Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Cell Tissue Res ; 369(3): 567-578, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28451751

RESUMO

The natriuretic peptides, Atrial-, B-type and C-type natriuretric peptides (ANP, BNP, CNP), are regulators of many endocrine tissues and exert their effects predominantly through the activation of their specific guanylyl cyclase receptors (GC-A and GC-B) to generate cGMP. Whereas cGMP-independent signalling has been reported in response to natriuretic peptides, this is mediated via either the clearance receptor (Npr-C) or a renal-specific NPR-Bi isoform, which both lack intrinsic guanylyl cyclase activity. Here, we report evidence of GC-B-dependent cGMP-independent signalling in pituitary GH3 cells. Stimulation of GH3 cells with CNP resulted in a rapid and sustained enhancement of ERK1/2 phosphorylation (P-ERK1/2), an effect that was not mimicked by dibutryl-cGMP. Furthermore, CNP-stimulated P-ERK1/2 occurred at concentrations below that required for cGMP accumulation. The effect of CNP on P-ERK1/2 was sensitive to pharmacological blockade of MEK (U0126) and Src kinases (PP2). Silencing of the GC-B1 and GC-B2 splice variants of the GC-B receptor by using targeted short interfering RNAs completely blocked the CNP effects on P-ERK1/2. CNP failed to alter GH3 cell proliferation or cell cycle distribution but caused a concentration-dependent increase in the activity of the human glycoprotein α-subunit promoter (αGSU) in a MEK-dependent manner. Finally, CNP also activated the p38 and JNK MAPK pathways in GH3 cells. These findings reveal an additional mechanism of GC-B signalling and suggest additional biological roles for CNP in its target tissues.


Assuntos
Guanilato Ciclase/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Peptídeo Natriurético Tipo C/farmacologia , Somatotrofos/metabolismo , Animais , Linhagem Celular , GMP Cíclico/metabolismo , Humanos , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Fosforilação/efeitos dos fármacos , Regiões Promotoras Genéticas/genética , Receptores Acoplados a Guanilato Ciclase/metabolismo , Somatotrofos/efeitos dos fármacos
2.
Cell Tissue Res ; 355(2): 425-36, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24352806

RESUMO

The guanylyl cyclases, GC-A and GC-B, are selective receptors for atrial and C-type natriuretic peptides (ANP and CNP, respectively). In the anterior pituitary, CNP and GC-B are major regulators of cGMP production in gonadotropes and yet mouse models of disrupted CNP and GC-B indicate a potential role in growth hormone secretion. In the current study, we investigate the molecular and pharmacological properties of the CNP/GC-B system in somatotrope lineage cells. Primary rat pituitary and GH3 somatolactotropes expressed functional GC-A and GC-B receptors that had similar EC50 properties in terms of cGMP production. Interestingly, GC-B signaling underwent rapid homologous desensitization in a protein phosphatase 2A (PP2A)-dependent manner. Chronic exposure to either CNP or ANP caused a significant down-regulation of both GC-A- and GC-B-dependent cGMP accumulation in a ligand-specific manner. However, this down-regulation was not accompanied by alterations in the sub-cellular localization of these receptors. Heterologous desensitization of GC-B signaling occurred in GH3 cells following exposure to either sphingosine-1-phosphate or thyrotrophin-releasing hormone (TRH). This heterologous desensitization was protein kinase C (PKC)-dependent, as pre-treatment with GF109203X prevented the effect of TRH on CNP/GC-B signaling. Collectively, these data indicate common and distinct properties of particulate guanylyl cyclase receptors in somatotropes and reveal that independent mechanisms of homologous and heterologous desensitization occur involving either PP2A or PKC. Guanylyl cyclase receptors thus represent potential novel therapeutic targets for treating growth-hormone-associated disorders.


Assuntos
Lactotrofos/enzimologia , Receptores do Fator Natriurético Atrial/metabolismo , Transdução de Sinais , Animais , Fator Natriurético Atrial/farmacologia , Sinalização do Cálcio/efeitos dos fármacos , Linhagem Celular , AMP Cíclico/metabolismo , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/genética , Endocitose/efeitos dos fármacos , Lactotrofos/efeitos dos fármacos , Ligantes , Camundongos , Peptídeo Natriurético Tipo C/farmacologia , Proteína Quinase C/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Transdução de Sinais/efeitos dos fármacos , Esfingolipídeos/metabolismo , Hormônio Liberador de Tireotropina/metabolismo
3.
J Cardiovasc Pharmacol Ther ; 15(1): 53-9, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20133496

RESUMO

Hydrogen sulfide (H( 2)S) is a biological mediator produced by enzyme-regulated pathways from L-cysteine, which is a substrate for cystathionine-gamma-lyase (CSE). In myocardium, endogenously and exogenously administered H(2)S has been shown to protect against ischemia-reperfusion injury. We hypothesized that L-cysteine exerts its protective action through stimulation of H(2)S production. Rat isolated hearts were Langendorff-perfused and underwent 35-minute regional ischemia and 120-minute reperfusion. L-cysteine perfusion from 10 minutes before ischemia until 10 minutes after reperfusion limited infarct size in a concentration-dependent manner, maximal at 1 mmol/L (control 36.4% +/- 2.4% vs L-cysteine 24.3% +/- 3.4%, P < .05). This protective action was attenuated by the CSE inhibitor, DL-propargylglycine (PAG) 1 mmol/L (31.4 +/- 5.9%, not significant vs control) but administration of the CSE cofactor pyridoxal-5'-phosphate (PLP) 50 mumol/L did not enhance the effect of L-cysteine. Ten minutes normoxic perfusion with L-cysteine 1 mmol/L caused a 3-fold increase in myocardial H(2)S concentration (0.64 +/- 0.16 vs 2.01 +/- 0.07 mumol/g protein, P < .01), an effect that was significantly attenuated by PAG (1.17 +/- 0.15 mumol/g protein). These data provide evidence that exogenous L-cysteine administration limits ischemia-reperfusion injury through a mechanism that appears to be at least partially dependent on H(2)S synthesis.


Assuntos
Cisteína/farmacologia , Sulfeto de Hidrogênio/metabolismo , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Traumatismo por Reperfusão Miocárdica/fisiopatologia , Alcinos/farmacologia , Análise de Variância , Animais , Modelos Animais de Doenças , Quimioterapia Combinada , Inibidores Enzimáticos/farmacologia , Glicina/análogos & derivados , Glicina/farmacologia , Sulfeto de Hidrogênio/análise , Masculino , Isquemia Miocárdica , Fosfato de Piridoxal/farmacologia , Ratos , Ratos Sprague-Dawley , Complexo Vitamínico B/farmacologia
4.
Curr Opin Pharmacol ; 9(2): 177-88, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19109069

RESUMO

Postconditioning is an intervention in which controlled, brief, intermittent periods of ischaemia at the onset of reperfusion protect myocardium from the lethal consequences of reperfusion ('reperfusion injury'). Postconditioning has been demonstrated in humans with acute myocardial infarction and offers the possibility of further limiting infarct size in patients undergoing reperfusion therapy. We review current research that focuses on the molecular mechanisms of postconditioning. The molecular pathways are incompletely mapped but they probably converge on suppression of mitochondrial permeability transition pore opening during early reperfusion, an event that is thought to promote cell death at reperfusion. A number of upstream signalling pathways, activated by autacoid factors, converge on this crucial target and these offer a range of realistic possibilities for pharmacological induction of a postconditioned state.


Assuntos
Cardiotônicos/uso terapêutico , Isquemia Miocárdica/tratamento farmacológico , Traumatismo por Reperfusão Miocárdica/tratamento farmacológico , Animais , Proteínas Reguladoras de Apoptose , Fator Natriurético Atrial/uso terapêutico , Autacoides/farmacologia , Sistemas de Liberação de Medicamentos , Humanos , Precondicionamento Isquêmico Miocárdico/métodos , Proteínas de Transporte da Membrana Mitocondrial/antagonistas & inibidores , Proteínas de Transporte da Membrana Mitocondrial/fisiologia , Poro de Transição de Permeabilidade Mitocondrial , Traumatismo por Reperfusão Miocárdica/terapia , Antagonistas de Entorpecentes , Agonistas do Receptor Purinérgico P1 , Antagonistas de Receptores Purinérgicos P1 , Receptores Opioides/agonistas , Receptores Opioides/uso terapêutico , Receptores Purinérgicos P1/uso terapêutico
5.
Heart Fail Rev ; 12(3-4): 279-91, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17516166

RESUMO

The myocardium represents a major source of several families of peptide hormones under normal physiological conditions and the plasma concentrations of many of these "cardiac peptides" (or related pro-peptide fragments) are substantially augmented in many cardiac disease states. In addition to well-characterised endocrine functions of several of the cardiac peptides, pleiotropic functions within the myocardium and the coronary vasculature represent a significant aspect of their actions in health and disease. Here, we focus specifically on the cardioprotective roles of four major peptide families in myocardial ischemia and reperfusion: adrenomedullin, kinins, natriuretic peptides and the urocortins. The patterns of early release of all these peptides are consistent with roles as autacoid cardioprotective mediators. Clinical and experimental research indicates the early release and upregulation of many of these peptides by acute ischemia and there is a convincing body of evidence showing that exogenously administered adrenomedullin, bradykinin, ANP, BNP, CNP and urocortins are all markedly protective against experimental myocardial ischemia-reperfusion injury through a conserved series of cytoprotective signal transduction pathways. Intriguingly, all the peptides examined so far have the potential to salvage against infarction when administered specifically during early reperfusion. Thus, the myocardial secretion of peptide hormones likely represents an early protective response to ischemia. Further work is required to explore the potential therapeutic manipulation of these peptides in acute coronary syndromes and their promise as biomarkers of acute myocardial ischemia.


Assuntos
Isquemia Miocárdica/prevenção & controle , Reperfusão Miocárdica , Miocárdio , Hormônios Peptídicos/fisiologia , Adrenomedulina , Bradicinina , Humanos , Infarto do Miocárdio/complicações , Isquemia Miocárdica/fisiopatologia , Peptídeos Natriuréticos , Transdução de Sinais
6.
Am J Physiol Heart Circ Physiol ; 292(6): H2598-606, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17220176

RESUMO

Intracellular signal transduction events in reperfusion following ischemia influence myocardial infarct development. Here we investigate the role of Rho kinase (ROCK) activation as a specific injury signal during reperfusion via attenuation of the reperfusion injury salvage kinase (RISK) pathway phosphatidylinositol 3-kinase (PI3K)/Akt/endothelial nitric oxide (NO) synthase (eNOS). Rat isolated hearts underwent 35 min of left coronary artery occlusion and 120 min of reperfusion. Phosphorylation of the ROCK substrate protein complex ezrin-radixin-moesin, assessed by immunoblotting and immunofluorescence, was used as a marker of ROCK activation. Infarct size was determined by tetrazolium staining, and terminal dUTP nick-end labeling (TUNEL) positivity was used as an index of apoptosis. The ROCK inhibitors fasudil or Y-27632 given 10 min before ischemia until 10 min after reperfusion reduced infarct size (control, 34.1 +/- 3.8%; 5 microM fasudil, 18.2 +/- 3.1%; 0.3 microM Y-27632, 19.4 +/- 4.4%; 5 microM Y-27632, 9.2 +/- 2.9%). When 5 microM Y-27632 was targeted specifically during early reperfusion, robust infarct limitation was observed (14.2 +/- 2.6% vs. control 33.4 +/- 4.4%, P<0.01). The protective action of Y-27632 given at reperfusion was attenuated by wortmannin (29.2 +/- 6.1%) and N(omega)-nitro-L-arginine methyl ester (30.4 +/- 5.7%), confirming a protective mechanism involving PI3K/Akt/NO. Ezrin-radixin-moesin phosphorylation in risk zone myocardium confirmed early and sustained ROCK activation during reperfusion and its inhibition by Y-27632. Inhibition of ROCK activation at reperfusion reduced the proportion of TUNEL-positive nuclei in the infarcted region. In conclusion, ROCK activation occurs specifically during early reperfusion. Inhibition of ROCK at reperfusion onset limits infarct size through an Akt/eNOS-dependent mechanism, suggesting that ROCK activation at reperfusion may be deleterious through suppression of the RISK pathway.


Assuntos
Apoptose/efeitos dos fármacos , Cardiotônicos/farmacologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Infarto do Miocárdio/metabolismo , Traumatismo por Reperfusão Miocárdica/metabolismo , Miocárdio/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais/efeitos dos fármacos , 1-(5-Isoquinolinasulfonil)-2-Metilpiperazina/análogos & derivados , 1-(5-Isoquinolinasulfonil)-2-Metilpiperazina/farmacologia , Amidas/farmacologia , Androstadienos/farmacologia , Animais , Cardiotônicos/uso terapêutico , Circulação Coronária/efeitos dos fármacos , Proteínas do Citoesqueleto/metabolismo , Ativação Enzimática , Técnicas In Vitro , Peptídeos e Proteínas de Sinalização Intracelular/antagonistas & inibidores , Masculino , Proteínas de Membrana/metabolismo , Proteínas dos Microfilamentos/metabolismo , Complexos Multiproteicos/metabolismo , Infarto do Miocárdio/enzimologia , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Infarto do Miocárdio/prevenção & controle , Traumatismo por Reperfusão Miocárdica/enzimologia , Traumatismo por Reperfusão Miocárdica/patologia , Traumatismo por Reperfusão Miocárdica/fisiopatologia , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Miocárdio/enzimologia , Miocárdio/patologia , NG-Nitroarginina Metil Éster/farmacologia , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo III/antagonistas & inibidores , Óxido Nítrico Sintase Tipo III/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação , Inibidores de Proteínas Quinases/uso terapêutico , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/metabolismo , Piridinas/farmacologia , Ratos , Ratos Sprague-Dawley , Wortmanina , Quinases Associadas a rho
7.
J Mol Cell Cardiol ; 41(2): 360-3, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16842816

