in vascular smooth muscle cells, strengthening the concept of PDEs as key actors in the subcellular compartmentation of cyclic nucleotides.


Assuntos
Miócitos de Músculo Liso/metabolismo , Nucleotídeos Cíclicos/metabolismo , Diester Fosfórico Hidrolases/metabolismo , Animais , Células Cultivadas , AMP Cíclico/metabolismo , GMP Cíclico/metabolismo , Masculino , Miócitos de Músculo Liso/efeitos dos fármacos , Inibidores de Fosfodiesterase/farmacologia , Ratos Wistar , Transdução de Sinais/efeitos dos fármacos , Citrato de Sildenafila/farmacologia
6.
Cardiovasc Res ; 115(1): 130-144, 2019 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-29939224

RESUMO

Aims: Regulation of vascular tone by 3',5'-cyclic adenosine monophosphate (cAMP) involves many effectors including the large conductance, Ca2+-activated, K+ (BKCa) channels. In arteries, cAMP is mainly hydrolyzed by type 3 and 4 phosphodiesterases (PDE3, PDE4). Here, we examined the specific contribution of BKCa channels to tone regulation by these PDEs in rat coronary arteries, and how this is altered in heart failure (HF). Methods and results: Concomitant application of PDE3 (cilostamide) and PDE4 (Ro-20-1724) inhibitors increased BKCa unitary channel activity in isolated myocytes from rat coronary arteries. Myography was conducted in isolated, U46619-contracted coronary arteries. Cilostamide (Cil) or Ro-20-1724 induced a vasorelaxation that was greatly reduced by iberiotoxin (IBTX), a BKCa channel blocker. Ro-20-1724 and Cil potentiated the relaxation induced by the ß-adrenergic agonist isoprenaline (ISO) or the adenylyl cyclase activator L-858051 (L85). IBTX abolished the effect of PDE inhibitors on ISO but did not on L85. In coronary arteries from rats with HF induced by aortic stenosis, contractility and response to acetylcholine were dramatically reduced compared with arteries from sham rats, but relaxation to PDE inhibitors was retained. Interestingly, however, IBTX had no effect on Ro-20-1724- and Cil-induced vasorelaxations in HF. Expression of the BKCa channel α-subunit, of a 98 kDa PDE3A and of a 80 kDa PDE4D were lower in HF compared with sham coronary arteries, while that of a 70 kDa PDE4B was increased. Proximity ligation assays demonstrated that PDE3 and PDE4 were localized in the vicinity of the channel. Conclusion: BKCa channels mediate the relaxation of coronary artery induced by PDE3 and PDE4 inhibition. This is achieved by co-localization of both PDEs with BKCa channels, enabling tight control of cAMP available for channel opening. Contribution of the channel is prominent at rest and on ß-adrenergic stimulation. This coupling is lost in HF.


Assuntos
Vasos Coronários/enzimologia , Nucleotídeo Cíclico Fosfodiesterase do Tipo 3/metabolismo , Nucleotídeo Cíclico Fosfodiesterase do Tipo 4/metabolismo , Insuficiência Cardíaca/enzimologia , Canais de Potássio Ativados por Cálcio de Condutância Alta/metabolismo , Vasodilatação , Animais , Vasos Coronários/efeitos dos fármacos , Vasos Coronários/fisiopatologia , Modelos Animais de Doenças , Insuficiência Cardíaca/fisiopatologia , Ativação do Canal Iônico , Masculino , Inibidores da Fosfodiesterase 3/farmacologia , Inibidores da Fosfodiesterase 4/farmacologia , Ratos Wistar , Transdução de Sinais , Vasodilatação/efeitos dos fármacos , Vasodilatadores/farmacologia
7.
Circulation ; 139(7): 932-948, 2019 02 12.
Artigo em Inglês | MEDLINE | ID: mdl-30586714

RESUMO

BACKGROUND: Monoallelic mutations in the gene encoding bone morphogenetic protein receptor 2 ( Bmpr2) are the main genetic risk factor for heritable pulmonary arterial hypertension (PAH) with incomplete penetrance. Several Bmpr2 transgenic mice have been reported to develop mild spontaneous PAH. In this study, we examined whether rats with the Bmpr2 mutation were susceptible to developing more severe PAH. METHODS: The zinc finger nuclease method was used to establish rat lines with mutations in the Bmpr2 gene. These rats were then characterized at the hemodynamic, histological, electrophysiological, and molecular levels. RESULTS: Rats with a monoallelic deletion of 71 bp in exon 1 (Δ 71 rats) showed decreased BMPRII expression and phosphorylated SMAD1/5/9 levels. Δ 71 Rats develop age-dependent spontaneous PAH with a low penetrance (16%-27%), similar to that in humans. Δ 71 Rats were more susceptible to hypoxia-induced pulmonary hypertension than wild-type rats. Δ 71 Rats exhibited progressive pulmonary vascular remodeling associated with a proproliferative phenotype and showed lower pulmonary microvascular density than wild-type rats. Organ bath studies revealed severe alteration of pulmonary artery contraction and relaxation associated with potassium channel subfamily K member 3 (KCNK3) dysfunction. High levels of perivascular fibrillar collagen and pulmonary interleukin-6 overexpression discriminated rats that developed spontaneous PAH and rats that did not develop spontaneous PAH. Finally, detailed assessments of cardiomyocytes demonstrated alterations in morphology, calcium (Ca2+), and cell contractility specific to the right ventricle; these changes could explain the lower cardiac output of Δ 71 rats. Indeed, adult right ventricular cardiomyocytes from Δ 71 rats exhibited a smaller diameter, decreased sensitivity of sarcomeres to Ca2+, decreased [Ca2+] transient amplitude, reduced sarcoplasmic reticulum Ca2+ content, and short action potential duration compared with right ventricular cardiomyocytes from wild-type rats. CONCLUSIONS: We characterized the first Bmpr2 mutant rats and showed some of the critical cellular and molecular dysfunctions described in human PAH. We also identified the heart as an unexpected but potential target organ of Bmpr2 mutations. Thus, this new genetic rat model represents a promising tool to study the pathogenesis of PAH.


Assuntos
Pressão Arterial/genética , Receptores de Proteínas Morfogenéticas Ósseas Tipo II/genética , Hipertensão Pulmonar/genética , Hipertensão Pulmonar/fisiopatologia , Mutação , Contração Miocárdica/genética , Artéria Pulmonar/fisiopatologia , Função Ventricular Direita/genética , Potenciais de Ação , Animais , Receptores de Proteínas Morfogenéticas Ósseas Tipo II/metabolismo , Sinalização do Cálcio , Modelos Animais de Doenças , Predisposição Genética para Doença , Hipertensão Pulmonar/metabolismo , Hipóxia/complicações , Miócitos Cardíacos/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Fenótipo , Fosforilação , Canais de Potássio de Domínios Poros em Tandem/metabolismo , Artéria Pulmonar/metabolismo , Ratos Mutantes , Proteínas Smad/metabolismo
8.
Circulation ; 137(21): 2256-2273, 2018 05 22.
Artigo em Inglês | MEDLINE | ID: mdl-29217642

RESUMO

BACKGROUND: Myocardial metabolic impairment is a major feature in chronic heart failure. As the major coenzyme in fuel oxidation and oxidative phosphorylation and a substrate for enzymes signaling energy stress and oxidative stress response, nicotinamide adenine dinucleotide (NAD+) is emerging as a metabolic target in a number of diseases including heart failure. Little is known on the mechanisms regulating homeostasis of NAD+ in the failing heart. METHODS: To explore possible alterations of NAD+ homeostasis in the failing heart, we quantified the expression of NAD+ biosynthetic enzymes in the human failing heart and in the heart of a mouse model of dilated cardiomyopathy (DCM) triggered by Serum Response Factor transcription factor depletion in the heart (SRFHKO) or of cardiac hypertrophy triggered by transverse aorta constriction. We studied the impact of NAD+ precursor supplementation on cardiac function in both mouse models. RESULTS: We observed a 30% loss in levels of NAD+ in the murine failing heart of both DCM and transverse aorta constriction mice that was accompanied by a decrease in expression of the nicotinamide phosphoribosyltransferase enzyme that recycles the nicotinamide precursor, whereas the nicotinamide riboside kinase 2 (NMRK2) that phosphorylates the nicotinamide riboside precursor is increased, to a higher level in the DCM (40-fold) than in transverse aorta constriction (4-fold). This shift was also observed in human failing heart biopsies in comparison with nonfailing controls. We show that the Nmrk2 gene is an AMP-activated protein kinase and peroxisome proliferator-activated receptor α responsive gene that is activated by energy stress and NAD+ depletion in isolated rat cardiomyocytes. Nicotinamide riboside efficiently rescues NAD+ synthesis in response to FK866-mediated inhibition of nicotinamide phosphoribosyltransferase and stimulates glycolysis in cardiomyocytes. Accordingly, we show that nicotinamide riboside supplementation in food attenuates the development of heart failure in mice, more robustly in DCM, and partially after transverse aorta constriction, by stabilizing myocardial NAD+ levels in the failing heart. Nicotinamide riboside treatment also robustly increases the myocardial levels of 3 metabolites, nicotinic acid adenine dinucleotide, methylnicotinamide, and N1-methyl-4-pyridone-5-carboxamide, that can be used as validation biomarkers for the treatment. CONCLUSIONS: The data show that nicotinamide riboside, the most energy-efficient among NAD precursors, could be useful for treatment of heart failure, notably in the context of DCM, a disease with few therapeutic options.


Assuntos
Cardiomiopatia Dilatada/tratamento farmacológico , Niacinamida/análogos & derivados , Proteínas Quinases Ativadas por AMP/metabolismo , Acrilamidas/uso terapêutico , Animais , Ácido Cítrico/metabolismo , Citocinas/genética , Citocinas/metabolismo , Suplementos Nutricionais , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Insuficiência Cardíaca/prevenção & controle , Metaboloma/efeitos dos fármacos , Camundongos , Camundongos Transgênicos , Miócitos Cardíacos/citologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , NAD/metabolismo , Niacinamida/uso terapêutico , Nicotinamida Fosforribosiltransferase/genética , Nicotinamida Fosforribosiltransferase/metabolismo , PPAR alfa/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/antagonistas & inibidores , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Piperidinas/uso terapêutico , Compostos de Piridínio , Ratos , Fator de Resposta Sérica/deficiência , Fator de Resposta Sérica/genética
9.
Eur J Med Chem ; 143: 473-490, 2018 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-29202409

RESUMO

A novel series of tubulin polymerization inhibitors, based on fluorinated derivatives of isocombretastatin A-4 was synthesized with the goal of evaluating the effect of these compounds on the proliferative activity. The introduction of fluorine atom was performed on the phenyl ring or at the linker between the two aromatic rings. The modification of isoCA-4 by introduction of difluoromethoxy group at the para-position (3i) and substitution of the two protons of the linker by two fluorine atoms (3m), produced the most active compounds in the series, with IC50 values of 0.15-2.2 nM (3i) and 0.1-2 nM (3m) respectively, against a panel of six cancer cell lines. Compounds 3i and 3m had greater antiproliferative activity in comparison with references CA-4 or isoCA-4, the presence of fluorine group leads to a significant enhancement of the antiproliferative activity. Molecular docking studies indicated that compounds 3i and 3m occupy the colchicine binding site of tubulin. Evaluation of cytotoxicity in Human noncancer cells indicated that the compounds 3i and 3m were practically ineffective in quiescent peripheral blood lymphocytes, and may have a selective antiproliferative activity against cancer cells. Analyses of cell cycle distribution, and morphological microtubules organization showed that compound 3m induced G2/M phase arrest and, dramatically disrupted the microtubule network.


Assuntos
Antineoplásicos/farmacologia , Desenho de Fármacos , Flúor/química , Estilbenos/farmacologia , Tubulina (Proteína)/metabolismo , Antineoplásicos/síntese química , Antineoplásicos/química , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Modelos Moleculares , Estrutura Molecular , Polimerização/efeitos dos fármacos , Estilbenos/síntese química , Estilbenos/química , Relação Estrutura-Atividade
10.
Circulation ; 133(14): 1371-85, 2016 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-26912814

RESUMO

BACKGROUND: Mutations in the KCNK3 gene have been identified in some patients suffering from heritable pulmonary arterial hypertension (PAH). KCNK3 encodes an outward rectifier K(+) channel, and each identified mutation leads to a loss of function. However, the pathophysiological role of potassium channel subfamily K member 3 (KCNK3) in PAH is unclear. We hypothesized that loss of function of KCNK3 is a hallmark of idiopathic and heritable PAH and contributes to dysfunction of pulmonary artery smooth muscle cells and pulmonary artery endothelial cells, leading to pulmonary artery remodeling: consequently, restoring KCNK3 function could alleviate experimental pulmonary hypertension (PH). METHODS AND RESULTS: We demonstrated that KCNK3 expression and function were reduced in human PAH and in monocrotaline-induced PH in rats. Using a patch-clamp technique in freshly isolated (not cultured) pulmonary artery smooth muscle cells and pulmonary artery endothelial cells, we found that KCNK3 current decreased progressively during the development of monocrotaline-induced PH and correlated with plasma-membrane depolarization. We demonstrated that KCNK3 modulated pulmonary arterial tone. Long-term inhibition of KCNK3 in rats induced distal neomuscularization and early hemodynamic signs of PH, which were related to exaggerated proliferation of pulmonary artery endothelial cells, pulmonary artery smooth muscle cell, adventitial fibroblasts, and pulmonary and systemic inflammation. Lastly, in vivo pharmacological activation of KCNK3 significantly reversed monocrotaline-induced PH in rats. CONCLUSIONS: In PAH and experimental PH, KCNK3 expression and activity are strongly reduced in pulmonary artery smooth muscle cells and endothelial cells. KCNK3 inhibition promoted increased proliferation, vasoconstriction, and inflammation. In vivo pharmacological activation of KCNK3 alleviated monocrotaline-induced PH, thus demonstrating that loss of KCNK3 is a key event in PAH pathogenesis and thus could be therapeutically targeted.


Assuntos
Hipertensão Pulmonar/fisiopatologia , Proteínas do Tecido Nervoso/fisiologia , Canais de Potássio de Domínios Poros em Tandem/fisiologia , Túnica Adventícia/patologia , Animais , Receptores de Proteínas Morfogenéticas Ósseas Tipo II/genética , Divisão Celular , Endotélio Vascular/patologia , Fibroblastos/patologia , Predisposição Genética para Doença , Hemodinâmica , Humanos , Hipertensão Pulmonar/induzido quimicamente , Hipertensão Pulmonar/complicações , Hipertensão Pulmonar/genética , Hipertrofia Ventricular Direita/etiologia , Inflamação , Masculino , Potenciais da Membrana , Monocrotalina/toxicidade , Mutação , Miócitos de Músculo Liso/patologia , Proteínas do Tecido Nervoso/antagonistas & inibidores , Proteínas do Tecido Nervoso/biossíntese , Proteínas do Tecido Nervoso/genética , Técnicas de Patch-Clamp , Canais de Potássio de Domínios Poros em Tandem/antagonistas & inibidores , Canais de Potássio de Domínios Poros em Tandem/biossíntese , Canais de Potássio de Domínios Poros em Tandem/genética , Ratos , Ratos Sprague-Dawley , Ratos Wistar , Sulfonamidas/farmacologia , Resistência Vascular , ortoaminobenzoatos/farmacologia
11.
Circulation ; 129(4): 451-62, 2014 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-24190960

RESUMO

BACKGROUND: ß1-2-adrenergic receptors (AR) are key regulators of cardiac contractility and remodeling in response to catecholamines. ß3-AR expression is enhanced in diseased human myocardium, but its impact on remodeling is unknown. METHODS AND RESULTS: Mice with cardiac myocyte-specific expression of human ß3-AR (ß3-TG) and wild-type (WT) littermates were used to compare myocardial remodeling in response to isoproterenol (Iso) or Angiotensin II (Ang II). ß3-TG and WT had similar morphometric and hemodynamic parameters at baseline. ß3-AR colocalized with caveolin-3, endothelial nitric oxide synthase (NOS) and neuronal NOS in adult transgenic myocytes, which constitutively produced more cyclic GMP, detected with a new transgenic FRET sensor. Iso and Ang II produced hypertrophy and fibrosis in WT mice, but not in ß3-TG mice, which also had less re-expression of fetal genes and transforming growth factor ß1. Protection from Iso-induced hypertrophy was reversed by nonspecific NOS inhibition at low dose Iso, and by preferential neuronal NOS inhibition at high-dose Iso. Adenoviral overexpression of ß3-AR in isolated cardiac myocytes also increased NO production and attenuated hypertrophy to Iso and phenylephrine. Hypertrophy was restored on NOS or protein kinase G inhibition. Mechanistically, ß3-AR overexpression inhibited phenylephrine-induced nuclear factor of activated T-cell activation. CONCLUSIONS: Cardiac-specific overexpression of ß3-AR does not affect cardiac morphology at baseline but inhibits the hypertrophic response to neurohormonal stimulation in vivo and in vitro, through a NOS-mediated mechanism. Activation of the cardiac ß3-AR pathway may provide future therapeutic avenues for the modulation of hypertrophic remodeling.


Assuntos
Ventrículos do Coração/patologia , Miócitos Cardíacos/metabolismo , Neurotransmissores/farmacologia , Óxido Nítrico Sintase/fisiologia , Receptores Adrenérgicos beta 3/metabolismo , Remodelação Ventricular/efeitos dos fármacos , Angiotensina II/efeitos adversos , Angiotensina II/farmacologia , Animais , Células Cultivadas , GMP Cíclico/fisiologia , Proteínas Quinases Dependentes de GMP Cíclico/fisiologia , Modelos Animais de Doenças , Ventrículos do Coração/fisiopatologia , Humanos , Hipertrofia/induzido quimicamente , Hipertrofia/patologia , Hipertrofia/fisiopatologia , Técnicas In Vitro , Isoproterenol/efeitos adversos , Isoproterenol/farmacologia , Masculino , Camundongos , Camundongos Transgênicos , Miócitos Cardíacos/patologia , Miócitos Cardíacos/fisiologia , Neurotransmissores/efeitos adversos , Receptores Adrenérgicos beta 3/genética , Transdução de Sinais/fisiologia , Remodelação Ventricular/fisiologia
12.
Cardiovasc Res ; 94(2): 304-15, 2012 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-22227153

RESUMO

The three isoforms of nitric oxide synthase (NOS), spatially confined in specific intracellular compartments in cardiac cells, have distinct roles in the regulation of contractility in pathophysiological situations. Recently, evidence has emerged that implicates NOS in modulating myocardial remodelling during cardiac stress, including after ischaemic insults. As long as they remain in a coupled state the NOS mostly attenuate hypertrophic remodelling through both cGMP-dependent and independent mechanisms. We review the evidence provided from the phenotype of genetic mouse models as well as from in vitro cell experiments dissecting the signalling effectors involved in the NOS-mediated regulation that justify new therapeutic interventions on the NOS-cGMP axis to attenuate the development of heart failure.


