Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
1.
Auton Autacoid Pharmacol ; 30(3): 185-92, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20085572

RESUMO

In vascular smooth muscle cells, relaxant actions of guanosine--3',5'-cyclic monophosphate (cGMP) are well recognized, but there is increasing evidence that cGMP also plays regulatory roles in vascular endothelium. However, the autacoid and endocrine mechanisms controlling cGMP production in endothelium are not well understood. The objective of these studies was to examine the mechanisms of cGMP accumulation in human umbilical vein endothelial cells (HUVEC) in response to natriuretic peptides. Expression in HUVEC of natriuretic peptide receptors, particulate guanylyl cyclases (GC)-A and GC-B, was confirmed by RT-PCR and Western blot analysis. In the presence of the phosphodiesterase inhibitor IBMX 500 microM, 3 h incubation of HUVEC with B-type natriuretic peptide (BNP) (preferential GC-A agonist) or C-type natriuretic peptide (CNP) (preferential GC-B agonist) stimulated concentration-dependent increases in cGMP production. At 10 and 100 nM, we observed two to three-fold greater potency of CNP compared to BNP. In the absence of IBMX, CNP-stimulated cGMP accumulation was significantly less than cGMP accumulation in response to sodium nitroprusside 1 mM. This greater sensitivity of GC-B-derived cGMP to phosphodiesterases suggests compartmentalization of two pools of cGMP from particulate and soluble guanylyl cyclases. Although CNP 100 nM and 1 microM was observed to increase nitrite + nitrate (stable metabolites of NO) production in HUVEC two-fold above basal level, the soluble guanylyl cyclase inhibitor ODQ 10 microM did not significantly modify CNP-stimulated cGMP accumulation suggesting that endothelial actions of CNP may be NO-independent. In conclusion, these studies indicate functional signaling by natriuretic peptides in endothelial cells, supporting possible roles of these mediators in regulating endothelial cell function.


Assuntos
GMP Cíclico/biossíntese , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Peptídeo Natriurético Tipo C/farmacologia , 1-Metil-3-Isobutilxantina/farmacologia , Células Cultivadas , GMP Cíclico/metabolismo , Inibidores Enzimáticos/farmacologia , Expressão Gênica/genética , Guanilato Ciclase/antagonistas & inibidores , Humanos , Peptídeo Natriurético Encefálico/farmacologia , Óxido Nítrico/metabolismo , Nitroprussiato/farmacologia , Oxidiazóis/farmacologia , Inibidores de Fosfodiesterase/farmacologia , Diester Fosfórico Hidrolases/metabolismo , Quinoxalinas/farmacologia , Receptores do Fator Natriurético Atrial/genética , Receptores de Superfície Celular/genética , Cordão Umbilical/citologia
2.
Br J Pharmacol ; 158(6): 1495-502, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19845676

RESUMO

BACKGROUND AND PURPOSE: Hyperlipidaemia interferes with cardioprotective mechanisms, but the cause of this phenomenon is largely unknown, although hyperlipidaemia impairs the cardioprotective NO-cGMP system. However, it is not known if natriuretic peptide-cGMP-protein kinase G (PKG) signalling is affected by hyperlipidaemia. Therefore, we investigated the cardioprotective efficacy of cGMP-elevating agents in hearts from normal and hyperlipidaemic rats. EXPERIMENTAL APPROACH: Male Wistar rats were rendered hyperlipidaemic by feeding with 2% cholesterol-enriched chow for 12 weeks. Hearts isolated from normal and hyperlipidaemic rats were perfused (Langendorff mode) and subjected to 30 min occlusion of the left main coronary artery, followed by 120 min reperfusion. 8-Br-cGMP (CG, 10 nM), B-type natriuretic peptide-32 (BNP, 10 nM), S-nitroso-N-acetyl-penicillamine (SNAP, 1 microM) were perfused from 10 min prior to coronary occlusion until the 15th min of reperfusion. Infarct size (% of ischaemic risk zone) was determined by triphenyltetrazolium staining. KEY RESULTS: Treatment with CG, SNAP or BNP decreased infarct size significantly in normal hearts from its control value of 41.6 +/- 2.9% to 15.5 +/- 2.4%, 23.3 +/- 3.0% and 25.3 +/- 4.6%, respectively (P < 0.05). Protection by BNP was abolished by co-perfusion of PKG inhibitors KT5823 (600 nM) or Rp-8pCPT-PET-cGMPs (1 microM), confirming its PKG dependence. In hearts from hyperlipidaemic rats, CG, SNAP or BNP failed to decrease infarct size. Hyperlipidaemia did not alter basal myocardial PKG content, but decreased its activity as assessed by phosphorylation of cardiac troponin I. CONCLUSIONS AND IMPLICATIONS: This is the first demonstration that defects in the cardioprotective cGMP-PKG system could be a critical biochemical anomaly in hyperlipidaemia.


Assuntos
Cardiotônicos/farmacologia , Proteínas Quinases Dependentes de GMP Cíclico/metabolismo , GMP Cíclico/metabolismo , Hiperlipidemias/complicações , Animais , GMP Cíclico/análogos & derivados , GMP Cíclico/farmacologia , Gorduras na Dieta/toxicidade , Modelos Animais de Doenças , Hiperlipidemias/fisiopatologia , Masculino , Infarto do Miocárdio/etiologia , Infarto do Miocárdio/prevenção & controle , Traumatismo por Reperfusão Miocárdica/fisiopatologia , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Peptídeo Natriurético Encefálico/farmacologia , Fosforilação , Ratos , Ratos Wistar , S-Nitroso-N-Acetilpenicilamina/farmacologia , Troponina I/metabolismo
3.
Br J Pharmacol ; 153(1): 1-3, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17952110

RESUMO

The last five years have witnessed a remarkable resurgence of interest in myocardial reperfusion injury. Reperfusion is absolutely essential to salvage ischaemic myocardium but experimental and clinical studies show that reperfusion-associated injury may mask the full benefits of prompt reperfusion in acute myocardial infarction. In the current issue of the British Journal of Pharmacology, Mudalagiri et al demonstrate a protective effect against simulated reperfusion injury using exogenously applied erythropoietin in human isolated myocardium. Crucially, the benefits of erythropoietin were observed when it was administered specifically during re-oxygenation. The demonstration that the protective effects of the cytokine were dependent on PI3-kinase/Akt and ERK1/2 activation provides compelling evidence that reperfusion injury salvage kinases (RISKs) are key survival mechanisms in human myocardium, as they are in experimental animal species. Although erythropoietin may be only one of several potential pharmacological approaches in human patients, this study establishes the important proof-of-principle that activation of RISKs is protective in human myocardium and could be a promising therapeutic target in acute myocardial infarction.


Assuntos
Eritropoetina/farmacologia , MAP Quinases Reguladas por Sinal Extracelular/fisiologia , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Fosfatidilinositol 3-Quinases/fisiologia , Proteínas Proto-Oncogênicas c-akt/fisiologia , Animais , Ativação Enzimática/efeitos dos fármacos , Humanos , Contração Miocárdica/efeitos dos fármacos , Proteínas Recombinantes
4.
Br J Pharmacol ; 152(6): 855-69, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17700722

RESUMO

It is clear that multiple signalling pathways regulate the critical balance between cell death and survival in myocardial ischaemia-reperfusion. Recent attention has focused on the activation of survival or salvage kinases, particularly during reperfusion, as a common mechanism of many cardioprotective interventions. The phosphatidyl inositol 3'-hydroxy kinase/Akt complex (PI3K/Akt) and p42/p44 mitogen-activated protein kinase cascades have been widely promoted in this respect but the cyclic guanosine 3',5'-monophosphate/cGMP-dependent protein kinase (cGMP/PKG) signal transduction cassette has been less systematically investigated as a survival cascade. We propose that activation of the cGMP/PKG signalling pathway, following activation of soluble or particulate guanylate cyclases, may play a pivotal role in survival signalling in ischaemia-reperfusion, especially in the classical preconditioning, delayed preconditioning and postconditioning paradigms. The resurgence of interest in reperfusion injury, largely as a result of postconditioning-related research, has confirmed that the cGMP/PKG pathway is a pivotal salvage mechanism in reperfusion. Numerous studies suggest that the infarct-limiting effects of preconditioning and postconditioning, exogenously donated nitric oxide (NO), natriuretic peptides, phosphodiesterase inhibitors, and other diverse drugs and mediators such as HMG co-A reductase inhibitors (statins), Rho-kinase inhibitors and adrenomedullin, whether given before and during ischaemia, or specifically at the onset of reperfusion, may be mediated by activation or enhancement of the cGMP pathway, either directly or indirectly via endogenous NO generation downstream of PI3K/Akt. Putative mechanisms of protection include PKG regulation of Ca(2+) homeostasis through the modification of sarcoplasmic reticulum Ca(2+) uptake mechanisms, and PKG-induced opening of ATP-sensitive K(+) channels during ischaemia and/or reperfusion. At present, significant technical obstacles in defining the precise roles played by cGMP/PKG signalling include the heavy reliance on pharmacological PKG inhibitors of uncertain selectivity, difficulties in determining PKG activity in intact tissue, and the growing recognition that intracellular compartmentalisation of the cGMP pool may contribute markedly to the nucleotide's biological actions and biochemical determination. Overall, the body of experimental evidence suggests that cGMP/PKG survival signalling ameliorates irreversible injury associated with ischaemia-reperfusion and may be a tractable therapeutic target.


Assuntos
Proteínas Quinases Dependentes de GMP Cíclico/fisiologia , GMP Cíclico/fisiologia , Traumatismo por Reperfusão Miocárdica/fisiopatologia , Transdução de Sinais/fisiologia , Animais , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Cardiotônicos/farmacologia , Cardiotônicos/uso terapêutico , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Proteínas Quinases Dependentes de GMP Cíclico/antagonistas & inibidores , Inibidores Enzimáticos/farmacologia , Inibidores Enzimáticos/uso terapêutico , Humanos , Traumatismo por Reperfusão Miocárdica/enzimologia , Traumatismo por Reperfusão Miocárdica/metabolismo , Peptídeos Natriuréticos/fisiologia , Inibidores de Fosfodiesterase/farmacologia , Inibidores de Fosfodiesterase/uso terapêutico
6.
Exp Mol Med ; 37(4): 311-22, 2005 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-16155408

RESUMO

Preconditioning of the myocardium rapidly induces a number of transcription factors, which are likely to be responsible for a cascade of transcriptional changes underlying the development of delayed adaptation. Identifying these changes provides insight into the molecular pathways elicited by sub-lethal ischaemia and the mechanism leading to delayed adaptation. Genes up-regulated in rabbit myocardium in vivo by ischaemic preconditioning following reperfusion for 2 h, 4 h and 6 h post-treatment were identified by representational difference analysis of cDNA (cDNA. RDA). The area of the left ventricle rendered ischaemic by preconditioning or the equivalent area of sham-treated animals was isolated and cDNA.RDA performed. Three novel genes and six genes with known function where identified, including the TGFbeta receptor interacting protein 1, the alpha isoform of the A subunit of PP2 and the cap binding protein NCBP1. To determine whether expression of these genes correlated with preconditioning per se, expression was measured in myocardium after both ischaemic as well as heat shock induced preconditioning following 2 h, 4 h, and 6 h reperfusion. These genes were induced in rabbit myocardium in vivo by both ischaemia and heat shock, consistent with a fundamental role in the development of delayed adaptation. The well described role of PP2 in modulating the mitogen-activated protein kinase pathway and promoting cell survival is consistent with our previous work, which identified the reperfusion injury salvage kinase pathway in mediating the protective effects of ischaemic preconditioning. Expression of Trip1 and Ncbp1 also implicates TGFbeta signalling pathways and RNA processing and transport in delayed adaptation to stress in the myocardium.


Assuntos
Regulação da Expressão Gênica , Precondicionamento Isquêmico Miocárdico , Isquemia Miocárdica/genética , Regulação para Cima , Animais , DNA Complementar/genética , Ventrículos do Coração/metabolismo , Masculino , RNA Mensageiro/análise , RNA Mensageiro/metabolismo , Coelhos
7.
Basic Res Cardiol ; 100(5): 387-96, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16010601

RESUMO

Adrenomedullin (AM) is a vascular-derived polypeptide that exerts numerous actions in cardiovascular homeostasis. Recent studies have demonstrated a cytoprotective action of exogenously applied or genetically over-expressed AM in experimental myocardial infarction. The present studies were undertaken to test the hypothesis that AM exerts its effects through direct augmentation of NO generation in the myocardium during early reperfusion. Rat isolated hearts underwent 35 min left coronary artery occlusion followed by 120 min reperfusion. Infarct size (as percentage of ischaemic riskzone) was determined by Evans' blue and tetrazolium double staining. AM 1 nM administered 5 min prior to and during the first 15 min of ischaemia did not significantly influence infarct size. However, the same concentration of AM given during the last 5 min ischaemia and first 15 min of reperfusion significantly limited infarct size (AM reperfusion 15.9 +/- 3.5% vs control 31.4 +/- 2.1%, P < 0.01). AM at reperfusion improved coronary flow and LV contractility. The protective effects of adrenomedullin were abolished in the presence of the NO synthase inhibitor, L-NAME 100 microM (infarct size 24.6 +/- 5.7%, P > 0.05 vs control). AM treatment at reperfusion was associated with augmented phosphorylation of the pro-survival kinase, Akt, determined by immunoblotting of tissue sampled 30 min following reperfusion. These studies provide the first evidence that AM exerts its cytoprotective action specifically during early reperfusion through a NO-dependent mechanism.


Assuntos
Infarto do Miocárdio/tratamento farmacológico , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Peptídeos/uso terapêutico , Adrenomedulina , Animais , Circulação Coronária/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/fisiologia , Masculino , Infarto do Miocárdio/complicações , Infarto do Miocárdio/patologia , NG-Nitroarginina Metil Éster/farmacologia , Óxido Nítrico/fisiologia , Fosfatidilinositol 3-Quinases/fisiologia , Ratos , Ratos Sprague-Dawley , Transdução de Sinais
8.
Cardiovasc Drugs Ther ; 18(2): 127-34, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15162074

RESUMO

Bradykinin is an important endogenous trigger of myocardial ischemic preconditioning (IPC). Through simultaneous inhibition of neutral endopeptidase and angiotensin converting enzyme, omapatrilat prevents enzymatic degradation of bradykinin. The aim of this study was to investigate if omapatrilat, through its ability to augment bradykinin levels, can augment a subthreshold IPC stimulus (Sub-IPC) and to compare the action of omapatrilat with the angiotensin-converting enzyme inhibitor, captopril. Langendorff perfused rat hearts were subjected to 35 min left coronary artery occlusion and 120 min reperfusion. Full IPC was induced with 5 min global ischemia/10 min reperfusion and substantially limited infarct size (21.5 +/- 3.5% of risk zone vs 53.4 +/- 2.0% in controls, P < 0.01). Sub-IPC (2 min global ischemia/10 min reperfusion) did not limit infarct size (48.4 +/- 3.8%). Omapatrilat (10 micromol/L) or captopril (200 micromol/L) were administered alone or in conjunction with Sub-IPC. Reduced infarct size comparable to that observed with the full IPC protocol was seen when sub-IPC was combined with either omapatrilat (19.7 +/- 2.5%) or captopril (20.3 +/- 4.9%). Omapatrilat alone caused modest reduction of infarct size (34.6 +/- 1.5%, P < 0.01 v control), an effect not observed with captopril. Hoe140, a selective kinin B(2) receptor antagonist, eliminated the cardioprotective effect of omaptrilat alone or in combination with sub-IPC. We conclude that omapatrilat elicits cardioprotection via inhibition of bradykinin degradation and that dual inhibition of angiotensin-converting enzyme and neutral endopeptidase may have beneficial effects beyond standard angiotensin-converting enzyme inhibitor therapy in patients with acute coronary syndromes who are at risk of myocardial infarction.


Assuntos
Inibidores da Enzima Conversora de Angiotensina/uso terapêutico , Precondicionamento Isquêmico Miocárdico/métodos , Infarto do Miocárdio/tratamento farmacológico , Piridinas/uso terapêutico , Receptores da Bradicinina/efeitos dos fármacos , Tiazepinas/uso terapêutico , Animais , Peso Corporal/efeitos dos fármacos , Captopril/uso terapêutico , Masculino , Tamanho do Órgão/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley
10.
Cardiovasc Res ; 55(3): 483-94, 2002 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-12160945

RESUMO

Myocardial protection conferred by ischemic preconditioning occurs in a bimodal time course. The early cardioprotection wanes rapidly and is succeeded by a delayed phase of protection reducing infarct development, myocardial stunning and arrhythmias. This 'second window' of preconditioning may be evident for up to 72 h. The current mechanistic paradigm for delayed preconditioning against infarction invokes roles for several freely-diffusible molecules, generated during the preconditioning period, that act in autocrine and/or paracrine fashion as triggers of cellular adaptation. These include adenosine, nitric oxide, reactive oxygen species and bradykinin. A role for adenosine receptor activation as a proximal molecular mechanism leading to delayed preconditioning against infarction was established in 1994. Pharmacological adenosine receptor blockade during preconditioning abolishes the acquisition of delayed protection, while transient adenosine A(1) or A(3) receptor activation fully recapitulates protection against infarction (but not against stunning or arrhythmias) 24 h later. Although nitric oxide is a co-trigger of delayed preconditioning, A(1) agonist-induced delayed protection is independent of nitric oxide production. Adenosine receptor activation causes the activation of a complex protein kinase signalling cascade and, putatively, the subsequent activation of gene transcription. The induction or post-translational regulation of several proteins is associated with A(1) agonist-induced delayed protection. These include the mitochondrial manganese-conjugated superoxide dismutase, and the 27-kDa heat shock protein. Opening of K(ATP) channels during the index ischaemic event is an obligatory downstream event mediating A(1) and A(3) agonist induced delayed protection. However, the mechanism of sub-acute regulation of K(ATP) channels following adenosine receptor activation is unknown. Evidence for induction of inducible nitric oxide synthase as a distal mechanism of A(1) agonist-induced delayed protection is equivocal.


Assuntos
Adenosina/fisiologia , Precondicionamento Isquêmico Miocárdico , Isquemia Miocárdica/metabolismo , Miocárdio/metabolismo , Animais , Humanos , Miocárdio Atordoado , Receptores Purinérgicos P1/metabolismo , Transdução de Sinais/fisiologia , Fatores de Tempo
12.
J Cardiovasc Pharmacol ; 38(6): 930-9, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11707697

RESUMO

Apoptosis contributes to myocardial cell death during ischemia and reperfusion, especially during reperfusion. Growth factor "survival" signaling attenuates apoptosis. We therefore examined the effects of transforming growth factor-beta1 (TGF-beta1) on reperfusion injury and assessed the role of p42/p44 MAPK signaling in TGF-beta1-induced protection. Rat ventricular myocytes were subjected to hypoxia and reoxygenation. TGF-beta1 (0.2 ng/ml) was applied to cells during reoxygenation and the extent of apoptosis was determined by TUNEL and annexin V binding assays. Further studies were conducted in intact rat hearts subjected to regional ischemia and reperfusion. TGF-beta1 (0.2 ng/ml) was perfused during early reperfusion. In cells, incubation with TGF-beta1 (0.2 ng/ml) during reoxygenation attenuated the extent of cell membrane damage (trypan blue uptake) and also reduced the numbers of TUNEL-and annexin V-positive cells. Reduction of apoptosis was abrogated by PD98059 (5 microM), an inhibitor of p42/p44 MAPK activation. TGF-beta1 activated p42/p44 MAPK transiently in normoxic myocytes. When intact hearts received TGF-beta1 (0.2 ng/ml) during early reperfusion, infarct size was reduced from 39.4 +/- 3.1% to 17.3 +/- 3.1% (p < 0.01). This protective action of TGF-beta1 was abrogated by PD98059. These studies are the first to show that TGF-beta attenuates cardiac myocyte apoptosis during early reperfusion and limits infarct size through p42/p44 MAPK activation.


Assuntos
Proteína Quinase 1 Ativada por Mitógeno/fisiologia , Proteínas Quinases Ativadas por Mitógeno/fisiologia , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Miocárdio/enzimologia , Fator de Crescimento Transformador beta/uso terapêutico , Animais , Animais Recém-Nascidos , Apoptose/efeitos dos fármacos , Hipóxia Celular , Células Cultivadas , Inibidores Enzimáticos/farmacologia , Flavonoides/farmacologia , Marcação In Situ das Extremidades Cortadas , Masculino , Proteína Quinase 1 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 3 Ativada por Mitógeno , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Infarto do Miocárdio/patologia , Infarto do Miocárdio/prevenção & controle , Traumatismo por Reperfusão Miocárdica/patologia , Ratos , Ratos Sprague-Dawley , Fatores de Tempo , Fator de Crescimento Transformador beta/farmacologia , Fator de Crescimento Transformador beta1
13.
Am J Physiol Heart Circ Physiol ; 281(3): H1458-64, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11514319

RESUMO

Bradykinin is an important endogenous mediator exerting acute protective effects in the ischemic myocardium. The aims of this study were to investigate whether exogenously administered bradykinin could evoke delayed myocardial protection and to determine whether any protection observed might be dependent on nitric oxide (NO) generation. Conscious rats received bradykinin (40 microg/kg iv) or saline, preceded 15-20 min earlier by the NO synthase inhibitor N(omega)-nitro-L-arginine methyl ester (L-NAME, 10 mg/kg ip) or saline. Twenty-four hours later, hearts were Langendorff perfused and subjected to 35 min of regional ischemia and 120 min of reperfusion. Infarct size was assessed using tetrazolium staining and expressed as a percentage of the risk zone. Bradykinin pretreatment reduced the infarct-to-risk ratio from 53.5 +/- 3.2% to 29.1 +/- 4.7% (P < 0.01). The administration of L-NAME before bradykinin abrogated the delayed protection (infarct size 52.3 +/- 5.0%) but alone did not influence infarct size (53.5 +/- 4.8%). These results are the first to demonstrate that bradykinin can evoke a delayed ("second window") enhancement of myocardial tolerance to ischemia, an action that is dependent on the early generation of NO.


Assuntos
Bradicinina/administração & dosagem , Citoproteção/efeitos dos fármacos , Infarto do Miocárdio/prevenção & controle , Miocárdio/metabolismo , Óxido Nítrico/metabolismo , Animais , Velocidade do Fluxo Sanguíneo/efeitos dos fármacos , Peso Corporal/efeitos dos fármacos , Circulação Coronária , Doença das Coronárias/complicações , Esquema de Medicação , Hemodinâmica/efeitos dos fármacos , Técnicas In Vitro , Injeções Intraperitoneais , Injeções Intravenosas , Masculino , Contração Miocárdica/efeitos dos fármacos , Infarto do Miocárdio/etiologia , Infarto do Miocárdio/patologia , Miocárdio/patologia , NG-Nitroarginina Metil Éster/administração & dosagem , Tamanho do Órgão/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley
14.
Basic Res Cardiol ; 96(4): 329-44, 2001 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11518189

RESUMO

The protection conferred by ischemic preconditioning (PC) of myocardium occurs in a bimodal time course. The early cardioprotection wanes rapidly and is succeeded by a delayed phase of protection. This "second window" lasts for up to 72 hours, depending on species and end-point. A widely adopted paradigm for delayed PC is the following: 1) freely diffusible molecules or radicals, generated during the PC period, act in autocrine and/or paracrine manner as triggers of cellular adaptation; 2) they cause the activation of a protein kinase signal cascade; 3) the activated kinases phosphorylate important substrate proteins. In the case of delayed PC, it is thought that the phosphorylation of transcription factors, initiating the synthesis of late appearing effector proteins that promote cell survival during subsequent ischemia, may be a crucial event. Investigation of the proximal components of this sequence has altered our perceptions of several biological mediators, previously thought to be short acting, including adenosine, NO, free radicals and bradykinin. Signal transduction components include protein kinase C, tyrosine kinases and various mitogen- and stress-activated protein kinases but their patterns of regulation are complex and as yet poorly defined. Gene expression is modified in a regulated fashion to induce new proteins that promote cell repair and to protect against subsequent ischemia-reperfusion insult. It is likely that the complex nature of the preconditioning stimulus causes the activation of a variety of transcription factors, regulating a large number of target genes. So far, attention has focussed on a small number of protein products as potential distal mediators of delayed preconditioning. These include the heat shock proteins, manganese superoxide dismutase, inducible nitric oxide synthase, the ATP-sensitive potassium channel and cyclo-oxygenase-2.


Assuntos
Precondicionamento Isquêmico Miocárdico , Adaptação Fisiológica/fisiologia , Animais , Cardiologia/tendências , Infarto do Miocárdio/patologia , Fatores de Tempo
15.
J Cardiovasc Pharmacol ; 38(2): 278-87, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11483878

RESUMO

Transient adenosine A1 receptor (A1R) activation induces a second window or delayed preconditioning against myocardial infarction 24-72 h later. Early generation of nitric oxide and delayed induction of nitric oxide synthase have been implicated in mediating delayed cardioprotection after ischemic preconditioning in rabbits. Recent evidence indicates that some of the regulatory roles of adenosine in cardiac tissue may be mediated by A1R-induced generation of nitric oxide. This study examined the role of nitric oxide in the mediation of A1R-induced delayed preconditioning against infarction. Pharmacologic preconditioning of rabbits with the selective A1R agonist 2-chloro-N6-cyclopentyladenosine 100 microg/kg (CCPA) significantly reduced myocardial infarct size compared with control animals, after 30 min regional ischemia and 2 h reperfusion in vivo 24 h later (27.3+/-4.7 vs. 46.0+/-3.7%, respectively; p = 0.001). Nonselective inhibition of nitric oxide synthase with N(G)-nitro-L-arginine methyl ester (10 mg/kg) before administration of CCPA did not affect this infarct limitation at 24 h. Selective inhibition of inducible nitric oxide synthase before the prolonged ischemic insult on day 2, with two structurally independent inducible nitric oxide synthase inhibitors, L-N(6)-(1-iminoethyl)-lysine (10 mg/kg) or aminoguanidine (300 mg/kg), did not abrogate the reduction in infarction observed by pharmacologic preconditioning with CCPA 24 h earlier. These results suggest that the second window or delayed protection against myocardial infarction observed 24 h after pharmacologic preconditioning with an adenosine A1 agonist occurs independently of either early generation of nitric oxide or subacute induction of inducible nitric oxide synthase.


Assuntos
Adenosina/análogos & derivados , Precondicionamento Isquêmico Miocárdico , Óxido Nítrico Sintase/biossíntese , Óxido Nítrico/metabolismo , Receptores Purinérgicos P1/metabolismo , Adenosina/farmacologia , Animais , Indução Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Hemodinâmica/efeitos dos fármacos , Hemodinâmica/fisiologia , Precondicionamento Isquêmico Miocárdico/métodos , Masculino , Óxido Nítrico Sintase Tipo II , Coelhos , Receptores Purinérgicos P1/fisiologia
16.
Circulation ; 103(25): 3111-6, 2001 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-11425777

RESUMO

BACKGROUND: The sulfonylurea glibenclamide (Glib) abolishes the cardioprotective effect of ischemic preconditioning (IP), presumably by inhibiting mitochondrial K(ATP) channel opening in myocytes. Glimepiride (Glim) is a new sulfonylurea reported to affect nonpancreatic K(ATP) channels less than does Glib. We examined the effects of Glim on IP and on the protection afforded by diazoxide (Diaz), an opener of mitochondrial K(ATP) channels. METHODS AND RESULTS: Rat hearts were Langendorff-perfused, subjected to 35 minutes of regional ischemia and 120 minutes of reperfusion, and assigned to 1 of the following treatment groups: (1) control; (2) IP of 2x 5 minutes each of global ischemia before lethal ischemia; or pretreatment with (3) 30 micromol/L Diaz, (4) 10 micromol/L Glim, (5) 10 micromol/L Glib, (6) IP+Glim, (7) IP+Glib, (8) Diaz+Glim, or (9) Diaz+Glib. IP limited infarct size (18.5+/-1% vs 43.7+/-3% in control, P<0.01) as did Diaz (22.2+/-4.7%, P<0.01). The protective actions of IP or Diaz were not abolished by Glim (18.5+/-3% in IP+Glim, 22.3+/-3% in Diaz+Glim; P<0.01 vs control). However, Glib abolished the infarct-limiting effects of IP and Diaz. Patch-clamp studies in isolated rat ventricular myocytes confirmed that both Glim and Glib (each at 1 micromol/L) blocked sarcolemmal K(ATP) currents. However, in isolated cardiac mitochondria, Glim (10 micromol/L) failed to block the effects of K(ATP) opening by GTP, in contrast to the blockade caused by Glib. CONCLUSIONS: Although it blocks sarcolemmal currents in rat cardiac myocytes, Glim does not block the beneficial effects of mitochondrial K(ATP) channel opening in the isolated rat heart. These data may have significant implications for the treatment of type 2 diabetes in patients with ongoing ischemic heart disease.


Assuntos
Diazóxido/farmacologia , Hipoglicemiantes/farmacologia , Precondicionamento Isquêmico Miocárdico , Isquemia Miocárdica/prevenção & controle , Compostos de Sulfonilureia/farmacologia , Animais , Glibureto/farmacologia , Guanosina Trifosfato/farmacologia , Coração/efeitos dos fármacos , Coração/fisiopatologia , Ventrículos do Coração/citologia , Ventrículos do Coração/efeitos dos fármacos , Hemodinâmica/efeitos dos fármacos , Técnicas In Vitro , Membranas Intracelulares/efeitos dos fármacos , Membranas Intracelulares/fisiologia , Masculino , Potenciais da Membrana/efeitos dos fármacos , Proteínas de Membrana/efeitos dos fármacos , Mitocôndrias Cardíacas/efeitos dos fármacos , Mitocôndrias Cardíacas/fisiologia , Infarto do Miocárdio/etiologia , Infarto do Miocárdio/patologia , Infarto do Miocárdio/prevenção & controle , Isquemia Miocárdica/complicações , Técnicas de Patch-Clamp , Canais de Potássio , Ratos , Ratos Sprague-Dawley , Função Ventricular
17.
J Am Coll Cardiol ; 37(7): 1996-2001, 2001 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-11401144

RESUMO

OBJECTIVES: We assessed the effect of angiotensin-converting enzyme (ACE) inhibition in combination with a subthreshold preconditioning (PC) stimulus to elicit delayed preconditioning against infarction in pig myocardium. BACKGROUND: Bradykinin triggers early PC. Angiotensin-converting enzyme inhibitors increase local bradykinin levels via inhibition of kinin breakdown and have been shown in experimental studies to augment early protection afforded by PC. A role for bradykinin in eliciting delayed PC has not so far been identified. METHODS: We used a two-day protocol. On day 1 (closed chest), pigs were either sham-operated (group 1) or preconditioned, using balloon catheter inflation of the left anterior descending (LAD) coronary artery, with either a full (4 x 5 min PC, group 2) or subthreshold PC stimulus (2 x 2 min PC, group 3). Additional groups were pre-treated with perindoprilat (0.06 mg/kg i.v.) before sham (group 4) or subthreshold PC (group 5). On day 2 (open chest), all pigs were subjected to 40 min occlusion of the LAD followed by 3 h of reperfusion. Infarct size was determined by tetrazolium staining. RESULTS: Group 1 had a mean infarct size of 42.8+/-3.2% of the risk zone. Preconditioning with 4 x 5 min reduced the infarct size to 19.5+/-3.9% (p < 0.05). Groups 3 and 4 had infarct sizes not statistically different from group 1. However, combining perindoprilat with subthreshold PC resulted in a significant limitation of the infarction (18.4+/-3.1% p < 0.05), comparable with group 2. CONCLUSIONS: This is the first study to show that ACE inhibition can augment a mild ischemic stimulus to induce a protected state 24 h later.


Assuntos
Inibidores da Enzima Conversora de Angiotensina/farmacologia , Precondicionamento Isquêmico Miocárdico/métodos , Animais , Arritmias Cardíacas , Feminino , Masculino , Infarto do Miocárdio , Suínos , Fatores de Tempo
18.
Heart ; 85(3): 331-6, 2001 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11179278

RESUMO

OBJECTIVE: To determine whether exercise is capable of protecting the myocardium from experimental infarction and to explore the involvement of protein kinase C, a key signalling protein, in the development of any protection observed. METHODS: Rats were exercised on a treadmill for 30 minutes at 23-27 m/min. Sham treated animals were placed on the stationary treadmill but not exercised. Twenty four hours later, hearts were Langendorff perfused and subjected to 35 minute left main coronary artery occlusion followed by 120 minute reperfusion. Infarct size was determined by tetrazolium staining and expressed as a percentage of the risk zone (I/R%). To examine the potential signalling pathway, animals were treated with either the selective protein kinase C inhibitor chelerythrine, 5 mg/kg intraperitoneally, or with vehicle 10 minutes before the exercise or sham treadmill period. RESULTS: In the non-exercised group, mean (SEM) I/R was 48.4 (3.0)%. In the exercised group, infarct size was reduced to 17.3 (3.0)% (p < 0.01). Infarct size limitation induced by exercise was abolished by chelerythrine (I/R 45.0 (6.0)%). Chelerythrine pretreatment alone did not have any effect on infarct size (I/R 51.1 (3.9)%). Differences in infarct size were independent of risk zone size and myocardial contractile function during ischaemia-reperfusion. CONCLUSIONS: Experimental moderate exercise induces protection against myocardial infarction 24 hours later. Protein kinase C activation during exercise appears to be an important signal mediator of this protective response.


Assuntos
Infarto do Miocárdio/prevenção & controle , Traumatismo por Reperfusão Miocárdica/fisiopatologia , Esforço Físico/fisiologia , Proteína Quinase C/fisiologia , Alcaloides , Animais , Benzofenantridinas , Hemodinâmica , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Fenantridinas/farmacologia , Condicionamento Físico Animal , Proteína Quinase C/antagonistas & inibidores , Ratos , Transdução de Sinais
20.
Br J Pharmacol ; 130(2): 197-200, 2000 May.
Artigo em Inglês | MEDLINE | ID: mdl-10807653

RESUMO

Ischaemia-reperfusion injury causes cell death by both necrosis and apoptosis. Caspase activation is a major event in apoptosis. We therefore examined the effect of caspase inhibitors during reperfusion upon myocardial infarction. Rat isolated hearts were subjected to 35 min coronary occlusion and 120 min reperfusion. Treatment groups were perfused with caspase inhibitors during early reperfusion. We assessed a non-selective caspase inhibitor (Z-VAD. fmk, 0.1 microM), a caspase-8 inhibitor (Z-IETD.fmk, 0.07 microM), a caspase-9 inhibitor (Z-LEHD.fmk, 0.07 microM) and a caspase-3 inhibitor (Ac-DEVD.cmk, 0.07 microM). All caspase inhibitors limited infarct size (infarct-risk ratio per cent: control 38.5+/-2.6; Z-VAD. fmk 24.6+/-3.4; Z-LEHD.fmk 19.3+/-2.4; Z-IETD.fmk 23.0+/-5.4; Ac-DEVD.cmk 27.8+/-3.3; P<0.05 when compared with control value, 1-way ANOVA). We conclude that caspase inhibition during early reperfusion protects myocardium against lethal reperfusion injury.


Assuntos
Inibidores de Caspase , Inibidores de Cisteína Proteinase/uso terapêutico , Infarto do Miocárdio/prevenção & controle , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Oligopeptídeos/uso terapêutico , Clorometilcetonas de Aminoácidos/uso terapêutico , Animais , Técnicas In Vitro , Masculino , Infarto do Miocárdio/enzimologia , Traumatismo por Reperfusão Miocárdica/enzimologia , Ratos , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA