Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 12(1): 4982, 2022 03 23.
Artigo em Inglês | MEDLINE | ID: mdl-35322094

RESUMO

Although cyanide's biological effects are pleiotropic, its most obvious effects are as a metabolic poison. Cyanide potently inhibits cytochrome c oxidase and potentially other metabolic enzymes, thereby unleashing a cascade of metabolic perturbations that are believed to cause lethality. From systematic screens of human metabolites using a zebrafish model of cyanide toxicity, we have identified the TCA-derived small molecule glyoxylate as a potential cyanide countermeasure. Following cyanide exposure, treatment with glyoxylate in both mammalian and non-mammalian animal models confers resistance to cyanide toxicity with greater efficacy and faster kinetics than known cyanide scavengers. Glyoxylate-mediated cyanide resistance is accompanied by rapid pyruvate consumption without an accompanying increase in lactate concentration. Lactate dehydrogenase is required for this effect which distinguishes the mechanism of glyoxylate rescue as distinct from countermeasures based solely on chemical cyanide scavenging. Our metabolic data together support the hypothesis that glyoxylate confers survival at least in part by reversing the cyanide-induced redox imbalances in the cytosol and mitochondria. The data presented herein represent the identification of a potential cyanide countermeasure operating through a novel mechanism of metabolic modulation.


Assuntos
Glioxilatos , Peixe-Zebra , Animais , Cianetos/toxicidade , Mamíferos , Ácido Pirúvico
2.
PLoS One ; 13(6): e0193889, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29879736

RESUMO

Cyanide is a potent toxic agent, and the few available antidotes are not amenable to rapid deployment in mass exposures. As a result, there are ongoing efforts to exploit different animal models to identify novel countermeasures. We have created a pipeline that combines high-throughput screening in zebrafish with subsequent validation in two mammalian small animal models as well as a porcine large animal model. We found that zebrafish embryos in the first 3 days post fertilization (dpf) are highly resistant to cyanide, becoming progressively more sensitive thereafter. Unbiased analysis of gene expression in response to several hours of ultimately lethal doses of cyanide in both 1 and 7 dpf zebrafish revealed modest changes in iron-related proteins associated with the age-dependent cyanide resistance. Metabolomics measurements demonstrated significant age-dependent differences in energy metabolism during cyanide exposure which prompted us to test modulators of the tricarboxylic acid cycle and related metabolic processes as potential antidotes. In cyanide-sensitive 7 dpf larvae, we identified several such compounds that offer significant protection against cyanide toxicity. Modulators of the pyruvate dehydrogenase complex, as well as the small molecule sodium glyoxylate, consistently protected against cyanide toxicity in 7 dpf zebrafish larvae. Together, our results indicate that the resistance of zebrafish embryos to cyanide toxicity during early development is related to an altered regulation of cellular metabolism, which we propose may be exploited as a potential target for the development of novel antidotes against cyanide poisoning.


Assuntos
Metaboloma/efeitos dos fármacos , Metabolômica , Cianeto de Potássio/intoxicação , Complexo Piruvato Desidrogenase/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/metabolismo , Animais , Suínos
3.
BMC Anesthesiol ; 17(1): 76, 2017 06 14.
Artigo em Inglês | MEDLINE | ID: mdl-28615047

RESUMO

BACKGROUND: Volatile anesthetics increase levels of the neurotransmitter nitric oxide (NO) and the secondary messenger molecule cyclic guanosine monophosphate (cGMP) in the brain. NO activates the enzyme guanylyl cyclase (GC) to produce cGMP. We hypothesized that the NO-GC-cGMP pathway contributes to anesthesia-induced unconsciousness. METHODS: Sevoflurane-induced loss and return of righting reflex (LORR and RORR, respectively) were studied in wild-type mice (WT) and in mice congenitally deficient in the GC-1α subunit (GC-1-/- mice). Spatial distributions of GC-1α and the GC-2α subunit in the brain were visualized by in situ hybridization. Brain cGMP levels were measured in WT and GC-1-/- mice after inhaling oxygen with or without 1.2% sevoflurane for 20 min. RESULTS: Higher concentrations of sevoflurane were required to induce LORR in GC-1-/- mice than in WT mice (1.5 ± 0.1 vs. 1.1 ± 0.2%, respectively, n = 14 and 14, P < 0.0001). Similarly, RORR occurred at higher concentrations of sevoflurane in GC-1-/- mice than in WT mice (1.0 ± 0.1 vs. 0.8 ± 0.1%, respectively, n = 14 and 14, P < 0.0001). Abundant GC-1α and GC-2α mRNA expression was detected in the cerebral cortex, medial habenula, hippocampus, and cerebellum. Inhaling 1.2% sevoflurane for 20 min increased cGMP levels in the brains of WT mice from 2.6 ± 2.0 to 5.5 ± 3.7 pmol/mg protein (n = 13 and 10, respectively, P = 0.0355) but not in GC-1-/- mice. CONCLUSION: Congenital deficiency of GC-1α abolished the ability of sevoflurane anesthesia to increase cGMP levels in the whole brain, and increased the concentration of sevoflurane required to induce LORR. Impaired NO-cGMP signaling raises the threshold for producing sevoflurane-induced unconsciousness in mice.


Assuntos
Anestésicos Inalatórios/farmacologia , Guanilato Ciclase/genética , Éteres Metílicos/farmacologia , Animais , Encéfalo/metabolismo , Guanosina Monofosfato/metabolismo , Camundongos Knockout , Reflexo de Endireitamento/efeitos dos fármacos , Sevoflurano
4.
Am J Physiol Heart Circ Physiol ; 310(11): H1790-800, 2016 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-27199131

RESUMO

Dysregulated nitric oxide (NO) signaling contributes to the pathogenesis of hypertension, a prevalent and often sex-specific risk factor for cardiovascular disease. We previously reported that mice deficient in the α1-subunit of the NO receptor soluble guanylate cyclase (sGCα1 (-/-) mice) display sex- and strain-specific hypertension: male but not female sGCα1 (-/-) mice are hypertensive on an 129S6 (S6) but not a C57BL6/J (B6) background. We aimed to uncover the genetic and molecular basis of the observed sex- and strain-specific blood pressure phenotype. Via linkage analysis, we identified a suggestive quantitative trait locus associated with elevated blood pressure in male sGCα1 (-/-)S6 mice. This locus encompasses Cyp4a12a, encoding the predominant murine synthase of the vasoconstrictor 20-hydroxy-5,8,11,14-eicosatetraenoic acid (20-HETE). Renal expression of Cyp4a12a in mice was associated with genetic background, sex, and testosterone levels. In addition, 20-HETE levels were higher in renal preglomerular microvessels of male sGCα1 (-/-)S6 than of male sGCα1 (-/-)B6 mice. Furthermore, treating male sGCα1 (-/-)S6 mice with the 20-HETE antagonist 20-hydroxyeicosa-6(Z),15(Z)-dienoic acid (20-HEDE) lowered blood pressure. Finally, 20-HEDE rescued the genetic background- and testosterone-dependent impairment of acetylcholine-induced relaxation in renal interlobar arteries associated with sGCα1 deficiency. Elevated Cyp4a12a expression and 20-HETE levels render mice susceptible to hypertension and vascular dysfunction in a setting of sGCα1 deficiency. Our data identify Cyp4a12a as a candidate sex-specific blood pressure-modifying gene in the context of deficient NO-sGC signaling.


Assuntos
Androgênios/farmacologia , Família 4 do Citocromo P450/genética , Ácidos Hidroxieicosatetraenoicos/metabolismo , Hipertensão/metabolismo , Guanilil Ciclase Solúvel/metabolismo , Animais , Pressão Sanguínea/efeitos dos fármacos , Pressão Sanguínea/fisiologia , Feminino , Ligação Genética , Hipertensão/genética , Rim/efeitos dos fármacos , Rim/metabolismo , Masculino , Camundongos , Camundongos Knockout , Locos de Características Quantitativas , Fatores Sexuais , Guanilil Ciclase Solúvel/genética , Testosterona/sangue
5.
Circ Res ; 117(9): 793-803, 2015 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-26259881

RESUMO

RATIONALE: The regulation of calcium (Ca(2+)) homeostasis by ß-adrenergic receptor (ßAR) activation provides the essential underpinnings of sympathetic regulation of myocardial function, as well as a basis for understanding molecular events that result in hypertrophic signaling and heart failure. Sympathetic stimulation of the ßAR not only induces protein phosphorylation but also activates nitric oxide-dependent signaling, which modulates cardiac contractility. Nonetheless, the role of nitric oxide in ßAR-dependent regulation of Ca(2+) handling has not yet been explicated fully. OBJECTIVE: To elucidate the role of protein S-nitrosylation, a major transducer of nitric oxide bioactivity, on ßAR-dependent alterations in cardiomyocyte Ca(2+) handling and hypertrophy. METHODS AND RESULTS: Using transgenic mice to titrate the levels of protein S-nitrosylation, we uncovered major roles for protein S-nitrosylation, in general, and for phospholamban and cardiac troponin C S-nitrosylation, in particular, in ßAR-dependent regulation of Ca(2+) homeostasis. Notably, S-nitrosylation of phospholamban consequent upon ßAR stimulation is necessary for the inhibitory pentamerization of phospholamban, which activates sarcoplasmic reticulum Ca(2+)-ATPase and increases cytosolic Ca(2+) transients. Coincident S-nitrosylation of cardiac troponin C decreases myocardial sensitivity to Ca(2+). During chronic adrenergic stimulation, global reductions in cellular S-nitrosylation mitigate hypertrophic signaling resulting from Ca(2+) overload. CONCLUSIONS: S-Nitrosylation operates in concert with phosphorylation to regulate many cardiac Ca(2+)-handling proteins, including phospholamban and cardiac troponin C, thereby playing an essential and previously unrecognized role in cardiac Ca(2+) homeostasis. Manipulation of the S-nitrosylation level may prove therapeutic in heart failure.


Assuntos
Cálcio/metabolismo , Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Óxido Nítrico/metabolismo , Receptores Adrenérgicos beta/metabolismo , Agonistas Adrenérgicos beta/farmacologia , Aldeído Oxirredutases , Animais , Proteínas de Ligação ao Cálcio/genética , Proteínas de Ligação ao Cálcio/metabolismo , Células Cultivadas , Hipertrofia , Immunoblotting , Isoproterenol/farmacologia , Camundongos Knockout , Camundongos Transgênicos , Mutação , Miocárdio/patologia , Miócitos Cardíacos/citologia , Fosforilação , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático , Transdução de Sinais/efeitos dos fármacos , Troponina I/genética , Troponina I/metabolismo
6.
Trends Cardiovasc Med ; 23(8): 312-8, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23755896

RESUMO

Hypertension is an important modifiable risk factor for coronary heart disease, congestive heart failure, stroke, end-stage renal disease, and peripheral vascular disease, but many of the molecular mechanisms and genetic factors underlying the development of the most common forms of human hypertension remain to be defined. Abundant evidence suggests that nitric oxide (NO) and one of its primary targets, the cyclic guanosine monophosphate (cGMP)-generating enzyme soluble guanylate cyclase (sGC), have a critical role in regulating blood pressure. The availability of murine models of hypertension and the revolution in human genetics research (e.g., genome-wide association studies [GWAS]), resulting in the identification of dozens of genetic loci that affect normal variation in blood pressure and susceptibility to hypertension, provide a unique opportunity to dissect the mechanisms by which NO-cGMP signaling regulates blood pressure and to gain important insights into the pathogenesis of hypertension. In this review, we will give an overview of the current knowledge relating to the role of sGC in the regulation of blood pressure, discussing data obtained from genetically modified mouse models as well as from human genetic studies.


Assuntos
Pressão Sanguínea/fisiologia , Guanilato Ciclase/genética , Hipertensão , Receptores Citoplasmáticos e Nucleares/genética , Animais , Predisposição Genética para Doença , Estudo de Associação Genômica Ampla , Humanos , Hipertensão/genética , Hipertensão/metabolismo , Modelos Animais , Modelos Cardiovasculares , Óxido Nítrico/metabolismo , Transdução de Sinais , Guanilil Ciclase Solúvel
7.
Am J Physiol Heart Circ Physiol ; 304(8): H1134-46, 2013 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-23417863

RESUMO

Myocardial depression is an important contributor to morbidity and mortality in septic patients. Nitric oxide (NO) plays an important role in the development of septic cardiomyopathy, but also has protective effects. Recent evidence has indicated that NO exerts many of its downstream effects on the cardiovascular system via protein S-nitrosylation, which is negatively regulated by S-nitrosoglutathione reductase (GSNOR), an enzyme promoting denitrosylation. We tested the hypothesis that reducing cardiomyocyte S-nitrosylation by increasing GSNOR activity can improve myocardial dysfunction during sepsis. Therefore, we generated mice with a cardiomyocyte-specific overexpression of GSNOR (GSNOR-CMTg mice) and subjected them to endotoxic shock. Measurements of cardiac function in vivo and ex vivo showed that GSNOR-CMTg mice had a significantly improved cardiac function after lipopolysaccharide challenge (LPS, 50 mg/kg) compared with wild-type (WT) mice. Cardiomyocytes isolated from septic GSNOR-CMTg mice showed a corresponding improvement in contractility compared with WT cells. However, systolic Ca(2+) release was similarly depressed in both genotypes after LPS, indicating that GSNOR-CMTg cardiomyocytes have increased Ca(2+) sensitivity during sepsis. Parameters of inflammation were equally increased in LPS-treated hearts of both genotypes, and no compensatory changes in NO synthase expression levels were found in GSNOR-overexpressing hearts before or after LPS challenge. GSNOR overexpression however significantly reduced total cardiac protein S-nitrosylation during sepsis. Taken together, our results indicate that increasing the denitrosylation capacity of cardiomyocytes protects against sepsis-induced myocardial depression. Our findings suggest that specifically reducing protein S-nitrosylation during sepsis improves cardiac function by increasing cardiac myofilament sensitivity to Ca(2+).


Assuntos
Cardiomiopatias/prevenção & controle , Glutationa Redutase/metabolismo , Contração Miocárdica/fisiologia , Miócitos Cardíacos/metabolismo , Óxido Nítrico/metabolismo , Choque Séptico/metabolismo , Álcool Desidrogenase , Animais , Cálcio/metabolismo , Cardiomiopatias/etiologia , Cardiomiopatias/fisiopatologia , Glutationa Redutase/genética , Lipopolissacarídeos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Miocárdio/metabolismo , Miócitos Cardíacos/fisiologia , Óxido Nítrico Sintase/metabolismo , Choque Séptico/complicações , Choque Séptico/fisiopatologia
8.
Curr Hypertens Rep ; 15(1): 47-58, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23233080

RESUMO

The nitric oxide (NO)-cyclic guanosine monophosphate (cGMP) signaling system is a well-characterized modulator of cardiovascular function, in general, and blood pressure, in particular. The availability of mice mutant for key enzymes in the NO-cGMP signaling system facilitated the identification of interactions with other blood pressure modifying pathways (e.g. the renin-angiotensin-aldosterone system) and of gender-specific effects of impaired NO-cGMP signaling. In addition, recent genome-wide association studies identified blood pressure-modifying genetic variants in genes that modulate NO and cGMP levels. Together, these findings have advanced our understanding of how NO-cGMP signaling regulates blood pressure. In this review, we will summarize the results obtained in mice with disrupted NO-cGMP signaling and highlight the relevance of this pathway as a potential therapeutic target for the treatment of hypertension.


Assuntos
GMP Cíclico/fisiologia , Hipertensão/fisiopatologia , Óxido Nítrico/fisiologia , Animais , Pressão Sanguínea/fisiologia , GMP Cíclico/metabolismo , Proteínas Quinases Dependentes de GMP Cíclico/metabolismo , Endotélio Vascular/fisiologia , Estudo de Associação Genômica Ampla , Humanos , Camundongos , Camundongos Mutantes , Modelos Animais , Óxido Nítrico/metabolismo , Transdução de Sinais/fisiologia , Vasoconstrição/fisiologia , Vasodilatação/fisiologia
9.
J Clin Invest ; 122(6): 2316-25, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22565307

RESUMO

Nitric oxide (NO) plays an essential role in regulating hypertension and blood flow by inducing relaxation of vascular smooth muscle. Male mice deficient in a NO receptor component, the α1 subunit of soluble guanylate cyclase (sGCα1), are prone to hypertension in some, but not all, mouse strains, suggesting that additional genetic factors contribute to the onset of hypertension. Using linkage analyses, we discovered a quantitative trait locus (QTL) on chromosome 1 that was linked to mean arterial pressure (MAP) in the context of sGCα1 deficiency. This region is syntenic with previously identified blood pressure-related QTLs in the human and rat genome and contains the genes coding for renin. Hypertension was associated with increased activity of the renin-angiotensin-aldosterone system (RAAS). Further, we found that RAAS inhibition normalized MAP and improved endothelium-dependent vasorelaxation in sGCα1-deficient mice. These data identify the RAAS as a blood pressure-modifying mechanism in a setting of impaired NO/cGMP signaling.


Assuntos
Genoma Humano , Guanilato Ciclase/genética , Hipertensão/genética , Locos de Características Quantitativas , Receptores Citoplasmáticos e Nucleares/genética , Sistema Renina-Angiotensina/genética , Sistemas do Segundo Mensageiro/genética , Vasodilatação/genética , Animais , GMP Cíclico/genética , GMP Cíclico/metabolismo , Endotélio Vascular/enzimologia , Feminino , Ligação Genética , Guanilato Ciclase/metabolismo , Humanos , Hipertensão/enzimologia , Masculino , Camundongos , Camundongos Knockout , Ratos , Receptores Citoplasmáticos e Nucleares/metabolismo , Renina/genética , Renina/metabolismo , Guanilil Ciclase Solúvel , Especificidade da Espécie
10.
Circulation ; 124(15): 1645-53, 2011 Oct 11.
Artigo em Inglês | MEDLINE | ID: mdl-21931083

RESUMO

BACKGROUND: Sudden cardiac arrest (CA) is a leading cause of death worldwide. Breathing nitric oxide (NO) reduces ischemia/reperfusion injury in animal models and in patients. The objective of this study was to learn whether inhaled NO improves outcomes after CA and cardiopulmonary resuscitation (CPR). METHODS AND RESULTS: Adult male mice were subjected to potassium-induced CA for 7.5 minutes whereupon CPR was performed with chest compression and mechanical ventilation. One hour after CPR, mice were extubated and breathed air alone or air supplemented with 40 ppm NO for 23 hours. Mice that were subjected to CA/CPR and breathed air exhibited a poor 10-day survival rate (4 of 13), depressed neurological and left ventricular function, and increased caspase-3 activation and inflammatory cytokine induction in the brain. Magnetic resonance imaging revealed brain regions with marked water diffusion abnormality 24 hours after CA/CPR in mice that breathed air. Breathing air supplemented with NO for 23 hours starting 1 hour after CPR attenuated neurological and left ventricular dysfunction 4 days after CA/CPR and markedly improved 10-day survival rate (11 of 13; P=0.003 versus mice breathing air). The protective effects of inhaled NO on the outcome after CA/CPR were associated with reduced water diffusion abnormality, caspase-3 activation, and cytokine induction in the brain and increased serum nitrate/nitrite levels. Deficiency of the α1 subunit of soluble guanylate cyclase, a primary target of NO, abrogated the ability of inhaled NO to improve outcomes after CA/CPR. CONCLUSIONS: These results suggest that NO inhalation after CA and successful CPR improves outcome via soluble guanylate cyclase-dependent mechanisms.


Assuntos
Reanimação Cardiopulmonar , Parada Cardíaca/terapia , Óxido Nítrico/administração & dosagem , Administração por Inalação , Ar , Animais , Apoptose , Pressão Sanguínea , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Encéfalo/fisiopatologia , Caspase 3/metabolismo , Citocinas/antagonistas & inibidores , Citocinas/biossíntese , Difusão , Ativação Enzimática/efeitos dos fármacos , Guanilato Ciclase/química , Guanilato Ciclase/genética , Guanilato Ciclase/metabolismo , Coração/efeitos dos fármacos , Coração/fisiopatologia , Parada Cardíaca/mortalidade , Parada Cardíaca/patologia , Parada Cardíaca/fisiopatologia , Mediadores da Inflamação/antagonistas & inibidores , Imageamento por Ressonância Magnética/métodos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Sistema Nervoso/fisiopatologia , Nitratos/sangue , Nitritos/sangue , Respiração , Solubilidade , Taxa de Sobrevida , Fatores de Tempo , Função Ventricular Esquerda , Função Ventricular Direita , Água/metabolismo
11.
Basic Res Cardiol ; 106(4): 635-43, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21394564

RESUMO

Nitric oxide (NO)-dependent soluble guanylate cyclase (sGC) activation is an important component of cardiac signal transduction pathways, including the cardioprotective signaling cascade induced by ischemic preconditioning (IPC). The sGCα subunit, which binds to the common sGCß1 subunit, exists in two different isoforms, sGCα1 and sGCα2, but their relative physiological roles remain unknown. In the present study, we studied Langendorff-perfused isolated hearts of genetically engineered mice lacking functional sGCα1 (sGCα1KO mice), which is the predominant isoform in the heart. Our results show that the loss of sGCα1 has a positive inotropic and lusitropic effect on basal cardiac function, indicating an important role for sGCα1 in regulating basal myocardial contractility. Surprisingly, IPC led to a similar 35-40% reduction in infarct size and concomitant protein kinase Cε (PKCε) phosphorylation in both wild-type (WT) and sGCα1KO hearts subjected to 40 min of global ischemia and reperfusion. Inhibition of the activation of all sGC isoforms by 1H-[1,2,4]oxadiazolo[4,3-a]quinoxaline-1-one (ODQ, 10 µmol/L) completely abolished the protection by IPC in WT and sGCα1KO hearts. NO-stimulated cGMP production was severely attenuated in sGCα1KO hearts compared to WT hearts, indicating that the sGCα2 isoform only produces minute amounts of cGMP after NO stimulation. Taken together, our results indicate that although sGCα1 importantly regulates cardiac contractility, it is not required for cardioprotection by IPC. Instead, our results suggest that possibly only minimal sGC activity, which in sGCα1KO hearts is provided by the sGCα2 isoform, is sufficient to transduce the cardioprotective signal induced by IPC via phosphorylation of PKCε.


Assuntos
Guanilato Ciclase/fisiologia , Precondicionamento Isquêmico Miocárdico , Contração Miocárdica , Animais , GMP Cíclico/biossíntese , Isoenzimas/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Miócitos Cardíacos/enzimologia
12.
Circulation ; 120(10): 888-96, 2009 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-19704099

RESUMO

BACKGROUND: Sudden cardiac arrest (CA) is one of the leading causes of death worldwide. We sought to evaluate the impact of hydrogen sulfide (H(2)S) on the outcome after CA and cardiopulmonary resuscitation (CPR) in mouse. METHODS AND RESULTS: Mice were subjected to 8 minutes of normothermic CA and resuscitated with chest compression and mechanical ventilation. Seven minutes after the onset of CA (1 minute before CPR), mice received sodium sulfide (Na(2)S) (0.55 mg/kg IV) or vehicle 1 minute before CPR. There was no difference in the rate of return of spontaneous circulation, CPR time to return of spontaneous circulation, and left ventricular function at return of spontaneous circulation between groups. Administration of Na(2)S 1 minute before CPR markedly improved survival rate at 24 hours after CPR (15/15) compared with vehicle (10/26; P=0.0001 versus Na(2)S). Administration of Na(2)S prevented CA/CPR-induced oxidative stress and ameliorated left ventricular and neurological dysfunction 24 hours after CPR. Delayed administration of Na(2)S at 10 minutes after CPR did not improve outcomes after CA/CPR. Cardioprotective effects of Na(2)S were confirmed in isolated-perfused mouse hearts subjected to global ischemia and reperfusion. Cardiomyocyte-specific overexpression of cystathionine gamma-lyase (an enzyme that produces H(2)S) markedly improved outcomes of CA/CPR. Na(2)S increased phosphorylation of nitric oxide synthase 3 in left ventricle and brain cortex, increased serum nitrite/nitrate levels, and attenuated CA-induced mitochondrial injury and cell death. Nitric oxide synthase 3 deficiency abrogated the protective effects of Na(2)S on the outcome of CA/CPR. CONCLUSIONS: These results suggest that administration of Na(2)S at the time of CPR improves outcome after CA possibly via a nitric oxide synthase 3-dependent signaling pathway.


Assuntos
Reanimação Cardiopulmonar , Parada Cardíaca/enzimologia , Parada Cardíaca/terapia , Sulfeto de Hidrogênio/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Apoptose/efeitos dos fármacos , Encéfalo/fisiopatologia , Reanimação Cardiopulmonar/efeitos adversos , Cardiotônicos/farmacologia , Cistationina gama-Liase/metabolismo , Coração/efeitos dos fármacos , Coração/fisiopatologia , Parada Cardíaca/mortalidade , Parada Cardíaca/fisiopatologia , Técnicas In Vitro , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias Cardíacas/efeitos dos fármacos , Contração Miocárdica , Traumatismo por Reperfusão Miocárdica/fisiopatologia , Miócitos Cardíacos/enzimologia , Sistema Nervoso/efeitos dos fármacos , Sistema Nervoso/fisiopatologia , Óxido Nítrico Sintase Tipo III/deficiência , Estresse Oxidativo/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Sulfetos/farmacologia , Taxa de Sobrevida , Regulação para Cima
13.
Am J Physiol Heart Circ Physiol ; 297(2): H654-63, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19502556

RESUMO

Altered cGMP signaling has been implicated in myocardial depression, morbidity, and mortality associated with sepsis. Previous studies, using inhibitors of soluble guanylate cyclase (sGC), suggested that cGMP generated by sGC contributed to the cardiac dysfunction and mortality associated with sepsis. We used sGC(alpha)(1)-deficient (sGC(alpha)(1)(-/-)) mice to unequivocally determine the role of sGC(alpha)(1)beta(1) in the development of cardiac dysfunction and death associated with two models of inflammatory shock: endotoxin- and TNF-induced shock. At baseline, echocardiographic assessment and invasive hemodynamic measurements of left ventricular (LV) dimensions and function did not differ between wild-type (WT) mice and sGC(alpha)(1)(-/-) mice on the C57BL/6 background (sGC(alpha)(1)(-/-B6) mice). At 14 h after endotoxin challenge, cardiac dysfunction was more pronounced in sGC(alpha)(1)(-/-B6) than WT mice, as assessed using echocardiographic and hemodynamic indexes of LV function. Similarly, Ca(2+) handling and cell shortening were impaired to a greater extent in cardiomyocytes isolated from sGC(alpha)(1)(-/-B6) than WT mice after endotoxin challenge. Importantly, morbidity and mortality associated with inflammatory shock induced by endotoxin or TNF were increased in sGC(alpha)(1)(-/-B6) compared with WT mice. Together, these findings suggest that cGMP generated by sGC(alpha)(1)beta(1) protects against cardiac dysfunction and mortality in murine inflammatory shock models.


Assuntos
Guanilato Ciclase/genética , Guanilato Ciclase/metabolismo , Receptores Citoplasmáticos e Nucleares/genética , Receptores Citoplasmáticos e Nucleares/metabolismo , Choque Cardiogênico , Disfunção Ventricular Esquerda , Animais , Pressão Sanguínea/fisiologia , Cálcio/metabolismo , Células Cultivadas , Modelos Animais de Doenças , Ecocardiografia , Endotoxinas/toxicidade , Ativação Enzimática/fisiologia , Regulação Enzimológica da Expressão Gênica/fisiologia , Estimativa de Kaplan-Meier , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Contração Miocárdica/fisiologia , Miócitos Cardíacos/citologia , Miócitos Cardíacos/fisiologia , Óxido Nítrico/metabolismo , Sepse/imunologia , Sepse/metabolismo , Sepse/mortalidade , Choque Cardiogênico/imunologia , Choque Cardiogênico/metabolismo , Choque Cardiogênico/mortalidade , Guanilil Ciclase Solúvel , Fator de Necrose Tumoral alfa/toxicidade , Disfunção Ventricular Esquerda/diagnóstico por imagem , Disfunção Ventricular Esquerda/imunologia , Disfunção Ventricular Esquerda/mortalidade , Pressão Ventricular/fisiologia
14.
Crit Care Med ; 37(1): 256-62, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19050616

RESUMO

OBJECTIVES: Despite advances in resuscitation methods, survival after out-of-hospital cardiac arrest remains low, at least in part, due to postcardiac arrest circulatory and neurologic failure. To elucidate the role of nitric oxide (NO) in the recovery from cardiac arrest and cardiopulmonary resuscitation (CPR), we studied the impact of NO synthase (NOS3)/cGMP signaling on cardiac and neurologic outcomes after cardiac arrest and CPR. DESIGN: Prospective, randomized, controlled study. SETTING: Animal research laboratory. SUBJECTS: Mice. INTERVENTIONS: Female wild-type (WT) mice, NOS3-deficient mice (NOS3-/-), NOS3-/- mice with cardiomyocyte-specific overexpression of NOS3 (NOS3-/-CSTg), and mice deficient for soluble guanylate cyclase alpha1 (sGCalpha1-/-) were subjected to potassium-induced cardiac arrest (9 min) followed by CPR. Cardiac and neurologic function and survival were assessed up to 24 hrs post-CPR. MEASUREMENTS AND MAIN RESULTS: Cardiac arrest and CPR markedly depressed myocardial function in NOS3-/- and sGCalpha1-/- but not in WT and NOS3-/-CSTg. Neurologic function score and 24 hrs survival rate was lower in NOS3-/- and sGCalpha1-/- compared with WT and NOS3-/-CSTg. Detrimental effects of deficiency of NOS3 or sGCalpha1 were associated with enhanced inflammation of heart and liver and increased cell death in heart, liver, and brain that were largely prevented by cardiomyocyte-restricted NOS3 overexpression. CONCLUSIONS: These results demonstrate an important salutary impact of NOS3/sGC signaling on the outcome of cardiac arrest. Myocardial NOS3 prevented postcardiac arrest myocardial dysfunction, attenuated end-organ damage, and improved neurologic outcome and survival. Our observations suggest that enhancement of cardiac NOS3 and/or sGC activity may improve outcome after cardiac arrest and CPR.


Assuntos
Reanimação Cardiopulmonar , Guanilato Ciclase/fisiologia , Parada Cardíaca/terapia , Óxido Nítrico Sintase Tipo III/fisiologia , Receptores Citoplasmáticos e Nucleares/fisiologia , Animais , Feminino , Camundongos , Guanilil Ciclase Solúvel , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...