Assuntos
Insuficiência Cardíaca/metabolismo , Miocárdio/metabolismo , Óxido Nítrico Sintase/fisiologia , Óxido Nítrico/metabolismo , Remodelação Ventricular/fisiologia , Animais , Insuficiência Cardíaca/fisiopatologia , Humanos , Camundongos , Miocárdio/patologia , Transdução de Sinais
13.
Curr Heart Fail Rep ; 8(3): 184-92, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21633786

RESUMO

Catecholamines play a key role in the regulation of cardiovascular function, classically through ß(1/2)-adrenoreceptors (AR) activation. After ß(3)-AR cloning in the late 1980s, convincing evidence for ß(3)-AR expression and function in cardiovascular tissues recently initiated a reexamination of their involvement in the pathophysiology of cardiovascular diseases. Their upregulation in diseased cardiovascular tissues and resistance to desensitization suggest they may be attractive therapeutic targets. They may substitute for inoperant ß(1/2)-AR to mediate vasodilation in diabetic or atherosclerotic vessels. In cardiac ventricle, their contractile effects are functionally antipathetic to those of ß(1/2)-AR; in normal heart, ß(3)-ARs may mediate a moderate negative inotropic effect, but in heart failure, it may protect against adverse effects of excessive catecholamine stimulation by action on excitation-contraction coupling, electrophysiology, or remodelling. Thus, prospective studies in animals and patients at different stages of heart failure should lead to identify the best therapeutic window to use ß(3)-AR agonists and/or antagonists.


Assuntos
Agonistas de Receptores Adrenérgicos beta 3/farmacologia , Antagonistas Adrenérgicos beta/farmacologia , Sistema Cardiovascular/efeitos dos fármacos , Insuficiência Cardíaca/tratamento farmacológico , Receptores Adrenérgicos beta 3/fisiologia , Envelhecimento/fisiologia , Animais , Sistema Cardiovascular/metabolismo , Sistema Cardiovascular/fisiopatologia , Diabetes Mellitus/tratamento farmacológico , Diabetes Mellitus/metabolismo , Diabetes Mellitus/fisiopatologia , Expressão Gênica/efeitos dos fármacos , Expressão Gênica/fisiologia , Insuficiência Cardíaca/metabolismo , Insuficiência Cardíaca/fisiopatologia , Humanos , Hipertensão/tratamento farmacológico , Hipertensão/metabolismo , Hipertensão/fisiopatologia , Hipotireoidismo/tratamento farmacológico , Hipotireoidismo/metabolismo , Hipotireoidismo/fisiopatologia , Cirrose Hepática/tratamento farmacológico , Cirrose Hepática/metabolismo , Cirrose Hepática/fisiopatologia , Modelos Animais , Contração Miocárdica/efeitos dos fármacos , Contração Miocárdica/fisiologia , Infarto do Miocárdio/tratamento farmacológico , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/fisiopatologia , Sepse/tratamento farmacológico , Sepse/metabolismo , Sepse/fisiopatologia , Remodelação Ventricular
14.
J Physiol ; 589(Pt 13): 3231-46, 2011 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-21486782

RESUMO

The acid-sensitive, two-pore domain K+ channel, TASK-1, contributes to the background K+ conductance and membrane potential (Em) of rat and human pulmonary artery smooth muscle cells (PASMCs), but its role in regulating tone remains elusive. This study aimed to clarify the role of TASK-1 by determining the functional properties of pulmonary artery (PA) from mice in which the TASK-1 gene was deleted (TASK-1/3 KO), in comparison with wild-type (WT) C57BL/6 controls. Small vessel wire myography was used to measure isometric tension developed by intact PA. Em and currents were recorded from freshly isolated PASMCs using the perforated patch-clamp technique. Reverse transcription-polymerase chain reaction (RT-PCR) was used to estimate K+ channel expression. We could find no difference between PA from WT and TASK-1/3 KO mice. They showed similar constrictor responses to a range of agonists and K+ concentrations, the K+ channel blockers 4-aminopyridine, tetraethylammonium ions and XE991. Treprostinil, proposed to dilate by activating TASK-1, was just as effective in TASK-1/3 KO arteries. Blocking Ca2+ influx with nifedipine (1 µM) or levcromakalim (10 µM) had no effect on resting tone in either strain. The resting Em of PASMCs and its responses to K+ channel blockers were unchanged in TASK-1/3 KO mice as were voltage-activated K+ currents, including the non-inactivating K+ current (IKN) measured at 0 mV. The Em was, however, depolarised in comparison with other species.Mouse IKN was much smaller than in rat and showed no sensitivity to pH. The results imply that TASK-1 does not form a functional channel in mouse PASMCs.


Assuntos
Contração Muscular/fisiologia , Músculo Liso Vascular/fisiologia , Proteínas do Tecido Nervoso/deficiência , Proteínas do Tecido Nervoso/fisiologia , Canais de Potássio de Domínios Poros em Tandem/deficiência , Canais de Potássio de Domínios Poros em Tandem/fisiologia , Artéria Pulmonar/fisiologia , Vasoconstrição/fisiologia , Animais , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Contração Muscular/genética , Músculo Liso Vascular/citologia , Proteínas do Tecido Nervoso/genética , Técnicas de Cultura de Órgãos , Técnicas de Patch-Clamp/métodos , Canais de Potássio de Domínios Poros em Tandem/genética , Artéria Pulmonar/metabolismo , Ratos , Ratos Sprague-Dawley , Vasoconstrição/genética
15.
J Physiol ; 588(Pt 13): 2305-14, 2010 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-20421283

RESUMO

Calcium-activated chloride channels (CaCCs) play important roles in several physiological processes. In vascular smooth muscle, activation of these ion channels by agonist-induced Ca(2+) release results in membrane depolarization and vasoconstriction. The molecular identity of vascular CaCCs is not fully defined. Here we present evidence that TMEM16A (or anoctamin 1), a member of the transmembrane 16 (TMEM16) protein family, forms CaCCs in pulmonary artery smooth muscle cells (PASMCs). Patch-clamp analysis in freshly isolated PASMCs revealed strongly outward-rectifying, slowly activating Ca(2+)-activated Cl(-) currents sharing a high degree of similarity with heterologous TMEM16A currents. TMEM16A mRNA was identified in rat and human pulmonary arteries and various other vascular smooth muscle cell types. Further analyses revealed that several TMEM16A splice variants were detected in rat PASMCs and that TMEM16F and TMEM16K were also expressed in these cells, while TMEM16B, TMEM16D and TMEM16E were all at least 50 times less abundantly expressed and the remaining TMEM16 family members were absent. Downregulation of TMEM16A gene expression in primary cultures of rat PASMCs, with small interfering RNAs, was accompanied by almost total loss of whole-cell CaCC currents. Based on these results, we propose that TMEM16A is the major constituent of the vascular calcium-activated chloride channel in rat pulmonary artery smooth muscle.


Assuntos
Cálcio/fisiologia , Canais de Cloreto/fisiologia , Miócitos de Músculo Liso/fisiologia , Artéria Pulmonar/fisiologia , Algoritmos , Animais , Anoctamina-1 , Células Cultivadas , Canais de Cloreto/genética , Eletrofisiologia , Humanos , Masculino , Artéria Pulmonar/citologia , RNA Interferente Pequeno/genética , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Soluções
16.
Adv Exp Med Biol ; 661: 405-17, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20204745

RESUMO

Smooth muscle cells regulate the diameter of pulmonary arteries and the resistance to blood flow in the pulmonary circulation. These cells are normally relaxed to maintain low intrinsic vessel tone, but are contracted in pulmonary arterial hypertension (PAH). Potassium channels in the smooth muscle cell help to maintain low tone by polarising the membrane and preventing Ca(2+) influx through voltage-operated Ca(2+) channels. There is a loss of K(+) channel activity in PAH, so drugs that open K(+) channels are predicted to have a beneficial effect, provided their action can be restricted to the pulmonary circulation. Here we review the myriad of K(+) channels that are expressed in pulmonary arteries and suggest the roles that each might play in regulating pulmonary artery tone. We conclude that members of the KCNQ family of K(+) channels, the most recent K(+) channels to be discovered in pulmonary artery, may be a useful therapeutic target for the treatment of PAH. KCNQ channels appear to be preferentially expressed in pulmonary arteries and drugs that modulate their activity have potent effects on pulmonary artery tone.


Assuntos
Canais de Potássio KCNQ/metabolismo , Isoformas de Proteínas/metabolismo , Artéria Pulmonar/efeitos dos fármacos , Artéria Pulmonar/metabolismo , Vasodilatadores/farmacologia , Animais , Ensaios Clínicos como Assunto , Humanos , Hipertensão Pulmonar/tratamento farmacológico , Canais de Potássio KCNQ/química , Canais de Potássio KCNQ/genética , Potenciais da Membrana/fisiologia , Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Artéria Pulmonar/citologia , Vasodilatadores/uso terapêutico
17.
Br J Pharmacol ; 158(3): 848-61, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19694728

RESUMO

BACKGROUND AND PURPOSE: Blood vessel culture is gaining interest for use with transfection-based techniques, but alters the contractile properties of the vessels. The present study tested the effects of culture on the intrinsic tone of rat pulmonary arteries (PAs) and examined the function and expression of K(+) channels regulating the resting membrane potential (E(m)) and tone of pulmonary artery smooth muscle cells (PASMCs). EXPERIMENTAL APPROACH: Rat intrapulmonary arteries were isolated and cultured under standard and modified conditions. Contractile responses of fresh and cultured PA were compared using vessel myograph. Electrophysiology experiments on isolated PASMCs used the patch-clamp technique. K(+) channel expression was quantified using reverse transcription and real-time PCR. KEY RESULTS: After 4 days in culture vessels contracted to phenylephrine, but relaxation to carbachol was significantly impaired. Contractile responses to 10 mM KCl, 4-aminopyridine and tetraethylammonium increased, and vessels developed an uncharacteristic relaxation response to Ca(2+)-free solution, nifedipine and levcromakalim. PASMCs from cultured vessels were depolarized and K(+) currents reduced, in association with down-regulation of K(v)1.5, K(v)2.1 and TWIK-related acid-sensitive K(+) channel-1 mRNA. These changes were partially reversed by increased oxygenation of the culture medium or removing the endothelium before culture. CONCLUSIONS AND IMPLICATIONS: Culture of PA for 3-4 days induced loss of functional K(+) channels, depolarization of PASMCs, Ca(2+) influx, intrinsic tone and spontaneous constrictions, similar to the effects of chronic hypoxia. This limits the use of cultured vessels for studying excitation-contraction coupling, although oxygenating the culture medium and removing the endothelium can help to retain normal smooth muscle function.


Assuntos
Canais de Potássio/fisiologia , Artéria Pulmonar/fisiologia , Animais , Cálcio/metabolismo , Hipóxia Celular , Endotélio Vascular/fisiologia , Masculino , Potenciais da Membrana , Contração Muscular , Tono Muscular , Miócitos de Músculo Liso/metabolismo , Técnicas de Cultura de Órgãos , Oxigênio/farmacologia , Técnicas de Patch-Clamp , Canais de Potássio/biossíntese , Canais de Potássio/genética , Artéria Pulmonar/citologia , Artéria Pulmonar/metabolismo , RNA Mensageiro/biossíntese , Ratos , Ratos Sprague-Dawley
18.
Eur Biophys J ; 38(3): 305-18, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18449536

RESUMO

Two-pore domain (K(2P)) channels emerged about a decade ago and since then have been an expanding area of interest. This is because their biophysical and pharmacological properties make them good candidates to support background potassium currents and membrane potential in many cell types. There is clear evidence for TREK-1 and TASK-1 in the heart and these channels are likely to regulate cardiac action potential duration through their regulation by stretch, polyunsaturated fatty acids, pH, and neurotransmitters. TREK-1 may also have a critical role in mediating the vasodilator response of resistance arteries to polyunsaturated fatty acids, thus contributing to their protective effect on the cardiovascular system. TASK-1, on the other hand, is a strong candidate for a role in hypoxic vasoconstriction of pulmonary arteries. Many other members of the K(2P) channel family have been identified in the cardiovascular system, although their functional roles are still to be demonstrated. This review provides an up to date summary of what is known about the involvement of members of the K(2P) channel family in cells of the heart and arterial circulation. Our knowledge of their roles will improve with the rapidly increasing interest in them and as new selective pharmacological tools emerge. As their physiological roles emerge, the K(2P) family of potassium channels may offer promising therapeutic solutions to target cardiovascular diseases.


Assuntos
Sistema Cardiovascular/metabolismo , Proteínas do Tecido Nervoso/fisiologia , Canais de Potássio de Domínios Poros em Tandem/fisiologia , Animais , Doenças Cardiovasculares/tratamento farmacológico , Doenças Cardiovasculares/metabolismo , Doenças Cardiovasculares/fisiopatologia , Circulação Coronária , Humanos , Concentração de Íons de Hidrogênio , Ativação do Canal Iônico/fisiologia , Miócitos Cardíacos/fisiologia , Neurotransmissores/fisiologia
19.
Int Immunopharmacol ; 7(7): 900-11, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17499192

RESUMO

Pulmonary fibrosis is characterized by excessive deposition of extracellular matrix in interstitium resulting in respiratory failure associated with inflammation showing mainly neutrophil (PMN) recruitment. The turn over of extracellular matrix is partially regulated by proteases such as metalloproteinases (MMPs) and their inhibitors (TIMPs). We investigated the impact of PMN depletion on the MMP/TIMP-1 imbalance and the development of fibrosis in mice induced by bleomycin (0.3 mg/mouse). Administration of 200 microL of rabbit anti-mouse PMN antibody i.p. blunted the neutrophil influx detected in BAL and in whole blood one day after bleomycin administration. At day(14), hydroxyproline content was increased both in anti-PMN treated and control mice, without any difference between groups. At day one, bleomycin elicited a raise in pro-MMP-9 level in BAL that was significantly attenuated in anti-PMN depleted mice, whereas TIMP-1 and MMP-2 release were similar in both groups at day(1) and day(14). Higher RNA levels were observed in PMN-treated mice at day(1) for MMP-9 and MMP-2 and at day(14) for MMP-2 only. At day(14), bleomycin elicited a raise of TIMP-1 protein and RNA levels regardless of anti-PMN treatment, whereas MMP-9 returned to basal level. Bleomycin enhanced MMP-8 level in BAL at day(14) only for the control group. The amount of MMP-8 was more important in BAL from anti-PMN treated mice than in control mice at day(1) and day(14). PMN-depletion and the associated modifications in pro-MMP-9/TIMP-1 imbalance in lung during the early inflammatory phase do not alter susceptibility to bleomycin-induced pulmonary fibrosis.


Assuntos
Metaloproteinases da Matriz Secretadas/metabolismo , Neutropenia/imunologia , Fibrose Pulmonar/imunologia , Inibidor Tecidual de Metaloproteinase-1/genética , Inibidor Tecidual de Metaloproteinase-1/metabolismo , Animais , Soro Antilinfocitário/administração & dosagem , Bleomicina , Líquido da Lavagem Broncoalveolar/química , Líquido da Lavagem Broncoalveolar/citologia , Líquido da Lavagem Broncoalveolar/imunologia , Colágeno/metabolismo , Hidroxiprolina/metabolismo , Técnicas Imunoenzimáticas , Interleucina-6/análise , Interleucina-6/genética , Depleção Linfocítica , Masculino , Metaloproteinases da Matriz Secretadas/genética , Camundongos , Camundongos Endogâmicos C57BL , Neutropenia/induzido quimicamente , Fibrose Pulmonar/induzido quimicamente , RNA Mensageiro/isolamento & purificação , RNA Mensageiro/metabolismo , Inibidor Tecidual de Metaloproteinase-1/análise
20.
J Pharmacol Exp Ther ; 322(1): 341-50, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17460149

RESUMO

The sigma(2)-receptor agonist, ifenprodil, was suggested as an inhibitor of G protein-coupled inwardly rectifying potassium channels. Nevertheless, an analysis of the role of sigma(2) receptors in cardiac electrophysiology has never been done. This work aims i) to identify the roles of cardiac sigma(2) receptors in the regulation of cardiac K(+) channel conductances and ii) to check whether sigma(2)-receptor agonists exhibit class III antiarrhythmic properties. The sigma(2)-receptor agonists ifenprodil, threo-ifenprodil, LNP250A [threo-8-[1-(4-hydroxyphenyl)-1-hydroxy-propan-2-yl]-1-phenyl-1,3,8-triazaspiro[4,5]decane-4-one] (a derivative of ifenprodil devoid of alpha(1)-adrenergic and N-methyl-d-aspartate glutamate receptor-blocking properties), and 1,3-di(2-tolyl)guanidine were used to discriminate the effects linked to sigma(2) receptors from those of the sigma(1) subtype, induced by (+/-)-N-allylnormetazocine (SKF-10,047). The sigma(2)-receptor antagonist 3-alpha-tropanyl-2(pCl-phenoxy)butyrate (SM-21) was employed to characterize sigma(2)-mediated effects in patch-clamp experiments. In rabbits, all sigma(2)-receptor agonists reduced phenylephrine-induced cardiac arrhythmias. They prolonged action potential duration in rabbit Purkinje fibers and reduced human ether-a-go-go-related gene (HERG) K(+) currents. (+)-SKF-10,047 was completely inactive in the last two tests. The effects of threo-ifenprodil were not antagonized by SM-21. In HERG-transfected COS-7 cells, SM-21 potentiated the ifenprodil-induced blockade of the HERG current. These data suggest that sigma(2)-receptor ligands block I(Kr) and that this effect could explain part of the antiarrhythmic properties of this ligands family. Nevertheless, an interaction with HERG channels not involving sigma(2) receptors seems to share this pharmacological property. This work shows for the first time that particular caution has to be taken toward ligands with affinity for sigma(2) receptors. The repolarization prolongation and the early-afterdepolarization can be responsible for "torsades de pointe" and sudden cardiac death.


Assuntos
Antiarrítmicos/farmacologia , Canais de Potássio Éter-A-Go-Go/antagonistas & inibidores , Coração/efeitos dos fármacos , Piperidinas/farmacologia , Bloqueadores dos Canais de Potássio/farmacologia , Receptores sigma/agonistas , Potenciais de Ação/efeitos dos fármacos , Animais , Pressão Sanguínea/efeitos dos fármacos , Células COS , Chlorocebus aethiops , Canal de Potássio ERG1 , Eletrocardiografia/efeitos dos fármacos , Canais de Potássio Éter-A-Go-Go/fisiologia , Feminino , Masculino , Fenazocina/análogos & derivados , Fenazocina/farmacologia , Fenilefrina/farmacologia , Ramos Subendocárdicos/efeitos dos fármacos , Ramos Subendocárdicos/fisiologia , Coelhos , Receptores sigma/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...