RESUMO

Exogenous administration or transfection of adrenomedullin (AM) affords protection against ischaemia-reperfusion injury. Here we have examined the role of endogenous AM in regulating the development of myocardial infarction. Wild type (WT) and AM(+/-) mice underwent 30 min regional myocardial ischaemia and 120 min reperfusion. In AM(+/-) hearts, tetrazolium-determined infarct size was greater than in WT controls (27.9 +/- 2.0 vs. 17.7 +/- 2.4%, P < 0.01) and mortality rate was increased (35% vs. 14%, P < 0.05). Treatment with exogenous recombinant AM (200 ng/kg) prior to coronary occlusion rescued the ischaemia-reperfusion intolerant phenotype of AM(+/-) mice and further limited infarct development in WT mice. Administration of recombinant AM was associated with augmented phosphorylation of Akt and eNOS in early reperfusion suggesting a role for AM in regulating this survival pathway. These studies provide the first evidence that expression of AM is a critical factor regulating myocardial tolerance to ischaemia-reperfusion injury.


Assuntos
Regulação da Expressão Gênica , Infarto do Miocárdio/metabolismo , Traumatismo por Reperfusão Miocárdica/metabolismo , Peptídeos/metabolismo , Transdução de Sinais , Adrenomedulina , Animais , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Masculino , Camundongos , Infarto do Miocárdio/prevenção & controle , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Óxido Nítrico Sintase Tipo II/metabolismo , Óxido Nítrico Sintase Tipo III , Proteína Oncogênica v-akt/metabolismo , Peptídeos/farmacologia , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacologia , Transdução de Sinais/efeitos dos fármacos
8.
Basic Res Cardiol ; 101(1): 53-60, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16328106

RESUMO

Hydrogen sulfide (H2S) is a gaseous mediator, produced by the metabolic pathways that regulate tissue concentrations of sulfur-containing amino acids. Recent studies indicate that endogenous or exogenous H2S exerts physiological effects in the cardiovascular system of vertebrates, possibly through modulation of K ATP channel opening. The present study was undertaken to examine the hypothesis that H2S is cytoprotective against myocardial ischemia-reperfusion injury and that this protective action is mediated by K ATP opening. Rat isolated hearts were Langendorff-perfused and underwent 30 min left main coronary artery occlusion and 120 min reperfusion. The resulting injury was assessed as infarct size, determined by tetrazolium staining. Treatment of hearts with the H2S-donor, NaHS, commencing 10 min prior to the onset of coronary occlusion and maintained until 10 min reperfusion, resulted in a concentration-dependent limitation of infarct size (control, 41.0 +/- 2.6% of risk zone; NaHS 0.1 microM, 33.9 +/- 2.1%, [0.05 > P < 0.1]; NaHS 1 microM, 20.2 +/- 2.1% [P < 0.01]). Pretreatment with the K ATP channel blockers glibenclamide 10 microM or sodium 5-hydroxydecanoate (5HD) 100 microM led to abrogation of the infarct-limiting effect of NaHS 1 microM (glibenclamide + NaHS 42.5 +/- 3.6%; 5HD + NaHS 44.7 +/- 2.2%). No statistically significant effects of NaHS treatment on coronary flow, heart rate or left ventricular developed pressure were observed in this experimental preparation. These data provide the first evidence that exogenous H2S protects against irreversible ischemia-reperfusion injury in myocardium and support the involvement of K ATP opening in the mechanism of action. Further work is required to elucidate the potential role of endogenous H2S as a cytoprotective mediator against myocardial ischemia-reperfusion injury, the mechanisms regulating its generation, and the nature of its interaction with protein targets such as the K ATP channel.


Assuntos
Sulfeto de Hidrogênio/uso terapêutico , Isquemia Miocárdica/tratamento farmacológico , Traumatismo por Reperfusão Miocárdica/tratamento farmacológico , Canais de Potássio/fisiologia , Trifosfato de Adenosina/metabolismo , Animais , Antiarrítmicos/farmacologia , Débito Cardíaco/efeitos dos fármacos , Ácidos Decanoicos/farmacologia , Glibureto/farmacologia , Frequência Cardíaca/efeitos dos fármacos , Hidroxiácidos/farmacologia , Técnicas In Vitro , Masculino , Contração Miocárdica/efeitos dos fármacos , Isquemia Miocárdica/fisiopatologia , Traumatismo por Reperfusão Miocárdica/fisiopatologia , Ratos , Ratos Sprague-Dawley
9.
Pharmacol Ther ; 105(2): 95-112, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15670621

RESUMO

During and following acute myocardial infarction, a variety of endogenous mediators are elevated, one of which is adrenomedullin (AM). AM is a multifunctional peptide that has been identified as having a putative beneficial role following an ischemic insult at both systemic and local levels. Classically described as a potent vasodilator, natriuretic, and diuretic agent, experimental infarct models also demonstrate AM to exhibit antiproliferative and antiapoptotic functions in the myocardium, counterregulating the effects of mediators such as angiotensin-II and endothelin-1. Less well documented are the angiogenic and inflammatory modulating potentials of AM, which may also contribute toward reducing adverse ventricular remodeling. The review examines clinical and experimental studies, looking at the effects of AM and cellular mechanisms that could be involved in mediating cardioprotective effects and ultimately optimizing left ventricular remodeling. Finally, the possibility of enhancing endogenous actions of AM by pharmacological intervention is considered.


Assuntos
Infarto do Miocárdio/metabolismo , Peptídeos/fisiologia , Adrenomedulina , Animais , Sobrevivência Celular , Vasos Coronários/metabolismo , Vasos Coronários/fisiopatologia , Homeostase , Humanos , Mediadores da Inflamação/metabolismo , Infarto do Miocárdio/fisiopatologia , Traumatismo por Reperfusão Miocárdica/metabolismo , Traumatismo por Reperfusão Miocárdica/fisiopatologia , Neovascularização Patológica/metabolismo , Estresse Oxidativo , Transdução de Sinais , Vasodilatação , Remodelação Ventricular
10.
Curr Pharm Des ; 10(20): 2477-82, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15320757

RESUMO

The natriuretic peptides are a family of widely distributed polypeptide mediators that exert a range of actions in several body systems. In cardiovascular homeostasis, the endocrine roles of the cardiac-derived atrial and B-type natriuretic peptide (ANP and BNP) in regulating central fluid volume and blood pressure have been recognised for two decades. However, there is a growing realisation that natriuretic peptide actions go far beyond their endocrine effects and that local (autocrine/paracrine) regulatory actions within the heart and coronary vasculature may be of comparable importance, especially in disease states where tissue and circulating levels of the peptides rise markedly. In acute myocardial ischaemia, release of BNP occurs rapidly from ventricular myocardium, prompting speculation that the early activation of the natriuretic peptide receptor/cGMP signalling system may be an important autocrine/paracrine response in cardiac ischaemia. The autocrine/paracrine actions include inotropic effects, the acute regulation of coronary vascular tone and the attenuation of the susceptibility of myocardium to ischaemic injury. The effects of longer-term upregulation of natriuretic peptide expression in the heart could include the suppression of growth and proliferative responses in a variety of myocardial and vascular cells. In a variety of preparations, acute exposure of epicardial coronary arteries to pharmacological concentrations of natriuretic peptides evokes vasorelaxation, although in coronary microvessels, evidence for a vasorelaxant action of the peptides is less consistent. The mechanisms of the coronary vasorelaxant action are unclear but limited evidence suggests an endothelium-dependent component. In ischaemic myocardium, acute treatment with BNP prior to and during coronary artery occlusion exerts a markedly protective, concentration-dependent infarct-limiting action. This cytoprotective effect of the natriuretic peptide signalling pathway might conceivably represent an alternative endogenous salvage pathway in myocardium which is potentially exploitable therapeutically. Taken together, the acute actions of natriuretic peptides on the coronary vasculature and in myocardial ischaemia suggest a profile of activity that may be therapeutically beneficial in the management of patients with acute coronary syndromes.


Assuntos
Fator Natriurético Atrial/farmacologia , Vasos Coronários/efeitos dos fármacos , Isquemia Miocárdica/tratamento farmacológico , Animais , Fator Natriurético Atrial/química , Fator Natriurético Atrial/fisiologia , Vasos Coronários/química , Vasos Coronários/metabolismo , Ventrículos do Coração/metabolismo , Ventrículos do Coração/patologia , Humanos , Isquemia Miocárdica/metabolismo , Isquemia Miocárdica/patologia
11.
Pharmacol Ther ; 101(2): 113-29, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14761702

RESUMO

The natriuretic peptides, atrial natriuretic peptide (ANP), brain natriuretic peptide (BNP), and C-type natriuretic peptide (CNP), are a family of polypeptide mediators exerting numerous actions in cardiovascular homeostasis. ANP and BNP are cardiac derived, being secreted and up-regulated in myocardium in response to many pathophysiological stimuli. CNP is an endothelium-derived mediator. The classical endocrine effects of ANP and BNP on fluid homeostasis and blood pressure, especially in conditions characterised by left ventricular dysfunction, are well recognised and extensively researched. However, there is accumulating evidence that, in addition to endocrine actions, ANP and BNP exhibit important autocrine and paracrine functions within the heart and coronary circulation. These include regulation of myocyte growth, inhibition of fibroblast proliferation and extracellular matrix deposition, a cytoprotective anti-ischaemic (preconditioning-like) function, and influences on coronary endothelium and vascular smooth muscle proliferation and contractility. Most if not all of these actions can be ascribed to particulate guanylyl cyclase activation because the ANP/BNP receptor, natriuretic peptide receptor (NPR)-A, has an intracellular guanylyl cyclase domain. Subsequent elevation of the intracellular second messenger cGMP may exert diverse physiological effects through activation of cGMP-dependent protein kinases (cGK), predominantly cGK-I. However, there appear to be other contributory mechanisms in several of these actions, including the augmentation of nitric oxide synthesis. These diverse actions may represent counterregulatory mechanisms in the pathophysiology of many cardiovascular diseases, not just those typified by left ventricular dysfunction. Ultimately, insights from the autocrine/paracrine actions of natriuretic peptides may provide routes to therapeutic application in cardiac diseases of natriuretic peptides and drugs that modify their availability.


Assuntos
Comunicação Autócrina/fisiologia , Coração/fisiologia , Peptídeos Natriuréticos/fisiologia , Comunicação Parácrina/fisiologia , Animais , Guanilato Ciclase/fisiologia , Contração Miocárdica/fisiologia , Isquemia Miocárdica/metabolismo , Isquemia Miocárdica/fisiopatologia , Receptores do Fator Natriurético Atrial/fisiologia , Transdução de Sinais/fisiologia , Vasodilatação/fisiologia
12.
Basic Res Cardiol ; 99(2): 71-5, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-14963664

RESUMO

The natriuretic peptides are a family of widely distributed, but evolutionarily conserved, polypeptide mediators that exert a range of actions throughout the body. In cardiovascular homeostasis, the endocrine roles of the cardiac-derived atrial and B-type natriuretic peptide (ANP and BNP) in regulating central fluid volume and blood pressure have been recognised for two decades. However, there is a growing realisation that natriuretic peptide actions go far beyond their volume regulating effects. These pleiotropic actions include local (autocrine/paracrine) regulatory actions of ANP and BNP within the heart, and of another natriuretic peptide, CNP, within the vessel wall. Effects on function and growth of the local tissue environment are likely to be of great importance, especially in disease states where tissue and circulating levels of ANP and BNP rise markedly. At present, the relevance of other natriuretic peptides (notably uroguanylin and DNP) to human physiology and pathology remain uncertain. Other articles in this issue of Basic Research in Cardiology review the molecular physiology of natriuretic peptide signalling, with a particular emphasis on the lessons from genetically targetted mice; the vascular activity of natriuretic peptides; the regulation and roles of natriuretic peptides in ischaemic myocardium; and the diagnostic, prognostic and therapeutic roles of natriuretic peptides in heart failure.


Assuntos
Peptídeos Natriuréticos , Animais , GMP Cíclico/metabolismo , História do Século XX , Humanos , Peptídeos Natriuréticos/história , Peptídeos Natriuréticos/metabolismo , Peptídeos Natriuréticos/fisiologia , Receptores de Neuropeptídeos/metabolismo , Transdução de Sinais
13.
J Mol Cell Cardiol ; 35(9): 1063-71, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12967629

RESUMO

Ischaemic preconditioning (IPC) protects the heart against myocardial infarction acutely as well as several hours later (e.g. 24-48 h). The mechanism of the profound cardioprotection is not completely explored. We hypothesized that PI3K/PDK1/Akt/mTOR/p70S6K-mediated pro-survival pathway is involved in delayed cardioprotection induced by IPC. Under Hypnorm-Diazepam anaesthesia, male New Zealand White rabbits were either sham-operated (SC) or preconditioned by four cycles of 5-min ischaemia and 10-min reperfusion on day 1. Twenty-four hours after recovery, the animals were anaesthetized with sodium pentobarbitone and subjected to 30-min ischaemia followed by 180-min reperfusion. Wortmannin (0.6 mg/kg, i.v.), an irreversible PI3 kinase (PI3K) inhibitor, rapamycin (0.25 mg/kg, i.v.), which prevents the phosphorylation of p70S6 kinase (p70S6K), or DMSO (control vehicle) was given 15 min prior to IPC. IPC significantly reduced infarct size compared to the control group (SC) (31.9 +/- 5.8% (n = 7) vs. 54.9 +/- 2.9% (n = 6), P < 0.05). Wortmannin and rapamycin alone had no effect on infarct size (56.3 +/- 1.6% (n = 6) and 54.7 +/- 3.8% (n = 6), respectively). However, when wortmannin or rapamycin were given prior to IPC the protection was completely abolished (49.9 +/- 2.8% (n = 6), 45.1 +/- 4.6% (n = 7), P < 0.05 vs. IPC). Western blot analysis showed that wortmannin, at a dose of 0.6 mg/kg, and rapamycin, at a dose of 0.25 mg/kg, were sufficient to prevent phosphorylation of Akt and p70S6K, respectively, when the inhibitors were given prior to IPC. We conclude that PI3K/PDK1/Akt/mTOR/p70S6K-signalling pathway plays an essential role in the development of the cardioprotection against infarction in rabbits.


Assuntos
Precondicionamento Isquêmico Miocárdico , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Serina-Treonina Quinases , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Androstadienos/farmacologia , Animais , Interações Medicamentosas , Inibidores Enzimáticos/farmacologia , Masculino , Modelos Biológicos , Infarto do Miocárdio/patologia , Infarto do Miocárdio/prevenção & controle , Inibidores de Fosfoinositídeo-3 Quinase , Proteínas Proto-Oncogênicas/efeitos dos fármacos , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Coelhos , Proteínas Quinases S6 Ribossômicas 70-kDa/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Sirolimo/farmacologia , Fatores de Tempo , Wortmanina
14.
Cardiovasc Drugs Ther ; 17(5-6): 415-25, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-15107596

RESUMO

AMP579, an adenosine A(1)/A(2A) receptor agonist, protects against myocardial infarction when given at the onset of reperfusion. However, it is unclear which receptor subtype mediates its protective actions. Anaesthetised rabbits were subjected to 30 min regional ischaemia/180 min reperfusion in vivo. AMP579 (30 microg kg(-1) bolus + 3 microg kg(-1) min(-1) for 70 min) reduced heart rate and mean arterial blood pressure with the latter being abolished with ZM241385 (a selective A(2A) receptor antagonist). AMP579 reduced infarct size from 46.0 +/- 3.4% in vehicle control hearts to 29.6 +/- 3.5% (P < 0.05), an effect that was attenuated in the presence of ZM241385, in a dose-dependent manner (38.2 +/- 4.9% at 1 mg kg(-1); 45.1 +/- 4.2% at 2.5 mg kg(-1)). CGS21680 (a selective A(2A) agonist, 30 microg kg(-1) bolus + 3 microg kg(-1) min(-1) for 70 min), or CCPA (a selective A(1) agonist, 50 microg kg(-1)), alone or in combination showed no protection (44.7 +/- 5.8%; 39.8 +/- 2.8%; 39.1 +/- 5.1%, respectively) when given at the commencement of reperfusion. Furthermore, we hypothesized that the prosurvival MEK1/2-Erk1/2 pathway was involved in the downstream mechanism of cardioprotection afforded by AMP579. PD098059, an inhibitor of MEK1/2 showed a dose dependent attenuation on infarct size (39.9 +/- 5.3% at 2 mg kg(-1); 48.3 +/- 5.7% at 4 mg kg(-1), i.v., respectively). PD098059 alone had no effect on infarct size (44.7 +/- 5.8%, 2 mg kg(-1), i.v.). We conclude that AMP579 limits myocardial infarction by activating A(2A) adenosine receptors that might be linked to further downstream kinases such as Erk1/2.


Assuntos
Agonistas do Receptor A1 de Adenosina , Agonistas do Receptor A2 de Adenosina , Imidazóis/farmacologia , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Traumatismo por Reperfusão Miocárdica/tratamento farmacológico , Piridinas/farmacologia , Animais , Relação Dose-Resposta a Droga , Flavonoides/farmacologia , Imidazóis/administração & dosagem , MAP Quinase Quinase 1 , MAP Quinase Quinase 2 , Masculino , Proteína Quinase 3 Ativada por Mitógeno , Quinases de Proteína Quinase Ativadas por Mitógeno/antagonistas & inibidores , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Infarto do Miocárdio/prevenção & controle , Traumatismo por Reperfusão Miocárdica/metabolismo , Proteínas Tirosina Quinases/antagonistas & inibidores , Piridinas/administração & dosagem , Coelhos
15.
Cardiovasc Res ; 55(3): 534-43, 2002 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-12160950

RESUMO

OBJECTIVE: We propose that ischemic preconditioning (IPC) and mitochondrial K(ATP) channel activation protect the myocardium by inhibiting mitochondrial permeability transition pore (MPTP) opening at reperfusion. METHODS: Isolated rat hearts were subjected to 35 min ischemia/120 min reperfusion and assigned to the following groups: (1) control; (2) IPC of 2x5 min each of preceding global ischemia; (3,4,5) 0.2 micromol/l cyclosporin A (CsA, which inhibits MPTP opening), 5 micromol/l FK506 (which inhibits the phosphatase calcineurin without inhibiting MPTP opening), or 20 micromol/l atractyloside (Atr, a MPTP opener) given at reperfusion; (6,7) pre-treatment with 30 micromol/l diazoxide (Diaz, a mitochondrial K(ATP) channel opener) or 200 nmol/l 2 chloro-N(6)-cyclopentyl-adenosine (CCPA, an adenosine A1 receptor agonist); (8) IPC+Atr; (9) Diaz+Atr; (10) CCPA+Atr. The effect of mitochondrial K(ATP) channel activation on calcium-induced MPTP opening in isolated calcein-loaded mitochondria was also assessed. RESULTS: IPC, CsA when given at reperfusion, and pre-treatment with diazoxide or CCPA all limited infarct size (19.9+/-2.6% in IPC; 24.6+/-1.9% in CsA, 18.0+/-1.7% in Diaz, 20.4+/-3.3% in CCPA vs. 44.7+/-2.0% in control, P<0.0001). Opening the MPTP with atractyloside at reperfusion abolished this cardio-protective effect (47.7+/-1.8% in IPC+Atr, 42.3+/-3.2% in Diaz+Atr, 51.2+/-1.6% in CCPA+Atr). Atractyloside and FK506, given at reperfusion, did not influence infarct size (45.7+/-2.1% in Atr and 43.1+/-3.6% in FK506 vs. 44.7+/-2.0% in control, P=NS). Diazoxide (30 micromol/l) was shown to reduce calcium-induced MPTP opening by 52.5+/-8.0% in calcein-loaded mitochondria. 5-Hydroxydecanoic acid (100 micromol/l) was able to abolish the cardio-protective effects of both diazoxide and IPC. CONCLUSION: One interpretation of these data is that IPC and mitochondrial K(ATP) channel activation may protect the myocardium by inhibiting MPTP opening at reperfusion.


Assuntos
Ciclosporina/farmacologia , Canais Iônicos/efeitos dos fármacos , Precondicionamento Isquêmico Miocárdico/métodos , Mitocôndrias Cardíacas/metabolismo , Isquemia Miocárdica/metabolismo , Adenosina/análogos & derivados , Adenosina/farmacologia , Análise de Variância , Animais , Atractilosídeo/farmacologia , Inibidores de Calcineurina , Ácidos Decanoicos/farmacologia , Diazóxido/farmacologia , Inibidores Enzimáticos/farmacologia , Hidroxiácidos/farmacologia , Masculino , Proteínas de Transporte da Membrana Mitocondrial , Poro de Transição de Permeabilidade Mitocondrial , Traumatismo por Reperfusão Miocárdica/metabolismo , Perfusão , Permeabilidade , Bloqueadores dos Canais de Potássio/farmacologia , Canais de Potássio/efeitos dos fármacos , Canais de Potássio Cálcio-Ativados/metabolismo , Agonistas do Receptor Purinérgico P1 , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Tacrolimo/farmacologia
16.
Basic Res Cardiol ; 97(4): 268-75, 2002 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12111036

RESUMO

Granulocytes, especially neutrophils, are recruited in myocardium during the evolution of acute myocardial infarction. Because the neutrophil reaction is most intense during reperfusion and because these cells are a rich source of toxic oxidant species and proteolytic enzymes, it has become a widely held view that neutrophils are an important mechanism of myocardial injury extension during reperfusion. However, on close examination the evidence underlying this contention is equivocal. The basic experimental situation can be summarised thus. (1) All forms of reperfusion injury (i.e., cytotoxic or lethal cell injury, myocardial stunning, endothelial dysfunction, and reperfusion-induced arrhythmias) can be observed in neutrophil-free conditions. (2) "Anti-neutrophil" interventions (e.g., anti-inflammatory drugs, adenosine, anti-neutrophil antisera, leukocyte filters and inhibitors of the various pathways of neutrophil adhesion) do not consistently prevent reperfusion injury and they certainly do not consistently limit infarct size. (3) The time course of neutrophil accumulation in post-ischaemic myocardium may be different to the time course of injury. (4) Despite more than two decades of research, no double-blind, randomised controlled clinical trial assessing an anti-neutrophil therapy in myocardial infarction has yet reported a positive benefit that is attributable to inhibition of neutrophil recruitment. The evidence weighs against a pivotal role of neutrophils as a causal factor in most forms of ischemia-reperfusion injury. An exception may be microvascular injury and capillary plugging leading to the "no-reflow" phenomenon but even here the evidence suggests that the extent of neutrophil accumulation and microvascular injury is determined by, rather than a cause of, myocyte necrosis.


Assuntos
Traumatismo por Reperfusão Miocárdica/imunologia , Neutrófilos/imunologia , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA