Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Pharmacol Rev ; 75(1): 159-216, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36753049

RESUMEN

Preconditioning, postconditioning, and remote conditioning of the myocardium enhance the ability of the heart to withstand a prolonged ischemia/reperfusion insult and the potential to provide novel therapeutic paradigms for cardioprotection. While many signaling pathways leading to endogenous cardioprotection have been elucidated in experimental studies over the past 30 years, no cardioprotective drug is on the market yet for that indication. One likely major reason for this failure to translate cardioprotection into patient benefit is the lack of rigorous and systematic preclinical evaluation of promising cardioprotective therapies prior to their clinical evaluation, since ischemic heart disease in humans is a complex disorder caused by or associated with cardiovascular risk factors and comorbidities. These risk factors and comorbidities induce fundamental alterations in cellular signaling cascades that affect the development of ischemia/reperfusion injury and responses to cardioprotective interventions. Moreover, some of the medications used to treat these comorbidities may impact on cardioprotection by again modifying cellular signaling pathways. The aim of this article is to review the recent evidence that cardiovascular risk factors as well as comorbidities and their medications may modify the response to cardioprotective interventions. We emphasize the critical need for taking into account the presence of cardiovascular risk factors as well as comorbidities and their concomitant medications when designing preclinical studies for the identification and validation of cardioprotective drug targets and clinical studies. This will hopefully maximize the success rate of developing rational approaches to effective cardioprotective therapies for the majority of patients with multiple comorbidities. SIGNIFICANCE STATEMENT: Ischemic heart disease is a major cause of mortality; however, there are still no cardioprotective drugs on the market. Most studies on cardioprotection have been undertaken in animal models of ischemia/reperfusion in the absence of comorbidities; however, ischemic heart disease develops with other systemic disorders (e.g., hypertension, hyperlipidemia, diabetes, atherosclerosis). Here we focus on the preclinical and clinical evidence showing how these comorbidities and their routine medications affect ischemia/reperfusion injury and interfere with cardioprotective strategies.


Asunto(s)
Poscondicionamiento Isquémico , Precondicionamiento Isquémico Miocárdico , Isquemia Miocárdica , Daño por Reperfusión Miocárdica , Animales , Humanos , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Daño por Reperfusión Miocárdica/prevención & control , Daño por Reperfusión Miocárdica/metabolismo , Isquemia Miocárdica/tratamiento farmacológico , Isquemia Miocárdica/prevención & control , Factores de Riesgo , Factores de Riesgo de Enfermedad Cardiaca , Isquemia
2.
Basic Res Cardiol ; 113(1): 6, 2018 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-29242986

RESUMEN

Conditioning-like infarct limitation by enhanced level of hydrogen sulfide (H2S) has been demonstrated in many animal models of myocardial ischemia/reperfusion injury (MIRI) in vivo. We sought to evaluate the effect of H2S on myocardial infarction across in vivo pre-clinical studies of MIRI using a comprehensive systematic review followed by meta-analysis. Embase, Pubmed and Web of Science were searched for pre-clinical investigation of the effect of H2S on MIRI in vivo. Retained records (6031) were subjected to our pre-defined inclusion criteria then were objectively critiqued. Thirty-two reports were considered eligible to be included in this study and were grouped, based on the time of H2S application, into preconditioning and postconditioning groups. Data were pooled using random effect meta-analysis. We also investigated the possible impact of different experimental variables and the risk of bias on the observed effect size. Preconditioning with H2S (n = 23) caused a significant infarct limitation of - 20.25% (95% CI - 25.02, - 15.47). Similarly, postconditioning with H2S (n = 40) also limited infarct size by - 21.61% (95% CI - 24.17, - 19.05). This cardioprotection was also robust and consistent following sensitivity analyses where none of the pre-defined experimental variables had a significant effect on the observed infarct limitation. H2S shows a significant infarct limitation across in vivo pre-clinical studies of MIRI which include data from 825 animals. This infarct-sparing effect is robust and consistent when H2S is applied before ischemia or at reperfusion, independently on animal size or sulfide source. Validating this infarct limitation using large animals from standard medical therapy background and with co-morbidities should be the way forward.


Asunto(s)
Sulfuro de Hidrógeno , Poscondicionamiento Isquémico , Precondicionamiento Isquémico Miocárdico , Animales
3.
Cell Tissue Res ; 369(3): 567-578, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28451751

RESUMEN

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.


Asunto(s)
Guanilato Ciclasa/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Péptido Natriurético Tipo-C/farmacología , Somatotrofos/metabolismo , Animales , Línea Celular , GMP Cíclico/metabolismo , Humanos , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Fosforilación/efectos de los fármacos , Regiones Promotoras Genéticas/genética , Receptores Acoplados a la Guanilato-Ciclasa/metabolismo , Somatotrofos/efectos de los fármacos
4.
Pharmacol Rev ; 66(4): 1142-74, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25261534

RESUMEN

Pre-, post-, and remote conditioning of the myocardium are well described adaptive responses that markedly enhance the ability of the heart to withstand a prolonged ischemia/reperfusion insult and provide therapeutic paradigms for cardioprotection. Nevertheless, more than 25 years after the discovery of ischemic preconditioning, we still do not have established cardioprotective drugs on the market. Most experimental studies on cardioprotection are still undertaken in animal models, in which ischemia/reperfusion is imposed in the absence of cardiovascular risk factors. However, ischemic heart disease in humans is a complex disorder caused by, or associated with, cardiovascular risk factors and comorbidities, including hypertension, hyperlipidemia, diabetes, insulin resistance, heart failure, altered coronary circulation, and aging. These risk factors induce fundamental alterations in cellular signaling cascades that affect the development of ischemia/reperfusion injury per se and responses to cardioprotective interventions. Moreover, some of the medications used to treat these risk factors, including statins, nitrates, and antidiabetic drugs, may impact cardioprotection by modifying cellular signaling. The aim of this article is to review the recent evidence that cardiovascular risk factors and their medication may modify the response to cardioprotective interventions. We emphasize the critical need to take into account the presence of cardiovascular risk factors and concomitant medications when designing preclinical studies for the identification and validation of cardioprotective drug targets and clinical studies. This will hopefully maximize the success rate of developing rational approaches to effective cardioprotective therapies for the majority of patients with multiple risk factors.


Asunto(s)
Cardiotónicos/farmacología , Enfermedades Cardiovasculares/prevención & control , Daño por Reperfusión Miocárdica/prevención & control , Animales , Diseño de Fármacos , Humanos , Poscondicionamiento Isquémico/métodos , Precondicionamiento Isquémico Miocárdico/métodos , Terapia Molecular Dirigida , Factores de Riesgo , Transducción de Señal/efectos de los fármacos
5.
Basic Res Cardiol ; 111(2): 23, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26912064

RESUMEN

Unmodified reperfusion therapy for acute myocardial infarction (AMI) is associated with irreversible myocardial injury beyond that sustained during ischemia. Studies in experimental models of ischemia/reperfusion and in humans undergoing reperfusion therapy for AMI have examined potential beneficial effects of nitric oxide (NO) supplemented at the time of reperfusion. Using a rigorous systematic search approach, we have identified and critically evaluated all the relevant experimental and clinical literature to assess whether exogenous NO given at reperfusion can limit infarct size. An inclusive search strategy was undertaken to identify all in vivo experimental animal and clinical human studies published in the period 1990-2014 where NO gas, nitrite, nitrate or NO donors were given to ameliorate reperfusion injury. Articles were screened at title and subsequently at abstract level, followed by objective full text analysis using a critical appraisal tool. In twenty-one animal studies, all NO treatments except nitroglycerin afforded protection against measures of reperfusion injury, including infarct size, creatinine kinase release, neutrophil accumulation and cardiac dysfunction. In three human AMI RCT's, there was no consistent evidence of infarct limitation associated with NO treatment as an adjunct to reperfusion. Despite experimental evidence that most NO treatments can reduce infarct size when given as adjuncts to reperfusion, the value of these interventions in clinical AMI is unproven. Our study raises issues for the design of further clinical studies and emphasises the need for improved design of animal studies to reflect more accurately the comorbidities and other confounding factors seen in clinical AMI.


Asunto(s)
Infarto del Miocardio/terapia , Reperfusión Miocárdica , Óxido Nítrico/uso terapéutico , Animales , Humanos
6.
Pharmacol Res ; 111: 442-451, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27378570

RESUMEN

Exogenous hydrogen sulfide (H2S) protects against myocardial ischemia/reperfusion injury but the mechanism of action is unclear. The present study investigated the effect of GYY4137, a slow-releasing H2S donor, on myocardial infarction given specifically at reperfusion and the signalling pathway involved. Thiobutabarbital-anesthetised rats were subjected to 30min of left coronary artery occlusion and 2h reperfusion. Infarct size was assessed by tetrazolium staining. In the first study, animals randomly received either no treatment or GYY4137 (26.6, 133 or 266µmolkg(-1)) by intravenous injection 10min before reperfusion. In a second series, involvement of PI3K and NO signalling were interrogated by concomitant administration of LY294002 or L-NAME respectively and the effects on the phosphorylation of Akt, eNOS, GSK-3ß and ERK1/2 during early reperfusion were assessed by immunoblotting. GYY4137 266µmolkg(-1) significantly limited infarct size by 47% compared to control hearts (P<0.01). In GYY4137-treated hearts, phosphorylation of Akt, eNOS and GSK-3ß was increased 2.8, 2.2 and 2.2 fold respectively at early reperfusion. Co-administration of L-NAME and GYY4137 attenuated the cardioprotection afforded by GYY4137, associated with attenuated phosphorylation of eNOS. LY294002 totally abrogated the infarct-limiting effect of GYY4137 and inhibited Akt, eNOS and GSK-3ß phosphorylation. These data are the first to demonstrate that GYY4137 protects the heart against lethal reperfusion injury through activation of PI3K/Akt signalling, with partial dependency on NO signalling and inhibition of GSK-3ß during early reperfusion. H2S-based therapeutic approaches may have value as adjuncts to reperfusion in the treatment of acute myocardial infarction.


Asunto(s)
Sulfuro de Hidrógeno/metabolismo , Morfolinas/farmacología , Infarto del Miocardio/prevención & control , Daño por Reperfusión Miocárdica/prevención & control , Miocardio/metabolismo , Compuestos Organotiofosforados/farmacología , Sustancias Protectoras/farmacología , Animales , Citoprotección , Modelos Animales de Enfermedad , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Hemodinámica/efectos de los fármacos , Masculino , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , Daño por Reperfusión Miocárdica/fisiopatología , Miocardio/patología , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Fosfatidilinositol 3-Quinasa/metabolismo , Fosforilación , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Factores de Tiempo
7.
Basic Res Cardiol ; 109(2): 402, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24477916

RESUMEN

B-type natriuretic peptide (BNP) and C-type natriuretic peptide (CNP), and (Cys-18)-atrial natriuretic factor (4-23) amide (C-ANF), are cytoprotective under conditions of ischemia-reperfusion, limiting infarct size. ATP-sensitive K(+) channel (KATP) opening is also cardioprotective, and although the KATP activation is implicated in the regulation of cardiac natriuretic peptide release, no studies have directly examined the effects of natriuretic peptides on cardiac KATP activity. Normoxic cardiomyocytes were patch clamped in the cell-attached configuration to examine sarcolemmal KATP (sKATP) activity. The KATP opener pinacidil (200 µM) increased the open probability of the patch (NPo; values normalized to control) at least twofold above basal value, and this effect was abolished by HMR1098 10 µM, a selective KATP blocker (5.23 ± 1.20 versus 0.89 ± 0.18; P < 0.001). We then examined the effects of BNP, CNP, C-ANF and 8Br-cGMP on the sKATP current. Bath application of BNP (≥10 nM) or CNP (≥0.01 nM) suppressed basal NPo (BNP: 1.00 versus 0.56 ± 0.09 at 10 nM, P < 0.001; CNP: 1.0 versus 0.45 ± 0.16, at 0.01 nM, P < 0.05) and also abolished the pinacidil-activated current at concentrations ≥10 nM. C-ANF (≥10 nM) enhanced KATP activity (1.00 versus 3.85 ± 1.13, at 100 nM, P < 0.05). The cGMP analog 8Br-cGMP 10 nM dampened the pinacidil-activated current (2.92 ± 0.60 versus 1.53 ± 0.32; P < 0.05). Natriuretic peptides modulate sKATP current in ventricular cardiomyocytes. This may be at least partially associated with their ability to augment intracellular cGMP concentrations via NPR-A/B, or their ability to bind NPR-C with high affinity. Although the mechanism of modulation requires elucidation, these preliminary data give new insights into the relationship between natriuretic peptide signaling and sKATP in the myocardium.


Asunto(s)
Canales KATP/fisiología , Miocitos Cardíacos/fisiología , Péptido Natriurético Encefálico/metabolismo , Péptido Natriurético Tipo-C/metabolismo , Animales , Ventrículos Cardíacos/citología , Activación del Canal Iónico/efectos de los fármacos , Activación del Canal Iónico/fisiología , Canales KATP/genética , Canales KATP/metabolismo , Masculino , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Miocitos Cardíacos/metabolismo , Péptido Natriurético Encefálico/farmacología , Péptido Natriurético Tipo-C/farmacología , Técnicas de Placa-Clamp , Cloruro de Potasio/farmacología , Ratas , Ratas Sprague-Dawley
8.
Cell Tissue Res ; 355(2): 425-36, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24352806

RESUMEN

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.


Asunto(s)
Lactotrofos/enzimología , Receptores del Factor Natriurético Atrial/metabolismo , Transducción de Señal , Animales , Factor Natriurético Atrial/farmacología , Señalización del Calcio/efectos de los fármacos , Línea Celular , AMP Cíclico/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/genética , Endocitosis/efectos de los fármacos , Lactotrofos/efectos de los fármacos , Ligandos , Ratones , Péptido Natriurético Tipo-C/farmacología , Proteína Quinasa C/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Transducción de Señal/efectos de los fármacos , Esfingolípidos/metabolismo , Hormona Liberadora de Tirotropina/metabolismo
9.
Cardiovasc Res ; 119(6): 1336-1351, 2023 06 13.
Artículo en Inglés | MEDLINE | ID: mdl-36718529

RESUMEN

AIMS: Remote ischaemic preconditioning (RIPC) is a robust cardioprotective intervention in preclinical studies. To establish a working and efficacious RIPC protocol in our laboratories, we performed randomized, blinded in vivo studies in three study centres in rats, with various RIPC protocols. To verify that our experimental settings are in good alignment with in vivo rat studies showing cardioprotection by limb RIPC, we performed a systematic review and meta-analysis. In addition, we investigated the importance of different study parameters. METHODS AND RESULTS: Male Wistar rats were subjected to 20-45 min cardiac ischaemia followed by 120 min reperfusion with or without preceding RIPC by 3 or 4 × 5-5 min occlusion/reperfusion of one or two femoral vessels by clamping, tourniquet, or pressure cuff. RIPC did not reduce infarct size (IS), microvascular obstruction, or arrhythmias at any study centres. Systematic review and meta-analysis focusing on in vivo rat models of myocardial ischaemia/reperfusion injury with limb RIPC showed that RIPC reduces IS by 21.28% on average. In addition, the systematic review showed methodological heterogeneity and insufficient reporting of study parameters in a high proportion of studies. CONCLUSION: We report for the first time the lack of cardioprotection by RIPC in rats, assessed in individually randomized, blinded in vivo studies, involving three study centres, using different RIPC protocols. These results are in discrepancy with the meta-analysis of similar in vivo rat studies; however, no specific methodological reason could be identified by the systematic review, probably due to the overall insufficient reporting of several study parameters that did not improve over the past two decades. These results urge for publication of more well-designed and well-reported studies, irrespective of the outcome, which are required for preclinical reproducibility, and the development of clinically translatable cardioprotective interventions.


Asunto(s)
Precondicionamiento Isquémico , Daño por Reperfusión Miocárdica , Ratas , Masculino , Animales , Ratas Wistar , Reproducibilidad de los Resultados , Precondicionamiento Isquémico/métodos , Daño por Reperfusión Miocárdica/prevención & control
10.
Free Radic Biol Med ; 166: 33-52, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33588049

RESUMEN

The morbidity and mortality from cardiovascular diseases (CVD) remain high. Metabolic diseases such as obesity, hyperlipidemia, diabetes mellitus (DM), non-alcoholic fatty liver disease (NAFLD) and non-alcoholic steatohepatitis (NASH) as well as hypertension are the most common comorbidities in patients with CVD. These comorbidities result in increased myocardial oxidative stress, mainly from increased activity of nicotinamide adenine dinucleotide phosphate oxidases, uncoupled endothelial nitric oxide synthase, mitochondria as well as downregulation of antioxidant defense systems. Oxidative and nitrosative stress play an important role in ischemia/reperfusion injury and may account for increased susceptibility of the myocardium to infarction and myocardial dysfunction in the presence of the comorbidities. Thus, while early reperfusion represents the most favorable therapeutic strategy to prevent ischemia/reperfusion injury, redox therapeutic strategies may provide additive benefits, especially in patients with heart failure. While oxidative and nitrosative stress are harmful, controlled release of reactive oxygen species is however important for cardioprotective signaling. In this review we summarize the current data on the effect of hypertension and major cardiometabolic comorbidities such as obesity, hyperlipidemia, DM, NAFLD/NASH on cardiac redox homeostasis as well as on ischemia/reperfusion injury and cardioprotection. We also review and discuss the therapeutic interventions that may restore the redox imbalance in the diseased myocardium in the presence of these comorbidities.


Asunto(s)
Enfermedades Cardiovasculares , Miocardio , Enfermedades Cardiovasculares/epidemiología , Enfermedades Cardiovasculares/metabolismo , Enfermedades Cardiovasculares/prevención & control , Humanos , Infarto/metabolismo , Miocardio/metabolismo , Oxidación-Reducción , Estrés Oxidativo
11.
Basic Res Cardiol ; 105(2): 257-66, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19714395

RESUMEN

We demonstrated previously that adrenomedullin (AM), when given during early reperfusion, limited infarct size in rat heart. The present study was undertaken to provide direct evidence of the NO-dependency of AM's cardioprotective action by assessing NO biosynthesis and involvement of the soluble guanylyl cyclase (sGC) pathway. Perfused hearts from male CD-1 mice were subjected to 30-min left coronary occlusion and 60-min reperfusion. Infarct size was determined by tetrazolium staining. AM 10 nM was administered from 20 min after coronary occlusion until 10 min after reperfusion. Coronary effluent was analysed for NO2- and NO3-, and myocardial samples were analysed for NO2-, NO3-, nitroso-adducts and cGMP concentration. To examine the role of NO/sGC signalling in the infarct-limiting action of AM, further hearts received the sGC inhibitor ODQ 2 microM. AM treatment stimulated NO synthesis, indicated by increased NO2- efflux in coronary effluent throughout reperfusion (summarised as area under curve, AM 29.2 +/- 3.9 vs. control 14.4 +/- 2.8 micromol min2 mL(-1), P < 0.05). AM limited infarct size (35.4 +/- 2.7 vs. 12.2 +/- 2.3%, P < 0.01), associated with a 2.45-fold increase (P < 0.05) in myocardial cGMP concentration at 10 min after reperfusion. ODQ abolished the infarct size-limiting effect of AM (28.9 +/- 4.3%). These data provide the first evidence that AM increases NO bioavailability in intact murine myocardium and confirm that the NO/sGC/cGMP pathway is central to the cytoprotective action of AM against ischaemia-reperfusion injury.


Asunto(s)
Adrenomedulina/metabolismo , GMP Cíclico/metabolismo , Guanilato Ciclasa/metabolismo , Infarto del Miocardio/metabolismo , Daño por Reperfusión Miocárdica/metabolismo , Óxido Nítrico/biosíntesis , Adrenomedulina/administración & dosificación , Animales , Técnicas In Vitro , Masculino , Ratones , Infarto del Miocardio/etiología , Infarto del Miocardio/patología , Infarto del Miocardio/prevención & control , Daño por Reperfusión Miocárdica/complicaciones , Daño por Reperfusión Miocárdica/patología , Daño por Reperfusión Miocárdica/prevención & control , Miocardio/patología , Transducción de Señal
12.
Basic Res Cardiol ; 105(5): 643-50, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20349314

RESUMEN

Nitric oxide (NO) and B-type natriuretic peptide (BNP) are protective against ischemia-reperfusion injury as they increase intracellular cGMP level via activation of soluble (sGC) or particulate guanylate cyclases (pGC), respectively. The aim of the present study was to examine if the cGMP-elevating mediators, NO and BNP, share a common downstream signaling pathway via cGMP-dependent protein kinase (PKG) in cardiac cytoprotection. Neonatal rat cardiac myocytes in vitro were subjected to 2.5 h simulated ischemia (SI) followed by 2 h reoxygenation. Cell viability was tested by trypan blue exclusion assay. PKG activity of cardiac myocytes was assessed by phospholamban (PLB) phosphorylation determined by western blot. Cell death was 34 +/- 2% after SI/reoxygenation injury in the control group. cGMP-inducing agents significantly decreased irreversible cell injury: the cGMP analog 8-bromo-cGMP (8-Br-cGMP, 10 nM) decreased it to 13 +/- 1% (p < 0.001), the direct NO-donor S-nitroso-N-acetylpenicillamine (SNAP, 1 microM) to 18 +/- 6% (p < 0.05) and BNP (10 nM) to 12 +/- 2% (p < 0.001), respectively. This protective effect was abolished by the selective PKG inhibitor KT-5823 (600 nM) in each case. As PLB is not a unique reporter for PKG activity since it is also phosphorylated by protein kinase A (PKA), we examined PLB phosphorylation in the presence of the PKA inhibitor KT-5720 (1 microM). The ratio of pPLB/PLB significantly increased after administration of both BNP and 8-Br-cGMP under ischemic conditions, which was abolished by the PKG inhibitor. This is the first demonstration that elevated cGMP produced either by the sGC activator SNAP or the pGC activator BNP exerts cytoprotective effects via a common downstream signaling pathway involving PKG activation.


Asunto(s)
Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Transducción de Señal/fisiología , Animales , Animales Recién Nacidos , Proteínas de Unión al Calcio/metabolismo , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Células Cultivadas , GMP Cíclico/análogos & derivados , GMP Cíclico/farmacología , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Péptido Natriurético Encefálico/metabolismo , Óxido Nítrico/metabolismo , Donantes de Óxido Nítrico/farmacología , Fosforilación/efectos de los fármacos , Fosforilación/fisiología , Ratas , Ratas Wistar , Transducción de Señal/efectos de los fármacos
13.
Curr Opin Pharmacol ; 9(2): 177-88, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19109069

RESUMEN

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.


Asunto(s)
Cardiotónicos/uso terapéutico , Isquemia Miocárdica/tratamiento farmacológico , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Animales , Proteínas Reguladoras de la Apoptosis , Factor Natriurético Atrial/uso terapéutico , Autacoides/farmacología , Sistemas de Liberación de Medicamentos , Humanos , Precondicionamiento Isquémico Miocárdico/métodos , Proteínas de Transporte de Membrana Mitocondrial/antagonistas & inhibidores , Proteínas de Transporte de Membrana Mitocondrial/fisiología , Poro de Transición de la Permeabilidad Mitocondrial , Daño por Reperfusión Miocárdica/terapia , Antagonistas de Narcóticos , Agonistas del Receptor Purinérgico P1 , Antagonistas de Receptores Purinérgicos P1 , Receptores Opioides/agonistas , Receptores Opioides/uso terapéutico , Receptores Purinérgicos P1/uso terapéutico
14.
Cell Biochem Funct ; 28(2): 95-106, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20104507

RESUMEN

Since the discovery of endogenously-produced hydrogen sulfide (H(2)S) in various tissues, there has been an explosion of interest in H(2)S as a biological mediator alongside other gaseous mediators, nitric oxide and carbon monoxide. The identification of enzyme-regulated H(2)S synthetic pathways in the cardiovascular system has led to a number of studies examining specific regulatory actions of H(2)S. We review evidence showing that endogenously-generated and exogenously-administered H(2)S exerts a wide range of actions in vascular and myocardial cells including vasodilator/vasoconstrictor effects via modification of the smooth muscle tone, induction of apoptosis and anti-proliferative responses in the smooth muscle cells, angiogenic actions, effects relevant to inflammation and shock, and cytoprotection in models of myocardial ischemia-reperfusion injury. Several molecular mechanisms of action of H(2)S have been described. These include interactions of H(2)S with NO, redox regulation of multiple signaling proteins and regulation of K(ATP) channel opening. The gaps in our current understanding of precise mechanisms, the absence of selective pharmacological tools and the limited availability of H(2)S measurement techniques for living tissues, leave many questions about physiological and pathophysiological roles of H(2)S unanswered at present. Nevertheless, this area of investigation is advancing rapidly. We believe H(2)S holds promise as an endogenous mediator controlling a wide range of cardiovascular cell functions and integrated responses under both physiological and pathological conditions and may be amenable to therapeutic manipulation.


Asunto(s)
Sulfuro de Hidrógeno/metabolismo , Músculo Liso Vascular/metabolismo , Animales , Presión Sanguínea , Bovinos , Humanos , Mediadores de Inflamación/metabolismo , Canales KATP/metabolismo , Músculo Liso Vascular/citología , Daño por Reperfusión Miocárdica/metabolismo , Óxido Nítrico/metabolismo , Ratas
15.
Regul Pept ; 146(1-3): 271-7, 2008 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-18022257

RESUMEN

Apelin, the endogenous ligand of the G protein-coupled APJ receptor, is a peptide mediator with emerging regulatory actions in the heart. The aim of the present studies was to explore potential roles of the apelin/APJ system in myocardial ischaemia/reperfusion injury. To determine the cardiac expression of apelin/APJ and potential regulation by acute ischaemic insult, Langendorff perfused rat hearts were subjected to regional ischaemia (left coronary artery occlusion, 35 min) or ischaemia followed by reperfusion (30 min). Apelin and APJ mRNA expression were then determined in ventricular myocardium by rt-PCR. Unlike APJ mRNA expression, which remained unchanged, apelin mRNA was upregulated 2.4 fold in ventricular myocardium from isolated rat hearts undergoing ischaemia alone, but returned back to control levels after 30 min reperfusion. We then proceeded to test the hypothesis that treatment with exogenous apelin is protective against ischaemia/reperfusion injury. Perfused hearts were subjected to 35 min left main coronary artery occlusion and 120 min reperfusion, after which infarct size was determined by tetrazolium staining. Exogenous Pyr(1)-apelin-13 (10(-8 )M) was perfused either from 5 min prior to 15 min after coronary occlusion, or from 5 min prior to 15 min after reperfusion. Whilst ineffective when used during ischaemia alone, apelin administered during reperfusion significantly reduced infarct size (47.6+/-2.6% of ischaemic risk zone compared to 62.6+/-2.8% in control, n=10 each, p<0.05) in hearts subject to temporary coronary occlusion followed by reperfusion. This protective effect was not abolished by co-administration of the PI3K inhibitor wortmannin (10(-7 )M, infarct size 49.8+/-4.1%, n=4) or the P70S6 kinase inhibitor rapamycin (10(-9 )M, 41.8+/-8.8%, n=4). In conclusion these results suggest that apelin may be a new and potentially important cardioprotective autacoid, upregulated rapidly after myocardial ischaemia and acting through an unknown pathway.


Asunto(s)
Proteínas Portadoras/farmacología , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Animales , Apelina , Proteínas Portadoras/biosíntesis , Péptidos y Proteínas de Señalización Intercelular , Masculino , Daño por Reperfusión Miocárdica/prevención & control , Miocardio/metabolismo , ARN Mensajero/biosíntesis , Ratas , Ratas Sprague-Dawley , Receptores Acoplados a Proteínas G/biosíntesis
16.
Basic Res Cardiol ; 102(6): 529-41, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17896117

RESUMEN

Natriuretic peptides are regulatory autacoids in the mammalian myocardium whose functions, mediated via particulate guanylyl cyclase/cGMP, may include cytoprotection against ischaemia-reperfusion injury. Previous work has identified that B-type natriuretic peptide (BNP) limits infarct size when administered prior to and during coronary occlusion through a K(ATP) channel-dependent mechanism. The present study examined the hypothesis that the protection afforded by BNP is mediated specifically at reperfusion in a postconditioning-like manner. Langendorff-perfused rat hearts were subjected to 35 min coronary artery occlusion and 120 min reperfusion, and infarct size was determined by tetrazolium staining. Postconditioning was effected by applying six 10-second periods of global ischaemia at the onset of reperfusion.Treatment with either BNP 10 nM or the NO donor S-nitroso-N-acetylpenicillamine (SNAP) 1-10 microM was commenced 5 min prior to reperfusion and continued until 10 min after reperfusion. Control infarct size (% of ischaemic risk zone) was 40.8 +/- 3.7%.BNP at reperfusion induced a significant limitation of infarct size (BNP 22.9 +/- 4.1% P<0.05 vs. control). Co-treatment at reperfusion with BNP and the K(ATP) channel blockers 5-hydroxydecanote (5HD, 100 microM), glibenclamide (Glib; 10 microM) or HMR1098 (10 microM) abolished the infarct-limiting effect of BNP (BNP + 5HD 41.0 +/- 3.9%, BNP + Glib 39.8 +/- 5.6%, BNP + HMR 1098 46.0 +/- 7.1%,P < 0.05 vs. BNP). BNP given together with L-NAME (100 microM) at reperfusion resulted in a marked loss of protection (BNP + L-NAME 53.1 +/- 3.8% P < 0.001 vs. BNP). In a second series of experiments, SNAP (1-10 microM) given at reperfusion was found not to be protective (SNAP 1 microM 30.2 +/- 4.9%, SNAP 2 microM 27.5 +/- 9.5%, SNAP 5 microM 39.2 +/- 5.7%, SNAP 10 microM 33.7 +/- 6.4%, not significant vs. control). In a third series of experiments, postconditioning significantly limited infarct size (14.9 +/- 3.6 % vs. control 34.5 +/- 4.9%, P < 0.01) and this effect of postconditioning was abolished in the presence of isatin (100 microM), a non-specific blocker of particulate guanylyl cyclases (35.1 +/- 6%, P < 0.05 vs. postconditioning). In conclusion, pharmacological activation of pGC by BNP can effectively induce protection against reperfusion injury, by mechanisms involving K(ATP) channel opening and endogenous NO synthase activation. Furthermore, endogenous activation of pGC could play a role in the mechanism of postconditioning.


Asunto(s)
Infarto del Miocardio/prevención & control , Daño por Reperfusión Miocárdica/prevención & control , Reperfusión Miocárdica/métodos , Natriuréticos/farmacología , Péptido Natriurético Encefálico/farmacología , Animales , Modelos Animales de Enfermedad , Canales KATP/efectos de los fármacos , Masculino , Contracción Miocárdica/efectos de los fármacos , Infarto del Miocardio/patología , Daño por Reperfusión Miocárdica/patología , Miocardio/metabolismo , Miocardio/patología , Óxido Nítrico/farmacología , Óxido Nítrico Sintasa/metabolismo , Ratas , Ratas Sprague-Dawley , Flujo Sanguíneo Regional/efectos de los fármacos
17.
Exp Gerontol ; 42(8): 807-14, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17532161

RESUMEN

Although in experimental hypertension the cardioprotective effects of ischemic preconditioning (PC) appear to be maintained, most studies have examined the short-term hypertension in juvenile animals. However, aging may be an additional factor that influences the effectiveness of PC. The aim of this study was to characterise the effects on PC of LVH and aging simultaneously. Hearts from spontaneously hypertensive rats (SHR) and age-matched normotensive Wistar-Kyoto rats (WKY) were studied. Excised hearts were Langendorff-perfused to give equivalent coronary flow per gram heart weight. The left main coronary artery was occluded for 35 min followed by 120 min reperfusion. Infarct size was determined by tetrazolium staining. Heart size was assessed as left ventricle/body weight ratio (LV/BW). PC was effected with 2 x 5 min periods of global ischemia prior to coronary occlusion. Hearts were studied at 3-4 months (juvenile), 7-8 months (mature) or 12-13 months (aging). LV/BW ratio in SHR increased relative to WKY controls by 20%, 32% and 40% in juvenile, mature and aging hearts, respectively, but ischemic risk zone size was similar in all groups (52-59% of LV). PC was equally effective at limiting infarct size in juvenile and mature SHR and WKY hearts but was ineffective in aging hearts from both WKY and SHR. Since angiotensin-converting enzyme inhibitors enhance sub-threshold PC in normal myocardium, we also examined the action of captopril (Cap) in aging hearts. Additional aging hearts received treatment with Cap 200 microM as an adjunct to PC. Although Cap+PC was able to induce modest protection in aging WKY hearts, this was not seen in aging SHR hearts. We conclude that PC is lost in longstanding hypertension through independent contributions of both hypertension and aging. These findings may have implications for the clinical development of preconditioning-based therapies since elderly patients with longstanding hypertension are at high risk of developing ischemic heart disease.


Asunto(s)
Envejecimiento/fisiología , Hipertensión/fisiopatología , Hipertensión/terapia , Precondicionamiento Isquémico Miocárdico , Anciano , Envejecimiento/patología , Inhibidores de la Enzima Convertidora de Angiotensina/uso terapéutico , Animales , Humanos , Hipertensión/patología , Hipertrofia Ventricular Izquierda/patología , Hipertrofia Ventricular Izquierda/fisiopatología , Hipertrofia Ventricular Izquierda/terapia , Técnicas In Vitro , Masculino , Infarto del Miocardio/prevención & control , Ratas , Ratas Endogámicas SHR , Ratas Endogámicas WKY
18.
J Ethnopharmacol ; 209: 203-209, 2017 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-28689799

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Viscum album L. (European mistletoe) is a hemiparasitic plant belonging to Loranthaceae family and has been used in Turkish traditional medicine for the treatment of cardiovascular disorders and heart diseases such as hypertension, tachycardia and angina pectoris. AIM OF THE STUDY: The present study investigated the cardioprotective effects of V. album leaf extracts in myocardial ischemia and reperfusion injury in rats. MATERIAL AND METHODS: Lyophilized aqueous (AVa) and methanolic (MVa) extracts of V. album were prepared from dried leaf. The isolated hearts were perfused with V. album extracts prior to and during 35min of ischemia induced by coronary artery occlusion. After 120min of coronary reperfusion, infarct size was determined by triphenyltetrazolium staining. RESULTS: Both AVa and MVa extracts reduced the extent of infarction compared with untreated control hearts, but protective effect of MVa had more potential in low concentration; infarct size as proportion of ischemic risk zone: AVa 17.5±1.5%; Mva 20.3±2.5%, both P<0.01 versus control 38.1±1.4%. This protective effect was comparable to infarct limitation induced by ischemic preconditioning (21.5±2.4%). Inhibition of nitric oxide synthesis with L-NG-nitroarginine methyl ester completely abrogated the protection afforded by both extracts. ATP-sensitive K+ channel blockade by glibenclamide abrogated the protection afforded by MVa while attenuating, but not abolishing, the protective action of Ava. CONCLUSIONS: This study provided the first experimental evidence that V. album leaf extracts can mediate nitric oxide-dependent cardioprotection against myocardial injury produced by ischemia/reperfusion insult. With this study, popular usage of V. album extracts in Turkish folk medicine as a remedy for cardiac diseases was justified.


Asunto(s)
Daño por Reperfusión Miocárdica/tratamiento farmacológico , Extractos Vegetales/farmacología , Hojas de la Planta/química , Viscum album/química , Animales , Corazón/efectos de los fármacos , Masculino , Infarto del Miocardio/fisiopatología , Isquemia Miocárdica/fisiopatología , Miocardio/patología , Extractos Vegetales/química , Ratas , Ratas Sprague-Dawley
19.
Br J Pharmacol ; 174(4): 287-301, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27930802

RESUMEN

BACKGROUND AND PURPOSE: H2 S protects myocardium against ischaemia/reperfusion injury. This protection may involve the cytosolic reperfusion injury salvage kinase (RISK) pathway, but direct effects on mitochondrial function are possible. Here, we investigated the potential cardioprotective effect of a mitochondria-specific H2 S donor, AP39, at reperfusion against ischaemia/reperfusion injury. EXPERIMENTAL APPROACH: Anaesthetized rats underwent myocardial ischaemia (30 min)/reperfusion (120 min) with randomization to receive interventions before reperfusion: vehicle, AP39 (0.01, 0.1, 1 µmol·kg-1 ), or control compounds AP219 and ADT-OH (1 µmol·kg-1 ). LY294002, L-NAME or ODQ were used to investigate the involvement of the RISK pathway. Myocardial samples harvested 5 min after reperfusion were analysed for RISK protein phosphorylation and isolated cardiac mitochondria were used to examine the direct mitochondrial effects of AP39. KEY RESULTS: AP39, dose-dependently, reduced infarct size. Inhibition of either PI3K/Akt, eNOS or sGC did not affect this effect of AP39. Western blot analysis confirmed that AP39 did not induce phosphorylation of Akt, eNOS, GSK-3ß or ERK1/2. In isolated subsarcolemmal and interfibrillar mitochondria, AP39 significantly attenuated mitochondrial ROS generation without affecting respiratory complexes I or II. Furthermore, AP39 inhibited mitochondrial permeability transition pore (PTP) opening and co-incubation of mitochondria with AP39 and cyclosporine A induced an additive inhibitory effect on the PTP. CONCLUSION AND IMPLICATIONS: AP39 protects against reperfusion injury independently of the cytosolic RISK pathway. This cardioprotective effect could be mediated by inhibiting PTP via a cyclophilin D-independent mechanism. Thus, selective delivery of H2 S to mitochondria may be therapeutically applicable for employing the cardioprotective utility of H2 S.


Asunto(s)
Daño por Reperfusión Miocárdica/prevención & control , Compuestos Organofosforados/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Quinasas/metabolismo , Transducción de Señal/efectos de los fármacos , Tionas/farmacología , Animales , Relación Dosis-Respuesta a Droga , Masculino , Daño por Reperfusión Miocárdica/metabolismo , Ratas , Ratas Sprague-Dawley , Relación Estructura-Actividad
20.
Pharmacol Ther ; 105(2): 95-112, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15670621

RESUMEN

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.


Asunto(s)
Infarto del Miocardio/metabolismo , Péptidos/fisiología , Adrenomedulina , Animales , Supervivencia Celular , Vasos Coronarios/metabolismo , Vasos Coronarios/fisiopatología , Homeostasis , Humanos , Mediadores de Inflamación/metabolismo , Infarto del Miocardio/fisiopatología , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/fisiopatología , Neovascularización Patológica/metabolismo , Estrés Oxidativo , Transducción de Señal , Vasodilatación , Remodelación Ventricular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA