Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
J Am Heart Assoc ; 9(4): e014691, 2020 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-32067589

RESUMEN

Background DJ-1 is a ubiquitously expressed protein typically associated with the development of early onset Parkinson disease. Recent data suggest that it also plays a role in the cellular response to stress. Here, we sought to determine the role DJ-1 plays in the development of heart failure. Methods and Results Initial studies found that DJ-1 deficient mice (DJ-1 knockout; male; 8-10 weeks of age) exhibited more severe left ventricular cavity dilatation, cardiac dysfunction, hypertrophy, and fibrosis in the setting of ischemia-reperfusion-induced heart failure when compared with wild-type littermates. In contrast, the overexpression of the active form of DJ-1 using a viral vector approach resulted in significant improvements in the severity of heart failure when compared with mice treated with a control virus. Subsequent studies aimed at evaluating the underlying protective mechanisms found that cardiac DJ-1 reduces the accumulation of advanced glycation end products and activation of the receptor for advanced glycation end products-thus, reducing glycative stress. Conclusions These results indicate that DJ-1 is an endogenous cytoprotective protein that protects against the development of ischemia-reperfusion-induced heart failure by reducing glycative stress. Our findings also demonstrate the feasibility of using a gene therapy approach to deliver the active form of DJ-1 to the heart as a therapeutic strategy to protect against the consequences of ischemic injury, which is a major cause of death in western populations.


Asunto(s)
Insuficiencia Cardíaca/etiología , Insuficiencia Cardíaca/metabolismo , Estrés Oxidativo/fisiología , Proteína Desglicasa DJ-1/metabolismo , Proteína Desglicasa DJ-1/fisiología , Animales , Modelos Animales de Enfermedad , Productos Finales de Glicación Avanzada/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
2.
J Am Heart Assoc ; 7(19): e009565, 2018 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-30371303

RESUMEN

Background Lymphatic vessels interconnect with blood vessels to form an elaborate system that aids in the control of tissue pressure and edema formation. Although the lymphatic system has been known to exist in a heart, little is known about the role the cardiac lymphatic system plays in the development of heart failure. Methods and Results Mice (C57 BL /6J, male, 8 to 12 weeks of age) were subjected to either myocardial ischemia or myocardial ischemia and reperfusion for up to 28 days. Analysis revealed that both models increased the protein expression of vascular endothelial growth factor C and VEGF receptor 3 starting at 1 day after the onset of injury, whereas a significant increase in lymphatic vessel density was observed starting at 3 days. Further studies aimed to determine the consequences of inhibiting the endogenous lymphangiogenesis response on the development of heart failure. Using 2 different pharmacological approaches, we found that inhibiting VEGF receptor 3 with MAZ -51 and blocking endogenous vascular endothelial growth factor C with a neutralizing antibody blunted the increase in lymphatic vessel density, blunted lymphatic transport, increased inflammation, increased edema, and increased cardiac dysfunction. Subsequent studies revealed that augmentation of the endogenous lymphangiogenesis response with vascular endothelial growth factor C treatment reduced inflammation, reduced edema, and improved cardiac dysfunction. Conclusions These results suggest that the endogenous lymphangiogenesis response plays an adaptive role in the development of ischemic-induced heart failure and supports the emerging concept that therapeutic lymphangiogenesis is a promising new approach for the treatment of cardiovascular disease.


Asunto(s)
Insuficiencia Cardíaca/etiología , Linfangiogénesis/fisiología , Vasos Linfáticos/patología , Isquemia Miocárdica/fisiopatología , Daño por Reperfusión Miocárdica/fisiopatología , Remodelación Ventricular/fisiología , Animales , Western Blotting , Células Cultivadas , Modelos Animales de Enfermedad , Endotelio Vascular/patología , Insuficiencia Cardíaca/patología , Insuficiencia Cardíaca/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Isquemia Miocárdica/complicaciones , Isquemia Miocárdica/patología , Daño por Reperfusión Miocárdica/complicaciones , Daño por Reperfusión Miocárdica/patología
3.
J Mol Cell Cardiol ; 116: 29-40, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29408195

