Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 182
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Biochem Biophys Res Commun ; 425(2): 230-6, 2012 Aug 24.
Artigo em Inglês | MEDLINE | ID: mdl-22828512

RESUMO

BACKGROUND: Apoptotic death of endothelial cells (EC) plays a crucial role for the development of ischemic injury. In the present study we investigated the impact of extracellular Adenosine-5'-triphosphate (ATP), either released from cells or exogenously added, on ischemia-induced apoptosis of human EC. METHODS AND RESULTS: To simulate ischemic conditions, cultured human umbilical vein endothelial cells (HUVEC) were exposed to 2 h of hypoxia (Po(2)<4mm Hg) in serum-free medium. Ischemia led to a 1.7-fold (+/-0.4; P<0.05) increase in EC apoptosis compared to normoxic controls as assessed by immunoblotting and immunocytochemistry of cleaved caspase-3. Ischemia-induced apoptosis was accompanied by a 2.3-fold (+/-0.5; P<0.05) increase of extracellular ATP detected by using a luciferin/luciferase assay. Addition of the soluble ecto-ATPase apyrase, enhancing ATP degradation, increased ischemia-induced caspase-3 cleavage. Correspondingly, inhibition of ATP breakdown by addition of the selective ecto-ATPase inhibitor ARL67156 significantly reduced ischemia-induced apoptosis. Extracellular ATP acts on membrane-bound P2Y- and P2X-receptors to induce intracellular signaling. Both, ATP and the P2Y-receptor agonist UTP significantly reduced ischemia-induced apoptosis in an equipotent manner, whereas the P2X-receptor agonist αß-me-ATP did not alter caspase-3 cleavage. The anti-apoptotic effects of ARL67156 and UTP were abrogated when P2-receptors were blocked by Suramin or PPADS. Furthermore, extracellular ATP led to an activation of MEK/ERK- and PI3K/Akt-signaling pathways. Accordingly, inhibition of MEK/ERK-signaling by UO126 or inhibition of PI3K/Akt-signaling by LY294002 abolished the anti-apoptotic effects of ATP. CONCLUSION: The data of the present study indicate that extracellular ATP counteracts ischemia-induced apoptosis of human EC by activating a P2Y-receptor-mediated signaling reducing caspase-3 cleavage.


Assuntos
Trifosfato de Adenosina/farmacologia , Apoptose/efeitos dos fármacos , Caspase 3/metabolismo , Citoproteção , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Isquemia/enzimologia , Agonistas do Receptor Purinérgico P2Y/farmacologia , Receptores Purinérgicos P2Y/metabolismo , Butadienos/farmacologia , Células Cultivadas , Cromonas/farmacologia , Inibidores Enzimáticos/farmacologia , Células Endoteliais da Veia Umbilical Humana/enzimologia , Células Endoteliais da Veia Umbilical Humana/patologia , Humanos , Isquemia/patologia , MAP Quinase Quinase Quinases/antagonistas & inibidores , MAP Quinase Quinase Quinases/metabolismo , Morfolinas/farmacologia , Nitrilas/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/metabolismo
2.
J Mol Cell Cardiol ; 53(1): 73-81, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22564366

RESUMO

AIMS: Since adenine nucleotide translocase 1 (ANT1) overexpression improved cardiac function in rats with activated renin-angiotensin system (RAS) and angiotensin II is known to enhance transforming growth factor ß (TGFß) signaling in cardiomyocytes, we assumed that ANT1 might modulate the classical TGFß/SMAD pathway. We therefore investigated whether the cardioprotective effect of ANT1 overexpression suppresses TGFß(1)-induced apoptosis, whether mitochondrial permeability transition pore (MPTP) regulation is involved, and SMAD signaling pathway is affected. METHODS AND RESULTS: Ventricular cardiomyocytes isolated from wild-type (WT) and ANT1 transgenic rats were treated with the apoptosis-inducing agent TGFß(1) (1 ng/ml). TGFß(1) treatment of WT cells enhanced the number of apoptotic cells by 31.8 ± 11.7% (p<0.01 vs. WT) measured by chromatin condensation. Apoptosis was blocked by 1µM cyclosporine A and by ANT1 overexpression. The protecting effect of ANT1 overexpression on TGFß(1)-induced apoptosis was verified by reduced caspase 3/7 activity and increased Bcl-2 expression. In addition, TGFß(1) decreased mitochondrial membrane potential as measured by JC-1 staining by 18.0 ± 3.7% in WT cardiomyocytes, but only by 7.2 ± 2.8% (p<0.05 vs. WT) in ANT1 cardiomyocytes. Cyclosporine A also attenuated the decline in mitochondrial membrane potential under TGFß(1) in WT cardiomyocytes. Determination of MPTP opening by Calcein assay in isolated cardiomyocytes and calcium retention assay in isolated mitochondria revealed a reduced open probability of MPTP after ANT1 overexpression. In addition to the effects of ANT1 on MPTP opening we investigated if ANT1 may interfere with the classical TGFß signaling pathway. Interestingly, ANT1-transgenic cardiomyocytes expressed less TGFß receptor II than WT cells. However, SMAD2 phosphorylation was already enhanced without TGFß(1) stimulation in these cells. Although no additional increase in SMAD2 phosphorylation was detectable after TGFß(1) treatment, SMAD signaling was still responsive to TGFß(1) indicated by an upregulation of SMAD7, a TGFß(1) target protein. CONCLUSION: Heart-specific overexpression of ANT1 leads to a reduced apoptotic response to TGFß(1) by preservation of the mitochondrial membrane potential, resistance to MPTP opening and altered TGFß signaling.


Assuntos
Apoptose/efeitos dos fármacos , Translocases Mitocondriais de ADP e ATP/genética , Proteínas de Transporte da Membrana Mitocondrial/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Fator de Crescimento Transformador beta1/farmacologia , Animais , Apoptose/genética , Células Cultivadas , Expressão Gênica , Masculino , Mitocôndrias Cardíacas/efeitos dos fármacos , Mitocôndrias Cardíacas/metabolismo , Poro de Transição de Permeabilidade Mitocondrial , Ratos , Ratos Sprague-Dawley , Transdução de Sinais , Transgenes
3.
Br J Pharmacol ; 165(1): 208-22, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21671901

RESUMO

BACKGROUND AND PURPOSE: Intermedin is a member of the calcitonin gene-related-peptide (CGRP) family expressed in endothelial cells and acts via calcitonin receptor-like receptors (CLRs). Here we have analysed the receptors for intermedin and its effect on the endothelial barrier in monolayers of human umbilical vein endothelial cells (HUVECs). EXPERIMENTAL APPROACH: We analysed the effect of intermedin on albumin permeability, contractile machinery, actin cytoskeleton and VE-cadherin in cultured HUVECs. KEY RESULTS: Intermedin concentration-dependently reduced basal endothelial permeability to albumin and antagonized thrombin-induced hyperpermeability. Intermedin was less potent (EC(50) 1.29 ± 0.12 nM) than adrenomedullin (EC(50) 0.24 ± 0.07 nM) in reducing endothelial permeability. These intermedin effects were inhibited by AM(22-52) and higher concentrations of αCGRP(8-37), with pA(2) values of αCGRP(8-37) of 6.4 for both intermedin and adrenomedullin. PCR data showed that HUVEC expressed only the CLR/RAMP2 receptor complex. Intermedin activated cAMP/PKA and cAMP/Epac signalling pathways. Intermedin's effect on permeability was blocked by inhibition of PKA but not of eNOS. Intermedin antagonized thrombin-induced contractile activation, RhoA activation and stress fibre formation. It also induced Rac1 activation, enhanced cell-cell adhesion and antagonized thrombin-induced loss of cell-cell adhesion. Treatment with a specific inhibitor of Rac1 prevented intermedin-mediated barrier stabilization. CONCLUSION AND IMPLICATIONS: Intermedin stabilized endothelial barriers in HUVEC monolayers via CLR/RAMP2 receptors. These effects were mediated via cAMP-mediated inactivation of contractility and strengthening of cell-cell adhesion. These findings identify intermedin as a barrier stabilizing agent and suggest intermedin as a potential treatment for vascular leakage in inflammatory conditions.


Assuntos
Células Endoteliais/efeitos dos fármacos , Células Endoteliais/fisiologia , Endotélio Vascular/fisiologia , Hormônios Peptídicos/farmacologia , Trombina/farmacologia , Junções Aderentes/efeitos dos fármacos , Junções Aderentes/fisiologia , Peptídeo Relacionado com Gene de Calcitonina/genética , Peptídeo Relacionado com Gene de Calcitonina/metabolismo , AMP Cíclico/genética , AMP Cíclico/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Cadeias Leves de Miosina/genética , Cadeias Leves de Miosina/metabolismo , RNA/genética , RNA/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Receptores de Adrenomedulina/genética , Receptores de Adrenomedulina/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Proteínas rac1 de Ligação ao GTP/genética , Proteínas rac1 de Ligação ao GTP/metabolismo , Proteína rhoA de Ligação ao GTP/genética , Proteína rhoA de Ligação ao GTP/metabolismo
4.
J Cell Physiol ; 224(1): 120-6, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20232299

RESUMO

Growth differentiation factor 15 (GDF15) is induced during heart failure development, and may influence different processes in cardiac remodeling. While its anti-apoptotic action under conditions of ischemia-reperfusion have been shown, it remained unclear if this is a broadly protective effect applicable to other apoptotic stimuli. Furthermore, effects on cardiac hypertrophy remained obscure. Therefore, we investigated the effects of GDF15 on induction of hypertrophy and apoptosis in ventricular cardiomyocytes. GDF15 (3 ng/ml) enhanced hypertrophic growth of cardiomyocytes as determined by an increase in cell size by 27 +/- 5% and rate of protein synthesis by 47 +/- 15%. In addition, a time and dose-dependent increase in SMAD-binding affinity was found, as well as enhanced phosphorylation of R-SMAD1. Inhibition of SMADs by transformation of cardiomyocytes with SMAD-decoy oligonucleotides abolished the hypertrophic growth effect. Specific inhibitors of PI3K (10 microM LY290042 or 10 nM wortmannin) or ERK (10 microM PD98059) also blocked GDF15-induced hypertrophy and SMAD activation. Apoptosis induction by three different agents, 100 nM angiotensin II, 1 ng/ml TGFbeta(1), or the NO-donor SNAP (100 microM) was blocked by addition of GDF15 (3 ng/ml). Scavenging of SMADs by transformation of cardiomyocytes with SMAD-decoy oligonucleotides abolished the anti-apoptotic effect of GDF15. In conclusion, GDF15 protects ventricular cardiomyocytes against different apoptotic stimuli and enhances hypertrophic growth. Hypertrophic signaling is thereby mediated via the kinases PI3K and ERK and the transcription factor R-SMAD1. Thus, GDF15 may influence cardiac remodeling via two different mechanisms, apoptosis protection and induction of hypertrophy.


Assuntos
Apoptose , Cardiomegalia/metabolismo , Cardiomegalia/patologia , Fator 15 de Diferenciação de Crescimento/metabolismo , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Transdução de Sinais , Fatores Etários , Angiotensina II/metabolismo , Animais , Apoptose/efeitos dos fármacos , Cardiomegalia/prevenção & controle , Células Cultivadas , MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Masculino , Miócitos Cardíacos/efeitos dos fármacos , Doadores de Óxido Nítrico/farmacologia , Oligonucleotídeos/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação , Inibidores de Proteínas Quinases/farmacologia , Ratos , Ratos Wistar , Transdução de Sinais/efeitos dos fármacos , Proteína Smad1/genética , Proteína Smad1/metabolismo , Fatores de Tempo , Fator de Crescimento Transformador beta1/metabolismo , Remodelação Ventricular
5.
Am J Physiol Cell Physiol ; 298(6): C1501-9, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20200209

RESUMO

Ischemia-induced apoptosis of endothelial cells may contribute to tissue injury, organ failure, and transplantation rejection. However, little is known about survival mechanisms capable to counteract endothelial apoptosis. This study investigated the potential role of an endogenous anti-apoptotic response elicited by transient hypoxia, capable to avert ongoing apoptosis in endothelial cells. Experiments were carried out in three different types of cultured endothelial cells (human umbilical vein, pig aorta, and from rat coronary microvasculature). As a pro-apoptotic challenge endothelial cells were cultured in serum-free medium and subjected to hypoxia for 2 h. We found that transient hypoxia reduced caspase 3 activation within 1 h of hypoxia. Accordingly, the number of apoptotic cells was reduced after 24 h of reoxygenation. This was true for all three cell types analyzed. Analysis of Akt and mitogen-activated protein kinase kinase (MEK)/extracellular signal-regulated kinase (ERK) pathways revealed that hypoxia induced a transient activation of ERK 2 but not of Akt. ERK 2 phosphorylation preceded the phosphorylation of pro-apoptotic molecule Bad at Ser112, an inhibitory phosphorylation site specific for ERK. The protective effects of hypoxia regarding Bad phosphorylation, caspase 3 activation, and apoptosis were abolished by MEK 1/2 inhibitors, PD98059 or UO126, as well as by antisense oligonucleotides directed against ERK 1/2. Furthermore, inhibition of this pathway inhibited hypoxia-induced increase in mitochondrial membrane potential. The present study demonstrates that transient hypoxia induces a novel survival mechanism that protects endothelial cells against apoptosis. This endogenous process involves MEK/ERK-mediated inhibition of the pro-apoptotic molecule Bad and caspase 3.


Assuntos
Apoptose , Células Endoteliais/enzimologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Animais , Apoptose/efeitos dos fármacos , Caspase 3/metabolismo , Hipóxia Celular , Sobrevivência Celular , Células Cultivadas , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/patologia , Ativação Enzimática , MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , MAP Quinases Reguladas por Sinal Extracelular/genética , Humanos , MAP Quinase Quinase Quinases/metabolismo , Potencial da Membrana Mitocondrial , Oligonucleotídeos Antissenso/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Ratos Wistar , Transdução de Sinais , Suínos , Fatores de Tempo , Proteína de Morte Celular Associada a bcl/metabolismo
6.
J Mol Cell Cardiol ; 48(1): 270-6, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19406125

RESUMO

Ischemic preconditioning has a powerful protective potential against a reperfusion-induced injury of the post-ischemic myocardium. Cardiomyocyte hypercontracture, i.e. excessive cell shortening, is an essential mechanism of the reperfusion-induced injury. Rigor contracture, i.e. Ca(2+)-independent contracture, has been shown to be an import component of the reperfusion-induced hypercontracture. Since rigor contracture is dependent on the rapidity of the metabolic recovery during reoxygenation, we hypothesized that preconditioning of the cardiomyocyte mitochondria may improve mitochondrial function to restore the energy balance during the initial phase of reoxygenation and may thus prevent rigor contracture. For this purpose adult rat cardiomyocytes were exposed to anoxia with subsequent reoxygenation. For preconditioning, cells were pre-treated with the mitochondrial ATP-sensitive K(+) channel opener diazoxide. Pre-treatment with 100 micromol/l diazoxide significantly reduced the reoxygenation-induced hypercontracture of cardiomyocytes due to an attenuation of the Ca(2+)-independent rigor-type contracture, which was accompanied by an acceleration of the phosphocreatine resynthesis during the initial phase of reoxygenation. Treatment with the mitochondrial ATP-sensitive K(+) channel antagonist 5-hydroxydecanoate (500 micromol/l) during preconditioning phase abolished these protective effects. Similarly, partial suppression of the mitochondrial function with 100 micromol/l NaCN during the reoxygenation phase abolished the diazoxide effects. Finally, in isolated rat hearts, preconditioning with diazoxide prior to global ischemia significantly improved left ventricular function and attenuated hypercontracture during reperfusion. This effect could be abolished by the treatment with 100 micromol/l NaCN during reperfusion. Taken together, pharmacological preconditioning of cardiomyocytes with diazoxide protects against the reoxygenation-induced rigor hypercontracture due to an improvement of the energy recovery at the onset of reoxygenation.


Assuntos
Diazóxido/farmacologia , Precondicionamento Isquêmico Miocárdico , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Animais , Ácidos Decanoicos/farmacologia , Hidroxiácidos/farmacologia , Hipóxia/fisiopatologia , Canais KATP/antagonistas & inibidores , Canais KATP/metabolismo , Masculino , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Mitocôndrias Cardíacas/efeitos dos fármacos , Mitocôndrias Cardíacas/metabolismo , Fosfocreatina/metabolismo , Ratos , Ratos Wistar , Cianeto de Sódio/farmacologia , Vasodilatadores/farmacologia
7.
Pflugers Arch ; 457(2): 485-91, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18581135

RESUMO

alpha-Adrenergic stimulation results in a positive adaptation of cardiomyocytes to increased cardiac work load by induction of hypertrophy and enhanced contraction. However, sustained adrenergic stimulation causes progression to heart failure. Under simultaneous activation of alpha- and beta-adrenoceptors by the naturally occurring catecholamine noradrenaline, beta1-stimulation inhibits alpha-adrenergic-stimulated hypertrophy. If beta-adrenergic stimulation may also influence cardiomyocyte contraction is not known yet. We now demonstrate that exposure of cardiomyocytes to noradrenaline or isoprenaline for 24 h results in a reduced cell shortening at low beating frequencies (0.5 Hz). At high beating frequencies (2 Hz), cell shortening was normal. beta-adrenergic stimulation enhances SERCA2A expression at the messenger RNA and protein level. This induction of the Ca(2+) pump SERCA2A by the transcription factor NFAT is responsible for maintenance of normal cell contraction at high beating frequencies since inhibition of NFAT by decoy-oligonucleotides impairs SERCA2A expression and cell shortening after beta-adrenergic stimulation. In conclusion, although reduced cell shortening is found under low beating frequencies, we demonstrate preservation of cardiomyocyte contraction at 2 Hz after exposure to beta-adrenergic stimuli, which indicate that adrenergic stimulation a priori does not cause impaired heart function. The increase of SERCA expression indicates an even improved Ca(2+) handling of the cells.


Assuntos
Agonistas alfa-Adrenérgicos/farmacologia , Agonistas Adrenérgicos beta/farmacologia , Cardiotônicos/farmacologia , Isoproterenol/farmacologia , Contração Miocárdica/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Norepinefrina/farmacologia , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/biossíntese , Animais , Cálcio/metabolismo , Estimulação Cardíaca Artificial , Células Cultivadas , Indução Enzimática , Frequência Cardíaca , Ventrículos do Coração/efeitos dos fármacos , Ventrículos do Coração/enzimologia , Masculino , Miócitos Cardíacos/enzimologia , Fatores de Transcrição NFATC/metabolismo , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/genética , Fatores de Tempo
9.
Dtsch Med Wochenschr ; 133(12): 586-90, 2008 Mar.
Artigo em Alemão | MEDLINE | ID: mdl-18335390

RESUMO

Rapid interventional restoration of coronary blood flow is the most effective therapy to limit infarct size in today}s cardiology. The early phase of reperfusion represents, however, a window of therapeutic opportunities largely unused in the clinic. Experimentally it has been clearly shown that the modalities of reperfusion have a substantial impact on infarct size, since reperfusion itself can damage the myocardium (reperfusion injury). The major cause for acute injury of the cardiomyocytes in reperfusion is their hypercontracture. Cytosolic Ca (2+) overload and malfunction of cell organelles, i. e. sarcoplasmic reticulum and mitochondria, determine the pathophysiology of reperfusion injury. The underlying mechanisms can be influenced in the first minutes of reperfusion by activation of protective signalling pathways (reperfusion therapy). First clinical studies confirm the efficacy of acute reperfusion therapy.


Assuntos
Traumatismo por Reperfusão Miocárdica/fisiopatologia , Reperfusão Miocárdica/métodos , Cálcio/metabolismo , Circulação Coronária , Humanos , Mitocôndrias Cardíacas/metabolismo , Transdução de Sinais , Resultado do Tratamento
10.
Acta Physiol (Oxf) ; 191(4): 267-74, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17825088

RESUMO

AIM: Angiotensin-converting enzyme (ACE) inhibitors like ramiprilat bind to ACE expressed on the cell surface of endothelial cells and induce cell-specific signalling including the activation of activator protein (AP)-1. The present study addressed the question whether ramiprilat exerts a similar effect on adult ventricular cardiomyocytes, i.e. activates the AP-1 or modifies contractile performance. It was further aimed to decide whether such effects depend on bradykinin receptors or whether they are directly mediated via ACE. METHODS: Adult rat ventricular cardiomyocytes were isolated and cultured. mRNA expression of ACE was investigated by RT-PCR, AP-1 activation by gel mobility shift assays, and cardiac contractile performance by electrical pacing of isolated cells and analysis of cell shortening via a line-camera. RESULTS: Cardiomyocytes stably express ACE. Ramiprilat increased maximal contraction velocity and shortened the time-to-peak of contraction. In contrast to effects evoked by bradykinin, such effects caused by ramiprilat were not attenuated by HOE 140, a bradykinin-receptor antagonist. These effects were also not attenuated in the presence of l-nitro-arginine, used to mimic bradykinin-dependent signalling. In cardiomyocytes, bradykinin but not ramiprilat activated AP-1. Ramiprilat activates AP-1 in endothelial cells that are known to respond to ramiprilat in this way. CONCLUSION: Ramiprilat exerts direct, bradykinin-receptor independent effects on cardiomyocytes that improve cellular function without a corresponding effect on AP-1 activation or induction of AP-1 dependent effects. This newly described effect of ramiprilat may contribute to the protective effects seen by application of ACE inhibitors.


Assuntos
Inibidores da Enzima Conversora de Angiotensina/administração & dosagem , Contração Miocárdica/fisiologia , Miócitos Cardíacos/fisiologia , Peptidil Dipeptidase A/metabolismo , Ramipril/análogos & derivados , Fator de Transcrição AP-1/metabolismo , Função Ventricular , Animais , Células Cultivadas , Relação Dose-Resposta a Droga , Ventrículos do Coração/citologia , Masculino , Contração Miocárdica/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Ramipril/administração & dosagem , Ratos , Ratos Wistar
11.
Proc Natl Acad Sci U S A ; 104(41): 16299-304, 2007 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-17901201

RESUMO

A comprehensive analysis of both the molecular genetic and phenotypic responses of any organism to the space flight environment has never been accomplished because of significant technological and logistical hurdles. Moreover, the effects of space flight on microbial pathogenicity and associated infectious disease risks have not been studied. The bacterial pathogen Salmonella typhimurium was grown aboard Space Shuttle mission STS-115 and compared with identical ground control cultures. Global microarray and proteomic analyses revealed that 167 transcripts and 73 proteins changed expression with the conserved RNA-binding protein Hfq identified as a likely global regulator involved in the response to this environment. Hfq involvement was confirmed with a ground-based microgravity culture model. Space flight samples exhibited enhanced virulence in a murine infection model and extracellular matrix accumulation consistent with a biofilm. Strategies to target Hfq and related regulators could potentially decrease infectious disease risks during space flight missions and provide novel therapeutic options on Earth.


Assuntos
Salmonella typhimurium/genética , Salmonella typhimurium/patogenicidade , Voo Espacial , Animais , Biofilmes/crescimento & desenvolvimento , Feminino , Expressão Gênica , Genes Bacterianos , Fator Proteico 1 do Hospedeiro/fisiologia , Ferro/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Análise de Sequência com Séries de Oligonucleotídeos , Proteômica , Regulon , Salmonelose Animal/etiologia , Salmonella typhimurium/fisiologia , Virulência , Simulação de Ausência de Peso
12.
Cardiovasc Res ; 74(3): 487-96, 2007 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-17408601

RESUMO

OBJECTIVES: Extracellular ATP stabilizes the endothelial barrier and inactivates the contractile machinery of endothelial cells. This inactivation relies on dephosphorylation of the regulatory myosin light chain (MLC) due to an activation of the MLC phosphatase (MLCP). To date, activation and function of MLCP in endothelial cells are only partially understood. METHODS: Here, the mechanism of extracellular ATP-mediated activation of MLCP was analyzed in human endothelial cells from umbilical veins. Cells were transfected with the endogenous protein phosphatase 1 (PP1)-specific inhibitor-2 (I-2). RESULTS: Overexpression of I-2 led to inhibition of PP1 activity and abrogation of the ATP-induced dephosphorylation of MLC. This indicates that the PP1 catalytic subunit is the principal phosphatase catalyzing the MLC dephosphorylation induced by extracellular ATP. As demonstrated by immunoprecipitation analysis, extracellular ATP recruits the PP1delta catalytic subunit and the myosin phosphatase targeting subunit (MYPT1) to form a complex. ATP stimulated dephosphorylation of MYPT1 at the inhibitory phosphorylation sites threonine 850 and 696. However, extracellular ATP failed to stimulate MYPT1 dephosphorylation in I-2-overexpressing cells. CONCLUSIONS: The present study shows for the first time that, in endothelial cells, extracellular ATP causes activation of MLCP through recruitment of PP1delta and MYPT1 into a MLCP holoenzyme complex and PP1-mediated reduction of the inhibitory phosphorylation of MYPT1.


Assuntos
Trifosfato de Adenosina/farmacologia , Células Endoteliais/enzimologia , Fosfatase de Miosina-de-Cadeia-Leve/metabolismo , Trifosfato de Adenosina/análogos & derivados , Amidas/farmacologia , Western Blotting , Células Cultivadas , Células Endoteliais/efeitos dos fármacos , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Humanos , Imunoprecipitação , Peptídeos e Proteínas de Sinalização Intracelular/antagonistas & inibidores , Toxinas Marinhas , Fosfatase de Miosina-de-Cadeia-Leve/antagonistas & inibidores , Nucleotidases/antagonistas & inibidores , Oxazóis/farmacologia , Fosfoproteínas Fosfatases/antagonistas & inibidores , Fosfoproteínas Fosfatases/metabolismo , Fosforilação , Proteína Fosfatase 1 , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas/genética , Proteínas/metabolismo , Antagonistas de Receptores Purinérgicos P1 , Piridinas/farmacologia , Teofilina/análogos & derivados , Teofilina/farmacologia , Trombina/farmacologia , Transfecção/métodos , Quinases Associadas a rho
13.
Pflugers Arch ; 454(1): 53-61, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17115225

RESUMO

Nitric oxide (NO) is increased under several pathophysiological, mainly inflammatory processes in the heart and has been characterized as an inducer of apoptosis in cardiomyocytes. The transcription factor activating protein-1 (AP-1) has been identified as a mediator of NO-induced apoptosis. Genes that are regulated by AP-1 under apoptotic conditions have not been identified yet. Therefore, we performed a microarray analysis with subsequent real-time polymerase chain reaction (PCR) to identify genes regulated by AP-1 in NO-induced ventricular cardiomyocytes of rats and tested the functional role of these genes in apoptosis. Cardiomyocytes were transformed with AP-1 decoy oligonucleotides for inhibition of AP-1 activity. These, as well as non-transformed control cells, were stimulated with the NO donor (+/-)-S-nitroso-N-acetylpenicillamine (SNAP, 100 microM) for 2 h. Some of the genes with differential gene expression on microarrays were further analysed by real-time PCR. Genes that are induced by SNAP were not identified. However, four genes, pyridoxal kinase, heat shock protein 10 (Hsp10), antigen identified by monoclonal antibodies 4F2 (4F2) and myosin light chain 2, were downregulated by SNAP in presence of AP-1. Pyridoxal kinase, Hsp10 and 4F2 have anti-apoptotic effects in unstimulated cells because downregulation of their expression by antisense oligos induced apoptosis in cardiomyocytes. An involvement of these genes in NO-induced apoptosis could only be proven for pyridoxal kinase. In conclusion, using microarray technology, we identified three anti-apoptotic genes (Hsp10, 4F2 and pyridoxal kinase) in ventricular cardiomyocytes, which may help the cells to resist some apoptotic stimuli. The downregulation of these genes results in cardiomyocyte apoptosis. Prevention of their downregulation may protect cardiomyocytes against apoptotic stimuli, and this may be of therapeutic benefit.


Assuntos
Apoptose/fisiologia , Chaperonina 10/genética , Proteína-1 Reguladora de Fusão/genética , Regulação da Expressão Gênica/fisiologia , Miócitos Cardíacos/fisiologia , Piridoxal Quinase/genética , Fator de Transcrição AP-1/fisiologia , Animais , Apoptose/genética , Sistemas Computacionais , Regulação para Baixo , Regulação da Expressão Gênica/efeitos dos fármacos , Ventrículos do Coração , Masculino , Análise em Microsséries , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Doadores de Óxido Nítrico/farmacologia , Oligonucleotídeos Antissenso/farmacologia , Reação em Cadeia da Polimerase/métodos , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , S-Nitroso-N-Acetilpenicilamina/farmacologia , Fator de Transcrição AP-1/antagonistas & inibidores
14.
J Mol Med (Berl) ; 84(11): 975-83, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16924465

RESUMO

Elevations in angiotensin II (AngII) and transforming growth factor (TGF-beta1) levels are often found under conditions leading to progression of heart failure. From several studies, it is evident that AngII enhances TGF-beta1 expression via activator protein 1 (AP-1) activation, and that this pathway is involved in hypertrophic growth of the heart muscle and in the development of cardiac fibrosis. We now continued characterization of the signaling pathway stimulated by AngII in ventricular cardiomyocytes of rat and analyzed if the enhancement of TGF-beta1 expression by AngII may also contribute to apoptosis induction, which is another predictor of heart failure progression. Stimulation of cardiomyocytes with 100 nM AngII for 2 h activated the transcription factors AP-1 and GATA by 68.6+/-23.9 or 70.7+/-9.8%. Induction of both factors was mediated by p38 mitogen-activated protein kinase (MAPK) because it was totally blocked using a specific inhibitor of the kinase (SB202190). When GATA was inhibited by transformation of cardiomyocytes with decoy oligonucleotides, AngII could not enhance TGF-beta1 expression. This inhibition was observed on the mRNA level in real-time polymerase chain reaction and on the protein level in Western blots. As a consequence, upon AngII stimulation for 24 h, release of TGF-beta1 from cardiomyocytes was also reduced from 240.5+/-50.4 to 130.5+/-22.1% (p<0.05). In contrast to the early induction of GATA and AP-1, the transcription factor similar to mothers against decapentaplegic homolog (SMAD) was induced by AngII after 24 h. This stimulation was dependent on TGF-beta1 because it was blocked by antibodies specific for TGF-beta1. Twenty-four hours after AngII addition, the number of apoptotic cardiomyocytes increased by 6.5+/-1.2%, and this apoptosis induction was blocked when SMAD activity was inhibited by transformation of cardiomyocytes with SMAD decoy oligonucleotides. In conclusion, the transcription factors AP-1 and GATA are activated by p38 MAPK upon AngII stimulation, and both are needed to enhance TGF-beta1 expression in ventricular cardiomyocytes. TGF-beta1 acts in an autocrine loop on the cells to induce apoptosis via SMAD signaling. Thus, the often-found correlation between AngII, TGF-beta1, AP-1, and SMAD in pathogenesis of heart disease reflects the proapoptotic signaling pathway induced by AngII in cardiomyocytes.


Assuntos
Angiotensina II/metabolismo , Apoptose , Comunicação Autócrina , Sistema de Sinalização das MAP Quinases , Miócitos Cardíacos/metabolismo , Fator de Crescimento Transformador beta1/biossíntese , Angiotensina II/farmacologia , Animais , Apoptose/efeitos dos fármacos , Comunicação Autócrina/efeitos dos fármacos , Western Blotting , Cardiomiopatias/metabolismo , Células Cultivadas , Fatores de Transcrição GATA/biossíntese , Ventrículos do Coração/citologia , Ventrículos do Coração/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Miócitos Cardíacos/efeitos dos fármacos , RNA Mensageiro/biossíntese , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteínas Smad/metabolismo , Fatores de Tempo , Fator de Transcrição AP-1/metabolismo , Fator de Crescimento Transformador beta1/genética , Regulação para Cima , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
15.
Basic Res Cardiol ; 100(4): 320-7, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15944810

RESUMO

Parathyroid hormone-related peptide (PTHrP) was found to improve contractile function of stunned myocardium in pigs. The peptide is released from coronary endothelial cells during ischemia and significantly improves post-ischemic recovery. The present study was aimed to decide whether such an induction of contractile responsiveness of the heart requires co-activation of adjacent cells or is a genuine phenomenon of cardiomyocytes. A second aim of this study was to decide whether such an improvement is linked to depressed cell function in general or oxidative inhibition. Isolated adult ventricular cardiomyocytes from rats were constantly paced at 0.5 Hz for 10 min. Cells were exposed to a brief oxidative inhibition by addition of 0.5 mmol/l potassium cyanide (KCN) in the presence of glucose. Under these conditions, cells stopped beating after 280 s on average. 30 s before they stopped to beat, cells had already developed a reduction in cell shortening, maximal relaxation and contraction velocity. In the co-presence of PTHrP (1-34) (100 nmol/l) cells continued to beat regular and did not develop reduced cell shortening, irrespectively of oxidative inhibition. In a second attempt, cells were exposed to the NO donor SNAP (100 micromol/l) or 8-bromocGMP (1 mmol/l). As expected both agents reduced cell shortening significantly. This reduction in cell shortening was attenuated in co-presence of PTHrP, too. Finally, we investigated the effect of PTHrP on cell shortening at different extracellular concentrations of calcium. Although, PTHrP increased intracellular calcium at 2 and 5 mmol/l extracellular calcium, respectively, it improved cell shortening only at 5 mmol/l extracellular calcium. Thus, the beneficial effect of PTHrP on cell shortening was independent from intracellular calcium but dependent on the steepness of the calcium gradient between intra- and extracellular calcium. In conclusion, our study shows that PTHrP is able to improve cell shortening rapidly and directly irrespectively of the reason for the reduced cell function. Improved electromechanical coupling rather than intracellular calcium handling seems to be the most important mechanism.


Assuntos
Contração Miocárdica/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Proteína Relacionada ao Hormônio Paratireóideo/farmacologia , Animais , Cálcio/metabolismo , GMP Cíclico/fisiologia , Masculino , Miócitos Cardíacos/fisiologia , Óxido Nítrico/fisiologia , Oxirredução , Cianeto de Potássio/farmacologia , Ratos , Ratos Wistar
16.
Cardiovasc Res ; 66(1): 123-31, 2005 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-15769455

RESUMO

OBJECTIVE: Reperfusion injury of the myocardium is characterised by development of cardiomyocyte hypercontracture. Previous studies have shown that cGMP-mediated stimuli protect against reperfusion injury, but the cellular mechanism is still unknown. METHODS: To simulate ischemia/reperfusion, adult rat cardiomyocytes were incubated anoxically (pH(o) 6.4) and then reoxygenated (pH(o) 7.4). Cytosolic calcium [Ca(2+)](i) (fura-2 ratio), pH(i) (BCECF ratio), cell length, and phospholamban phosphorylation were analysed. Under simulated ischemia cardiomyocytes develop [Ca(2+)](i) overload. When reoxygenated they rapidly undergo hypercontracture, triggered by oscillations of [Ca(2+)](i). We investigated whether cGMP-mediated stimuli can modulate [Ca(2+)](i) or pH(i) recovery and whether this contributes to their protective effect. Membrane-permeable cGMP analogues, 8-bromo-cGMP (1 mmol/L) or 8-pCPT-cGMP (10 micrommol/L), or a receptor-mediated activator of particulate guanylyl cyclase, urodilatin (1 micromol/L), were applied. RESULTS: The investigated stimuli protect against reoxygenation-induced hypercontracture (cell length as percent of end-ischemic length; control: 68+/-1.6; 8-bromo-cGMP: 88+/-1.5*; 8-pCPT-cGMP: 84+/-2.9*; urodilatin: 87+/-1.1*; n=24; *p<0.05). Recovery from [Ca(2+)](i) overload after 2 min reoxygenation [fura-2 ratio (a.u.); control: 1.43+/-0.15; 8-bromo-cGMP: 1.86+/-0.15*; 8-pCPT-cGMP: 1.92+/-0.19*; urodilatin: 1.93+/-0.24*; n=25; *p<0.05] was accelerated, and the frequency of [Ca(2+)](i) oscillations (min(-1)) was significantly reduced (control: 49+/-5.0 min(-1); 8-bromo-cGMP: 18+/-3.5* min(-1); 8-pCPT-cGMP: 18+/-4.5* min(-1); urodilatin: 16+/-4.1* min(-1); n=24; *p<0.05). cGMP-mediated stimuli increased sarcoplasmic Ca(2+) sequestration (caffeine-releasable Ca(2+) pool: 2-3 fold increase vs. control). Inhibition of sarcoplasmic Ca(2+)-ATPase (SERCA) by thapsigargin (150 nmol/L) or of protein kinase G with KT-5823 (1 micromol/L) abolished the effect of these stimuli on [Ca(2+)](i) recovery. The investigated stimuli significantly enhanced phospholamban phosphorylation. CONCLUSIONS: We conclude that cGMP-dependent signals activate SERCA via a protein kinase G-dependent phosphorylation of phospholamban. The increase in SERCA activity seems to reduce peak [Ca(2+)](i) and [Ca(2+)](i) oscillation during reoxygenation and to attenuate the excessive activation of the contractile machinery that otherwise leads to the development of hypercontracture.


Assuntos
Cálcio/metabolismo , GMP Cíclico/análogos & derivados , GMP Cíclico/metabolismo , GMP Cíclico/farmacologia , Traumatismo por Reperfusão Miocárdica/metabolismo , Miócitos Cardíacos/metabolismo , Animais , ATPases Transportadoras de Cálcio/antagonistas & inibidores , ATPases Transportadoras de Cálcio/metabolismo , Carbazóis/farmacologia , Tamanho Celular/efeitos dos fármacos , Proteínas Quinases Dependentes de GMP Cíclico/antagonistas & inibidores , Citosol/metabolismo , Concentração de Íons de Hidrogênio , Indóis/farmacologia , Masculino , Microscopia de Fluorescência , Miócitos Cardíacos/efeitos dos fármacos , Ratos , Ratos Wistar , Retículo Sarcoplasmático/metabolismo , Tapsigargina/farmacologia
17.
Acta Physiol Scand ; 183(2): 161-9, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15676057

RESUMO

AIMS: Endothelin-1 (ET-1) promotes endothelial cell growth. Endothelial cell proliferation involves the activation of Ca2+-activated K+ channels. In this study, we investigated whether Ca2+-activated K+ channels with big conductance (BK(Ca)) contribute to endothelial cell proliferation induced by ET-1. METHODS: The patch-clamp technique was used to analyse BK(Ca) activity in endothelial cells derived from human umbilical cord veins (HUVEC). Endothelial proliferation was examined using cell counts and measuring [3H]-thymidine incorporation. Changes of intracellular Ca2+ levels were examined using fura-2 fluorescence imaging. RESULTS: Characteristic BK(Ca) were identified in cultured HUVEC. Continuous perfusion of HUVEC with 10 nmol L(-1) ET-1 caused a significant increase of BK(Ca) open-state probability (n = 14; P < 0.05; cell-attached patches). The ET(B)-receptor antagonist (BQ-788, 1 micromol L(-1)) blocked this effect. Stimulation with Et-1 (10 nmol L(-1)) significantly increased cell growth by 69% (n = 12; P < 0.05). In contrast, the combination of ET-1 (10 nmol L(-1)) and the highly specific BK(Ca) blocker iberiotoxin (IBX; 100 nmol L(-1)) did not cause a significant increase in endothelial cell growth. Ca2+ dependency of ET-1-induced proliferation was tested using the intracellular Ca2+-chelator BAPTA (10 micromol L(-1)). BAPTA abolished ET-1 induced proliferation (n = 12; P < 0.01). In addition, ET-1-induced HUVEC growth was significantly reduced, if cells were kept in a Ca2+-reduced solution (0.3 mmol L(-1)), or by the application of 2 aminoethoxdiphenyl borate (100 micromol L(-1)) which blocks hyperpolarization-induced Ca2+ entry (n = 12; P < 0.05). CONCLUSION: Activation of BK(Ca) by ET-1 requires ET(B)-receptor activation and induces a capacitative Ca2+ influx which plays an important role in ET-1-mediated endothelial cell proliferation.


Assuntos
Ácido Egtázico/análogos & derivados , Células Endoteliais/fisiologia , Endotelina-1/fisiologia , Canais de Potássio Cálcio-Ativados/fisiologia , Cálcio/metabolismo , Cálcio/fisiologia , Contagem de Células , Divisão Celular/efeitos dos fármacos , Divisão Celular/fisiologia , Células Cultivadas , Quelantes/farmacologia , Meios de Cultura , Relação Dose-Resposta a Droga , Ácido Egtázico/farmacologia , Condutividade Elétrica , Células Endoteliais/efeitos dos fármacos , Antagonistas do Receptor de Endotelina B , Humanos , Potenciais da Membrana/fisiologia , Oligopeptídeos , Peptídeos/farmacologia , Piperidinas , Canais de Potássio Cálcio-Ativados/antagonistas & inibidores
18.
Am J Physiol Cell Physiol ; 287(5): C1246-55, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15475517

RESUMO

cAMP-mediated signaling mechanisms may destabilize or stabilize the endothelial barrier, depending on the origin of endothelial cells. Here, microvascular coronary [coronary endothelial cells (CEC)] and macrovascular aortic endothelial cell (AEC) monolayers with opposite responses to cAMP were analyzed. Macromolecule permeability, isometric force, activation state of contractile machinery [indicated by phosphorylation of regulatory myosin light chains (MLC), activity of MLC kinase, and MLC phosphatase], and dynamic changes of adhesion complex proteins (translocation of VE-cadherin and paxillin) were determined. cAMP signaling was stimulated by the adenosine receptor agonist 5'-N-(ethylcarboxamido)-adenosine (NECA), the beta-adrenoceptor agonist isoproterenol (Iso), or by the adenylyl cyclase activator forskolin (FSK). Permeability was increased in CEC and decreased in AEC on stimulation with NECA, Iso, or FSK. The effects could be inhibited by the PKA inhibitor Rp-8-CPT-cAMPS and imitated by the PKA activator Sp-cAMPS. Under cAMP/PKA-dependent stimulation, isometric force and MLC phosphorylation were reduced in monolayers of either cell type, due to an activation of MLC phosphatase. In CEC but not in AEC, FSK induced delocalization of VE-cadherin and paxillin from cellular adhesion complexes as indicated by cell fractionation and immunofluorescence microscopy. In conclusion, decline in contractile activation and isometric force contribute to cAMP/PKA-mediated stabilization of barrier function in AEC. In CEC, this stabilizing effect is overruled by cAMP-induced disintegration of cell adhesion structures.


Assuntos
AMP Cíclico/metabolismo , Células Endoteliais/metabolismo , Endotélio Vascular/metabolismo , Transdução de Sinais/fisiologia , Animais , Aorta/citologia , Aorta/fisiologia , Adesão Celular/fisiologia , Células Cultivadas , Vasos Coronários/citologia , Vasos Coronários/fisiologia , Proteínas Quinases Dependentes de AMP Cíclico , Células Endoteliais/ultraestrutura , Endotélio Vascular/ultraestrutura , Ativação Enzimática/fisiologia , Junções Intercelulares/ultraestrutura , Masculino , Microscopia de Fluorescência , Cadeias Leves de Miosina/metabolismo , Quinase de Cadeia Leve de Miosina/metabolismo , Fosfatase de Miosina-de-Cadeia-Leve/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Ratos , Ratos Wistar , Suínos
19.
Am J Physiol Cell Physiol ; 286(4): C807-12, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15001425

RESUMO

Phosphorylation of endothelial myosin light chains (MLC) is a key mechanism in control of endothelial contractile machinery. Extracellular ATP influences endothelial MLC phosphorylation by either activation of Ca(2+)-dependent MLC kinase or Ca(2+)-independent MLC phosphatase. Here, the role of the MEK/MAPK pathway in this signaling was investigated in porcine aortic endothelial cells. Phosphorylation of ERK2 and phosphorylation of MLC were analyzed in cultured aortic endothelial cells. ATP (10 microM) increased ERK2 phosphorylation from basal 17 +/- 3 to 53 +/- 4%, an effect suppressed in the presence of the MEK inhibitors PD-98059 (20 microM) or U0126 (10 microM). Phosphorylation of ERK2 was not dependent on the ATP-induced cytosolic Ca(2+) rise, because it was unaltered when this was suppressed by the Ca(2+) chelator BAPTA (10 microM) or xestospongin C (3 microM), an inhibitor of the inositol 1,4,5-trisphosphate-sensitive Ca(2+) release mechanism of the endoplasmic reticulum. Phosphorylation of ERK2 was neither induced by the adenosine analog 5'-(N-ethylcarboxamido)adenosine (1 microM) nor inhibited in the presence of the adenosine receptor antagonist 8-phenyltheophylline (10 microM). ATP increased MLC kinase activity, and this was blocked in presence of PD-98059. ATP also increased MLC phosphatase activity, which was not inhibited by PD-98059. The MEK/MAPK pathway is a Ca(2+)-independent part of ATP signaling toward MLC kinase but not of ATP signaling toward MLC phosphatase.


Assuntos
Trifosfato de Adenosina/metabolismo , Endotélio Vascular/enzimologia , Sistema de Sinalização das MAP Quinases/fisiologia , Cadeias Leves de Miosina/metabolismo , Animais , Aorta/citologia , Cálcio/metabolismo , Células Cultivadas , Endotélio Vascular/citologia , Inibidores Enzimáticos/farmacologia , Flavonoides/farmacologia , Proteína Quinase 1 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Quinase de Cadeia Leve de Miosina/metabolismo , Fosfatase de Miosina-de-Cadeia-Leve/metabolismo , Fosforilação , Suínos
20.
Cardiovasc Res ; 61(3): 365-71, 2004 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-14962469

RESUMO

During the past decade, the understanding has grown that control of the conditions of reperfusion is critical for salvaging ischemic-reperfused myocardium. The first few minutes of reperfusion constitute a critical phase, as here lethal tissue injury in addition to that already developed during ischemia may be initiated. The identification of the mechanisms of reperfusion-induced cell death opens a new window of opportunity for cardioprotection in the clinic. Development of cardiomyocyte hypercontracture is a predominant feature of reperfusion injury. We and others have shown that control of hypercontracture in reperfusion reduces the extent of tissue injury. On the cellular level, it was shown that reperfusion-induced hypercontracture might either originate from a rigor-type mechanism, when energy recovery proceeds very slowly, or from Ca2+ overload, when energy recovery is rapid but cytosolic Ca2+ load is high. These two mechanisms can be influenced by various interventions that either connect with cytosolic Ca2+ control or myofibrillar Ca2+ sensitivity or with mitochondrial energy production. These experimental approaches will hopefully lead to novel strategies for clinical cardioprotection during the early phase of reperfusion.


Assuntos
Infarto do Miocárdio/terapia , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Cálcio/metabolismo , Humanos , Contração Miocárdica , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/patologia , Miocárdio/metabolismo , Miocárdio/patologia , Necrose , Sódio/metabolismo , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...