RESUMEN

BACKGROUND: Hydrogen sulfide (H2S) is an important regulator of mitochondrial bioenergetics, but its role in regulating mitochondrial biogenesis is not well understood. Using both genetic and pharmacological approaches, we sought to determine if H2S levels directly influenced cardiac mitochondrial content. RESULTS: Mice deficient in the H2S-producing enzyme, cystathionine γ-lyase (CSE KO) displayed diminished cardiac mitochondrial content when compared to wild-type hearts. In contrast, mice overexpressing CSE (CSE Tg) and mice supplemented with the orally active H2S-releasing prodrug, SG-1002, displayed enhanced cardiac mitochondrial content. Additional analysis revealed that cardiac H2S levels influenced the nuclear localization and transcriptional activity of peroxisome proliferator-activated receptor γ coactivator 1α (PGC1α) with higher levels having a positive influence and lower levels having a negative influence. Studies aimed at evaluating the underlying mechanisms found that H2S required AMP-activated protein kinase (AMPK) to induce PGC1α signaling and mitochondrial biogenesis. Finally, we found that restoring H2S levels with SG-1002 in the setting of heart failure increased cardiac mitochondrial content, improved mitochondrial respiration, improved ATP production efficiency, and improved cardiac function. CONCLUSIONS: Together, these results suggest that hydrogen sulfide is an important regulator of cardiac mitochondrial content and establishes that exogenous hydrogen sulfide can induce mitochondrial biogenesis via an AMPK-PGC1α signaling cascade.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Sulfuro de Hidrógeno/farmacología , Mitocondrias Cardíacas/metabolismo , Biogénesis de Organelos , Animales , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , ADN Mitocondrial/genética , Activación Enzimática/efectos de los fármacos , Insuficiencia Cardíaca/metabolismo , Humanos , Ratones Endogámicos C57BL , Mitocondrias Cardíacas/efectos de los fármacos , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Fosforilación/efectos de los fármacos , Proteína Fosfatasa 2/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Azufre/metabolismo
4.
Am J Cardiovasc Dis ; 6(3): 118-28, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27679746

RESUMEN

Angiotensin II (Ang II) modulates blood pressure and atherosclerosis development through its vascular type-1 (AT1R) and type-2 (AT2R) receptors, which have opposing effects. AT2R activation produces hypotension, and is anti-atherogenic. Targeted overexpression of AT2Rs in vascular smooth muscle cells (VSMCs) indicates that these effects are due to increased nitric oxide (NO) generation. However, the role of endogenous VSMC AT2Rs in these events is unknown. Effect of 7-day low-dose Ang II-infusion (12 µg/kg/hr) on blood pressure was tested in 9-week-old apoE((-/-)) mice fed a low or high cholesterol diet (LCD or HCD, respectively). Cardiac output was measured by echocardiography. Immunohistochemistry was performed to localize and quantify AT2Rs and p-Ser(1177)-endothelial nitric oxide synthase (eNOS) levels in the aortic arch. PD123319 and GW-9662 were used to selectively block the AT2R and peroxisome proliferator-activated receptor-γ (PPAR-γ), respectively. Ang II infusion decreased blood pressure by 12 mmHg (P < 0.001) in LCD/apoE((-/-)) mice without altering cardiac output; a response blocked by PD123319. Although, AT2R stimulation neither activated eNOS (p-Ser(1177)-eNOS) nor changed plasma NO metabolites, it caused an ~6-fold increase in VSMC PPAR-γ levels (P < 0.001) and the AT2R-mediated hypotension was abolished by GW-9662. AT2R-mediated hypotension was also inhibited by HCD, which selectively decreased VSMC AT2R expression by ~6-fold (P < 0.01). These findings suggest a novel pathway for the Ang II/AT2R-mediated hypotensive response that involves PPAR-γ, and is down regulated by a HCD.

5.
J Mol Cell Cardiol ; 97: 56-66, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27108530

RESUMEN

Recent data indicates that DJ-1 plays a role in the cellular response to stress. Here, we aimed to examine the underlying molecular mechanisms mediating the actions of DJ-1 in the heart following myocardial ischemia-reperfusion (I/R) injury. In response to I/R injury, DJ-1 KO mice displayed increased areas of infarction and worsened left ventricular function when compared to WT mice, confirming a protective role for DJ-1 in the heart. In an effort to evaluate the potential mechanism(s) responsible for the increased injury in DJ-1 KO mice, we focused on SUMOylation, a post-translational modification process that regulates various aspects of protein function. DJ-1 KO hearts after I/R injury were found to display enhanced accumulation of SUMO-1 modified proteins and reduced SUMO-2/3 modified proteins. Further analysis, revealed that the protein expression of the de-SUMOylation enzyme SENP1 was reduced, whereas the expression of SENP5 was enhanced in DJ-1 KO hearts after I/R injury. Finally, DJ-1 KO hearts were found to display enhanced SUMO-1 modification of dynamin-related protein 1, excessive mitochondrial fission, and dysfunctional mitochondria. Our data demonstrates that the activation of DJ-1 in response to myocardial I/R injury protects the heart by regulating the SUMOylation status of Drp1 and attenuating excessive mitochondrial fission.


Asunto(s)
Mitocondrias Cardíacas/genética , Mitocondrias Cardíacas/metabolismo , Dinámicas Mitocondriales/genética , Daño por Reperfusión Miocárdica/genética , Daño por Reperfusión Miocárdica/metabolismo , Proteína Desglicasa DJ-1/genética , Proteína Desglicasa DJ-1/metabolismo , Animales , Biopsia , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Masculino , Ratones , Ratones Noqueados , Daño por Reperfusión Miocárdica/patología , Miocitos Cardíacos/metabolismo , Estrés Oxidativo , Proteína Desglicasa DJ-1/deficiencia , Proteolisis , Ratas , Especies Reactivas de Oxígeno , Sumoilación
6.
Circ Heart Fail ; 9(4): e002368, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27056879

RESUMEN

BACKGROUND: Therapeutic strategies aimed at increasing hydrogen sulfide (H2S) levels exert cytoprotective effects in various models of cardiovascular injury. However, the underlying mechanism(s) responsible for this protection remain to be fully elucidated. Nuclear factor E2-related factor 2 (Nrf2) is a cellular target of H2S and facilitator of H2S-mediated cardioprotection after acute myocardial infarction. Here, we tested the hypothesis that Nrf2 mediates the cardioprotective effects of H2S therapy in the setting of heart failure. METHODS AND RESULTS: Mice (12 weeks of age) deficient in Nrf2 (Nrf2 KO; C57BL/6J background) and wild-type littermates were subjected to ischemic-induced heart failure. Wild-type mice treated with H2S in the form of sodium sulfide (Na2S) displayed enhanced Nrf2 signaling, improved left ventricular function, and less cardiac hypertrophy after the induction of heart failure. In contrast, Na2S therapy failed to provide protection against heart failure in Nrf2 KO mice. Studies aimed at evaluating the underlying cardioprotective mechanisms found that Na2S increased the expression of proteasome subunits, resulting in an increased proteasome activity and a reduction in the accumulation of damaged proteins. In contrast, Na2S therapy failed to enhance the proteasome and failed to attenuate the accumulation of damaged proteins in Nrf2 KO mice. Additionally, Na2S failed to improve cardiac function when the proteasome was inhibited. CONCLUSIONS: These findings indicate that Na2S therapy enhances proteasomal activity and function during the development of heart failure in an Nrf2-dependent manner and that this enhancement leads to attenuation in cardiac dysfunction.


Asunto(s)
Fármacos Cardiovasculares/farmacología , Insuficiencia Cardíaca/prevención & control , Sulfuro de Hidrógeno/farmacología , Isquemia Miocárdica/tratamiento farmacológico , Miocardio/enzimología , Factor 2 Relacionado con NF-E2/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Sulfuros/farmacología , Animales , Fármacos Cardiovasculares/metabolismo , Modelos Animales de Enfermedad , Estrés del Retículo Endoplásmico/efectos de los fármacos , Insuficiencia Cardíaca/enzimología , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/fisiopatología , Sulfuro de Hidrógeno/metabolismo , Hipertrofia Ventricular Izquierda/enzimología , Hipertrofia Ventricular Izquierda/fisiopatología , Hipertrofia Ventricular Izquierda/prevención & control , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Isquemia Miocárdica/enzimología , Isquemia Miocárdica/genética , Isquemia Miocárdica/fisiopatología , Factor 2 Relacionado con NF-E2/deficiencia , Factor 2 Relacionado con NF-E2/genética , Estrés Oxidativo/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Sulfuros/metabolismo , Factores de Tiempo , Función Ventricular Izquierda/efectos de los fármacos
7.
Nitric Oxide ; 46: 145-56, 2015 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-25575644

RESUMEN

Diabetic cardiomyopathy is a significant contributor to the morbidity and mortality associated with diabetes and metabolic syndrome. However, the underlying molecular mechanisms that lead to its development have not been fully elucidated. Hydrogen sulfide (H2S) is an endogenously produced signaling molecule that is critical for the regulation of cardiovascular homeostasis. Recently, therapeutic strategies aimed at increasing its levels have proven cardioprotective in models of acute myocardial ischemia-reperfusion injury and heart failure. The precise role of H2S in the pathogenesis of diabetic cardiomyopathy has not yet been established. Therefore, the goal of the present study was to evaluate circulating and cardiac H2S levels in a murine model of high fat diet (HFD)-induced cardiomyopathy. Diabetic cardiomyopathy was produced by feeding mice HFD (60% fat) chow for 24 weeks. HFD feeding reduced both circulating and cardiac H2S and induced hallmark features of type-2 diabetes. We also observed marked cardiac dysfunction, evidence of cardiac enlargement, cardiac hypertrophy, and fibrosis. H2S therapy (SG-1002, an orally active H2S donor) restored sulfide levels, improved some of the metabolic perturbations stemming from HFD feeding, and attenuated HFD-induced cardiac dysfunction. Additional analysis revealed that H2S therapy restored adiponectin levels and suppressed cardiac ER stress stemming from HFD feeding. These results suggest that diminished circulating and cardiac H2S levels play a role in the pathophysiology of HFD-induced cardiomyopathy. Additionally, these results suggest that H2S therapy may be of clinical importance in the treatment of cardiovascular complications stemming from diabetes.


Asunto(s)
Cardiomiopatías Diabéticas/fisiopatología , Dieta Alta en Grasa , Estrés del Retículo Endoplásmico/efectos de los fármacos , Sulfuro de Hidrógeno/farmacología , Proteínas Quinasas Activadas por AMP/metabolismo , Adiponectina/metabolismo , Administración Oral , Animales , Corazón/efectos de los fármacos , Sulfuro de Hidrógeno/química , Masculino , Ratones , Ratones Endogámicos C57BL , Miocardio/química , Transducción de Señal/efectos de los fármacos
8.
Med Gas Res ; 4(1): 20, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25525500

RESUMEN

BACKGROUND: Coronary artery disease remains the principal cause of death in patients with diabetes mellitus. Diabetic mice display exacerbated injury following myocardial ischemia-reperfusion (MI/R) and are resistant to most therapeutic interventions. We have reported that sodium sulfide (Na2S) therapy confers cardioprotection during MI/R in non-diabetic mice. Here we tested the hypothesis that Na2S therapy would limit the extent of myocardial injury following MI/R when administered at the time of reperfusion. METHODS AND RESULTS: Diabetic mice (db/db, 12 weeks of age) were subjected to transient myocardial ischemia for a period of 30 minutes followed by reperfusion up to 24 hours. Na2S (0.05 to 1 mg/kg) or saline (vehicle) was administered into the left ventricular lumen at the time of reperfusion. Na2S therapy significantly decreased myocardial injury in the db/db diabetic mouse, as evidenced by a reduction in infarct size and circulating troponin-I levels. The reduction in myocardial injury was also associated with a reduction in oxidative stress and a decrease in cleaved caspase-3 expression. In an effort to evaluate the signaling mechanism responsible for the observed cardioprotection, additional groups of mice were sacrificed during early reperfusion. Hearts were excised and processed for Western blot analysis. These studies revealed that Na2S therapy activated the Erk1/2 arm of the Reperfusion Injury Salvage Kinase (RISK) pathway. CONCLUSION: These findings provide important information that myocardial Erk1/2 activation by Na2S therapy following MI/R sets into motion events, which ultimately lead to cardioprotection in the setting of diabetes.

9.
Circ Res ; 114(8): 1281-91, 2014 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-24599803

RESUMEN

RATIONALE: Nitric oxide (NO) bioavailability is reduced in the setting of heart failure. Nitrite (NO2) is a critically important NO intermediate that is metabolized to NO during pathological states. We have previously demonstrated that sodium nitrite ameliorates acute myocardial ischemia/reperfusion injury. OBJECTIVE: No evidence exists as to whether increasing NO bioavailability via nitrite therapy attenuates heart failure severity after pressure-overload-induced hypertrophy. METHODS AND RESULTS: Serum from patients with heart failure exhibited significantly decreased nitrosothiol and cGMP levels. Transverse aortic constriction was performed in mice at 10 to 12 weeks. Sodium nitrite (50 mg/L) or saline vehicle was administered daily in the drinking water postoperative from day 1 for 9 weeks. Echocardiography was performed at baseline and at 1, 3, 6, and 9 weeks after transverse aortic constriction to assess left ventricular dimensions and ejection fraction. We observed increased cardiac nitrite, nitrosothiol, and cGMP levels in mice treated with nitrite. Sodium nitrite preserved left ventricular ejection fraction and improved left ventricular dimensions at 9 weeks (P<0.001 versus vehicle). In addition, circulating and cardiac brain natriuretic peptide levels were attenuated in mice receiving nitrite (P<0.05 versus vehicle). Western blot analyses revealed upregulation of Akt-endothelial nitric oxide-nitric oxide-cGMP-GS3Kß signaling early in the progression of hypertrophy and heart failure. CONCLUSIONS: These results support the emerging concept that nitrite therapy may be a viable clinical option for increasing NO levels and may have a practical clinical use in the treatment of heart failure.


Asunto(s)
Citoprotección/fisiología , Insuficiencia Cardíaca/tratamiento farmacológico , Óxido Nítrico/fisiología , Transducción de Señal/fisiología , Nitrito de Sodio/uso terapéutico , Disfunción Ventricular Izquierda/tratamiento farmacológico , Anciano , Animales , Disponibilidad Biológica , GMP Cíclico/metabolismo , Modelos Animales de Enfermedad , Femenino , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/fisiopatología , Hemodinámica/efectos de los fármacos , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Transducción de Señal/efectos de los fármacos , Nitrito de Sodio/farmacología , Volumen Sistólico/efectos de los fármacos , Disfunción Ventricular Izquierda/fisiopatología
10.
Proc Natl Acad Sci U S A ; 111(8): 3182-7, 2014 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-24516168

RESUMEN

Previous studies have demonstrated that hydrogen sulfide (H2S) protects against multiple cardiovascular disease states in a similar manner as nitric oxide (NO). H2S therapy also has been shown to augment NO bioavailability and signaling. The purpose of this study was to investigate the impact of H2S deficiency on endothelial NO synthase (eNOS) function, NO production, and ischemia/reperfusion (I/R) injury. We found that mice lacking the H2S-producing enzyme cystathionine γ-lyase (CSE) exhibit elevated oxidative stress, dysfunctional eNOS, diminished NO levels, and exacerbated myocardial and hepatic I/R injury. In CSE KO mice, acute H2S therapy restored eNOS function and NO bioavailability and attenuated I/R injury. In addition, we found that H2S therapy fails to protect against I/R in eNOS phosphomutant mice (S1179A). Our results suggest that H2S-mediated cytoprotective signaling in the setting of I/R injury is dependent in large part on eNOS activation and NO generation.


Asunto(s)
Citoprotección/fisiología , Sulfuro de Hidrógeno/metabolismo , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Óxido Nítrico Sintasa de Tipo III/metabolismo , Óxido Nítrico/metabolismo , Transducción de Señal/fisiología , Alanina Transaminasa/sangre , Análisis de Varianza , Animales , Aspartato Aminotransferasas/sangre , Western Blotting , Cromatografía Líquida de Alta Presión , Cistationina gamma-Liasa/genética , Citoprotección/efectos de los fármacos , Sulfuro de Hidrógeno/farmacología , Inmunohistoquímica , Ratones , Ratones Noqueados , Mitocondrias/fisiología , Daño por Reperfusión Miocárdica/metabolismo , Estrés Oxidativo/fisiología , Consumo de Oxígeno/fisiología , Troponina I/metabolismo
11.
J Mol Cell Cardiol ; 64: 1-10, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23962643

RESUMEN

The infarct sparing effects of exercise are evident following both long-term and short-term training regimens. Here we compared the infarct-lowering effects of nitrite therapy, voluntary exercise, and the combination of both following myocardial ischemia-reperfusion (MI/R) injury. We also compared the degree to which each strategy increased cardiac nitrite levels, as well as the effects of each strategy on the nitrite reductase activity of the heart. Mice subjected to voluntary wheel running (VE) for 4weeks displayed an 18% reduction in infarct size when compared to sedentary mice, whereas mice administered nitrite therapy (25mg/L in drinking water) showed a 53% decrease. However, the combination of VE and nitrite exhibited no further protection than VE alone. Although the VE and nitrite therapy mice showed similar nitrite levels in the heart, cardiac nitrite reductase activity was significantly reduced in the VE mice. Additionally, the cardiac protein expression of myoglobin, a known nitrite reductase, was also reduced after VE. Further studies revealed that cardiac NFAT activity was lower after VE due to a decrease in calcineurin activity and an increase in GSK3ß activity. These data suggest that VE downregulates cardiac myoglobin levels by inhibiting calcineurin/NFAT signaling. Additionally, these results suggest that the modest infarct sparing effects of VE are the result of a decrease in the hearts ability to reduce nitrite to nitric oxide during MI/R.


Asunto(s)
Regulación de la Expresión Génica , Daño por Reperfusión Miocárdica/metabolismo , Miocardio/metabolismo , Mioglobina/genética , Nitrito Reductasas/metabolismo , Condicionamiento Físico Animal , Animales , Cardiotónicos/administración & dosificación , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Modelos Animales de Enfermedad , Activación Enzimática , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Masculino , Ratones , Ratones Noqueados , Proteínas de Microfilamentos , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Mioglobina/metabolismo , Óxido Nítrico/metabolismo , Nitritos/administración & dosificación , Nitritos/metabolismo
12.
Am J Physiol Heart Circ Physiol ; 304(9): H1215-24, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23479260

RESUMEN

Hydrogen sulfide (H2S) therapy protects nondiabetic animals in various models of myocardial injury, including acute myocardial infarction and heart failure. Here, we sought to examine whether H2S therapy provides cardioprotection in the setting of type 2 diabetes. H2S therapy in the form of sodium sulfide (Na2S) beginning 24 h or 7 days before myocardial ischemia significantly decreased myocardial injury in db/db diabetic mice (12 wk of age). In an effort to evaluate the signaling mechanism responsible for the observed cardioprotection, we focused on the role of nuclear factor E2-related factor (Nrf2) signaling. Our results indicate that diabetes does not alter the ability of H2S to increase the nuclear localization of Nrf2, but does impair aspects of Nrf2 signaling. Specifically, the expression of NADPH quinine oxidoreductase 1 was increased after the acute treatment, whereas the expression of heme-oxygenase-1 (HO-1) was only increased after 7 days of treatment. This discrepancy was found to be the result of an increased nuclear expression of Bach1, a known repressor of HO-1 transcription, which blocked the binding of Nrf2 to the HO-1 promoter. Further analysis revealed that 7 days of Na2S treatment overcame this impairment by removing Bach1 from the nucleus in an Erk1/2-dependent manner. Our findings demonstrate for the first time that exogenous administration of Na2S attenuates myocardial ischemia-reperfusion injury in db/db mice, suggesting the potential therapeutic effects of H2S in treating a heart attack in the setting of type 2 diabetes.


Asunto(s)
Diabetes Mellitus Tipo 2/complicaciones , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Sulfuro de Hidrógeno/uso terapéutico , Precondicionamiento Isquémico Miocárdico , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Factor 2 Relacionado con NF-E2/metabolismo , Animales , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , Expresión Génica/efectos de los fármacos , Hemo-Oxigenasa 1/genética , Hemo-Oxigenasa 1/metabolismo , Sistema de Señalización de MAP Quinasas , Masculino , Ratones , Ratones Endogámicos C57BL , Daño por Reperfusión Miocárdica/complicaciones , Daño por Reperfusión Miocárdica/metabolismo , NAD(P)H Deshidrogenasa (Quinona)/genética , NAD(P)H Deshidrogenasa (Quinona)/metabolismo , Sulfuros/uso terapéutico
13.
Circulation ; 127(10): 1116-27, 2013 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-23393010

RESUMEN

BACKGROUND: Cystathionine γ-lyase (CSE) produces H2S via enzymatic conversion of L-cysteine and plays a critical role in cardiovascular homeostasis. We investigated the effects of genetic modulation of CSE and exogenous H2S therapy in the setting of pressure overload-induced heart failure. METHODS AND RESULTS: Transverse aortic constriction was performed in wild-type, CSE knockout, and cardiac-specific CSE transgenic mice. In addition, C57BL/6J or CSE knockout mice received a novel H2S donor (SG-1002). Mice were followed up for 12 weeks with echocardiography. We observed a >60% reduction in myocardial and circulating H2S levels after transverse aortic constriction. CSE knockout mice exhibited significantly greater cardiac dilatation and dysfunction than wild-type mice after transverse aortic constriction, and cardiac-specific CSE transgenic mice maintained cardiac structure and function after transverse aortic constriction. H2S therapy with SG-1002 resulted in cardioprotection during transverse aortic constriction via upregulation of the vascular endothelial growth factor-Akt-endothelial nitric oxide synthase-nitric oxide-cGMP pathway with preserved mitochondrial function, attenuated oxidative stress, and increased myocardial vascular density. CONCLUSIONS: Our results demonstrate that H2S levels are decreased in mice in the setting of heart failure. Moreover, CSE plays a critical role in the preservation of cardiac function in heart failure, and oral H2S therapy prevents the transition from compensated to decompensated heart failure in part via upregulation of endothelial nitric oxide synthase and increased nitric oxide bioavailability.


Asunto(s)
Cardiotónicos/uso terapéutico , Insuficiencia Cardíaca/tratamiento farmacológico , Insuficiencia Cardíaca/enzimología , Sulfuro de Hidrógeno/uso terapéutico , Óxido Nítrico Sintasa de Tipo III/biosíntesis , Regulación hacia Arriba/efectos de los fármacos , Animales , Cardiotónicos/administración & dosificación , Cistationina gamma-Liasa/deficiencia , Cistationina gamma-Liasa/genética , Insuficiencia Cardíaca/fisiopatología , Sulfuro de Hidrógeno/administración & dosificación , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Óxido Nítrico Sintasa de Tipo III/fisiología , Regulación hacia Arriba/fisiología
14.
Arterioscler Thromb Vasc Biol ; 33(4): 744-51, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23349187

RESUMEN

OBJECTIVE: The aim of this study was to determine whether thioredoxin 1 (Trx1) mediates the cardioprotective effects of hydrogen sulfide (H2S) in a model of ischemic-induced heart failure (HF). APPROACH AND RESULTS: Mice with a cardiac-specific overexpression of a dominant negative mutant of Trx1 and wild-type littermates were subjected to ischemic-induced HF. Treatment with H2S as sodium sulfide (Na2S) not only increased the gene and protein expression of Trx1 in the absence of ischemia but also augmented the HF-induced increase in both. Wild-type mice treated with Na2S experienced less left-ventricular dilatation, improved left-ventricular function, and less cardiac hypertrophy after the induction of HF. In contrast, Na2S therapy failed to improve any of these parameters in the dominant negative mutant of Trx1 mice. Studies aimed at evaluating the underlying cardioprotective mechanisms found that Na2S therapy inhibited HF-induced apoptosis signaling kinase-1 signaling and nuclear export of histone deacetylase 4 in a Trx1-dependent manner. CONCLUSIONS: These findings provide novel information that the upregulation of Trx1 by Na2S therapy in the setting of HF sets into motion events, such as the inhibition of apoptosis signaling kinase-1 signaling and histone deacetylase 4 nuclear export, which ultimately leads to the attenuationof left-ventricular remodeling.


Asunto(s)
Cardiotónicos/farmacología , Insuficiencia Cardíaca/tratamiento farmacológico , Miocardio/metabolismo , Sulfuros/farmacología , Tiorredoxinas/metabolismo , Transporte Activo de Núcleo Celular , Animales , Cardiotónicos/metabolismo , Modelos Animales de Enfermedad , Genes Reporteros , Insuficiencia Cardíaca/diagnóstico por imagen , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/fisiopatología , Hemodinámica/efectos de los fármacos , Histona Desacetilasas/metabolismo , Sulfuro de Hidrógeno/metabolismo , Hipertrofia Ventricular Izquierda/metabolismo , Hipertrofia Ventricular Izquierda/prevención & control , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Luciferasas/genética , Luciferasas/metabolismo , MAP Quinasa Quinasa Quinasa 5/antagonistas & inhibidores , MAP Quinasa Quinasa Quinasa 5/metabolismo , Masculino , Ratones , Ratones Transgénicos , Mutación , Miocardio/patología , Factores de Transcripción NFATC/genética , ARN Mensajero/metabolismo , Transducción de Señal/efectos de los fármacos , Volumen Sistólico/efectos de los fármacos , Sulfuros/metabolismo , Tiorredoxinas/genética , Factores de Tiempo , Ultrasonografía , Regulación hacia Arriba , Función Ventricular Izquierda/efectos de los fármacos , Remodelación Ventricular/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
15.
Circ Res ; 108(12): 1448-58, 2011 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-21527738

RESUMEN

RATIONALE: Exercise training confers sustainable protection against ischemia-reperfusion injury in animal models and has been associated with improved survival following a heart attack in humans. It is still unclear how exercise protects the heart, but it is apparent that endothelial nitric oxide synthase (eNOS) and nitric oxide (NO) play a role. OBJECTIVE: To determine the role of ß(3)-adrenergic receptors (ß(3)-ARs), eNOS activation, and NO metabolites (nitrite and nitrosothiols) in the sustained cardioprotective effects of exercise. METHODS AND RESULTS: Here we show that voluntary exercise reduces myocardial injury in mice following a 4-week training period and that these protective effects can be sustained for at least 1 week following the cessation of the training. The sustained cardioprotective effects of exercise are mediated by alterations in the phosphorylation status of eNOS (increase in serine 1177 and decrease in threonine 495), leading to an increase in NO generation and storage of NO metabolites (nitrite and nitrosothiols) in the heart. Further evidence revealed that the alterations in eNOS phosphorylation status and NO generation were mediated by ß(3)-AR stimulation and that in response to exercise a deficiency of ß(3)-ARs leads to an exacerbation of myocardial infarction following ischemia-reperfusion injury. CONCLUSIONS: Our findings clearly demonstrate that exercise protects the heart against myocardial ischemia-reperfusion injury by stimulation of ß(3)-ARs and increased cardiac storage of nitric oxide metabolites (ie, nitrite and nitrosothiols).


Asunto(s)
Ejercicio Físico , Daño por Reperfusión Miocárdica/prevención & control , Óxido Nítrico/metabolismo , Nitritos/metabolismo , Compuestos Nitrosos/metabolismo , Condicionamiento Físico Animal , Receptores Adrenérgicos beta 3/metabolismo , Adolescente , Adulto , Animales , Activación Enzimática , Humanos , Masculino , Ratones , Ratones Noqueados , Daño por Reperfusión Miocárdica/genética , Daño por Reperfusión Miocárdica/metabolismo , Óxido Nítrico/genética , Óxido Nítrico Sintasa de Tipo III/genética , Óxido Nítrico Sintasa de Tipo III/metabolismo , Receptores Adrenérgicos beta 3/genética , Factores de Tiempo
16.
Pharmacol Res ; 62(4): 289-97, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20542117

RESUMEN

Gasotransmitters are lipid soluble, endogenously produced gaseous signaling molecules that freely permeate the plasma membrane of a cell to directly activate intracellular targets, thus alleviating the need for membrane-bound receptors. The gasotransmitter family consists of three members: nitric oxide (NO), carbon monoxide (CO), and hydrogen sulfide (H(2)S). H(2)S is the latest gasotransmitter to be identified and characterized and like the other members of the gasotransmitter family, H(2)S was historically considered to be a toxic gas and an environmental/occupational hazard. However with the discovery of its presence and enzymatic production in mammalian tissues, H(2)S has gained much attention as a physiological signaling molecule. Also, much like NO and CO, H(2)S's role in ischemia/reperfusion (I/R) injury has recently begun to be elucidated. As such, modulation of endogenous H(2)S and administration of exogenous H(2)S has now been demonstrated to be cytoprotective in various organ systems through diverse signaling mechanisms. This review will provide a detailed description of the role H(2)S plays in different model systems of I/R injury and will also detail some of the mechanisms involved with its cytoprotection.


Asunto(s)
Sulfuro de Hidrógeno/metabolismo , Daño por Reperfusión/metabolismo , Animales , Citoprotección/efectos de los fármacos , Humanos , Sulfuro de Hidrógeno/farmacología
17.
Circulation ; 122(1): 11-9, 2010 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-20566952

RESUMEN

BACKGROUND: Hydrogen sulfide (H(2)S) is an endogenous signaling molecule with potent cytoprotective effects. The present study evaluated the therapeutic potential of H(2)S in murine models of heart failure. METHODS AND RESULTS: Heart failure was induced by subjecting mice either to permanent ligation of the left coronary artery for 4 weeks or to 60 minutes of left coronary artery occlusion followed by reperfusion for 4 weeks. Transgenic mice with cardiac-restricted overexpression of the H(2)S-generating enzyme cystathione gamma-lyase (alphaMHC-CGL-Tg(+)) displayed a clear protection against left ventricular structural and functional impairment as assessed by echocardiography in response to ischemia-induced heart failure, as well as improved survival in response to permanent myocardial ischemia. Exogenous H(2)S therapy (Na(2)S; 100 microg/kg) administered at the time of reperfusion (intracardiac) and then daily (intravenous) for the first 7 days after myocardial ischemia also protected against the structural and functional deterioration of the left ventricle by attenuating oxidative stress and mitochondrial dysfunction. Additional experiments aimed at elucidating some of the protective mechanisms of H(2)S therapy found that 7 days of H(2)S therapy increased the phosphorylation of Akt and increased the nuclear localization of 2 transcription factors, nuclear respiratory factor 1 and nuclear factor-E2-related factor (Nrf2), that are involved in increasing the levels of endogenous antioxidants, attenuating apoptosis, and increasing mitochondrial biogenesis. CONCLUSIONS: The results of the present study suggest that either the administration of exogenous H(2)S or the modulation of endogenous H(2)S production may be of therapeutic benefit in the treatment of ischemia-induced heart failure.


Asunto(s)
Cistationina gamma-Liasa/genética , Insuficiencia Cardíaca/tratamiento farmacológico , Sulfuro de Hidrógeno/uso terapéutico , Isquemia Miocárdica/complicaciones , Animales , Peso Corporal , Cardiomegalia/enzimología , Cardiomegalia/prevención & control , Regulación Enzimológica de la Expresión Génica , Insuficiencia Cardíaca/etiología , Insuficiencia Cardíaca/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Isquemia Miocárdica/genética , Factor 2 Relacionado con NF-E2/efectos de los fármacos , Factor 2 Relacionado con NF-E2/metabolismo , Factor Nuclear 1 de Respiración/efectos de los fármacos , Factor Nuclear 1 de Respiración/metabolismo , Tamaño de los Órganos , Sulfatos/farmacología , Tasa de Supervivencia , Vasodilatación/efectos de los fármacos , Vasodilatación/genética , Disfunción Ventricular Izquierda/genética , Función Ventricular Izquierda/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA