Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Am J Physiol Heart Circ Physiol ; 309(8): H1271-80, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26254336

RESUMO

Levels of the HER2/ErbB2 protein in the heart are upregulated in some women during breast cancer therapy, and these women are at high risk for developing heart dysfunction after sequential treatment with anti-ErbB2/trastuzumab or doxorubicin. Doxorubicin is known to increase oxidative stress in the heart, and thus we considered the possibility that ErbB2 protein influences the status of cardiac antioxidant defenses in cardiomyocytes. In this study, we measured reactive oxygen species (ROS) in cardiac mitochondria and whole hearts from mice with cardiac-specific overexpression of ErbB2 (ErbB2(tg)) and found that, compared with control mice, high levels of ErbB2 in myocardium result in lower levels of ROS in mitochondria (P = 0.0075) and whole hearts (P = 0.0381). Neonatal cardiomyocytes isolated from ErbB2(tg) hearts have lower ROS levels and less cellular death (P < 0.0001) following doxorubicin treatment. Analyzing antioxidant enzyme levels and activities, we found that ErbB2(tg) hearts have increased levels of glutathione peroxidase 1 (GPx1) protein (P < 0.0001) and GPx activity (P = 0.0031) in addition to increased levels of two known GPx activators, c-Abl (P = 0.0284) and Arg (P < 0.0001). Interestingly, although mitochondrial ROS emission is reduced in the ErbB2(tg) hearts, oxygen consumption rates and complex I activity are similar to control littermates. Compared with these in vivo studies, H9c2 cells transfected with ErbB2 showed less cellular toxicity and produced less ROS (P < 0.0001) after doxorubicin treatment but upregulated GR activity (P = 0.0237) instead of GPx. Our study shows that ErbB2-dependent signaling contributes to antioxidant defenses and suggests a novel mechanism by which anticancer therapies involving ErbB2 antagonists can harm myocardial structure and function.


Assuntos
Antineoplásicos/toxicidade , Antioxidantes/metabolismo , Doxorrubicina/toxicidade , Glutationa Peroxidase/metabolismo , Cardiopatias/prevenção & controle , Mitocôndrias Cardíacas/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Receptor ErbB-2/metabolismo , Animais , Animais Recém-Nascidos , Morte Celular/efeitos dos fármacos , Linhagem Celular , Relação Dose-Resposta a Droga , Glutationa Redutase/metabolismo , Cardiopatias/induzido quimicamente , Cardiopatias/enzimologia , Cardiopatias/genética , Cardiopatias/patologia , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mitocôndrias Cardíacas/enzimologia , Mitocôndrias Cardíacas/patologia , Miócitos Cardíacos/enzimologia , Miócitos Cardíacos/patologia , Proteínas Tirosina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-abl/metabolismo , Ratos , Receptor ErbB-2/genética , Glutationa Peroxidase GPX1
2.
Clin Sci (Lond) ; 129(7): 561-74, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26186741

RESUMO

In Type I diabetic (T1DM) patients, both peaks of hyperglycaemia and increased sympathetic tone probably contribute to impair systolic and diastolic function. However, how these stressors eventually alter cardiac function during T1DM is not fully understood. In the present study, we hypothesized that impaired mitochondrial energy supply and excess reactive oxygen species (ROS) emission is centrally involved in T1DM cardiac dysfunction due to metabolic/redox stress and aimed to determine the mitochondrial sites implicated in these alterations. To this end, we used isolated myocytes and mitochondria from Sham and streptozotocin (STZ)-induced T1DM guinea pigs (GPs), untreated or treated with insulin. Relative to controls, T1DM myocytes exhibited higher oxidative stress when challenged with high glucose (HG) combined with ß-adrenergic stimulation [via isoprenaline (isoproterenol) (ISO)], leading to contraction/relaxation deficits. T1DM mitochondria had decreased respiration with complex II and IV substrates and markedly lower ADP phosphorylation rates and higher H2O2 emission when challenged with oxidants to mimic the more oxidized redox milieu present in HG + ISO-treated cardiomyocytes. Since in T1DM hearts insulin-sensitivity is preserved and a glucose-to-fatty acid (FA) shift occurs, we next tested whether insulin therapy or acute palmitate (Palm) infusion prevents HG + ISO-induced cardiac dysfunction. We found that insulin rescued proper cardiac redox balance, but not mitochondrial respiration or contractile performance. Conversely, Palm restored redox balance and preserved myocyte function. Thus, stressors such as peaks of HG and adrenergic hyperactivity impair mitochondrial respiration, hampering energy supply while exacerbating ROS emission. Our study suggests that an ideal therapeutic measure to treat metabolically/redox-challenged T1DM hearts should concomitantly correct energetic and redox abnormalities to fully maintain cardiac function.


Assuntos
Diabetes Mellitus Tipo 1/metabolismo , Peróxido de Hidrogênio/química , Mitocôndrias/metabolismo , Animais , Glicemia/metabolismo , Cálcio/metabolismo , Modelos Animais de Doenças , Ácidos Graxos/metabolismo , Cobaias , Insulina/metabolismo , Masculino , Microscopia de Fluorescência , Mitocôndrias Cardíacas/metabolismo , Células Musculares/citologia , Contração Muscular , Miócitos Cardíacos/metabolismo , Oxirredução , Oxigênio/metabolismo , Fosforilação , Espécies Reativas de Oxigênio/metabolismo , Receptores Adrenérgicos beta/metabolismo , Sarcômeros/metabolismo
3.
J Biol Chem ; 286(38): 33669-77, 2011 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-21832082

RESUMO

Respiring mitochondria produce H(2)O(2) continuously. When production exceeds scavenging, H(2)O(2) emission occurs, endangering cell functions. The mitochondrial peroxidase peroxiredoxin-3 reduces H(2)O(2) to water using reducing equivalents from NADPH supplied by thioredoxin-2 (Trx2) and, ultimately, thioredoxin reductase-2 (TrxR2). Here, the contribution of this mitochondrial thioredoxin system to the control of H(2)O(2) emission was studied in isolated mitochondria and cardiomyocytes from mouse or guinea pig heart. Energization of mitochondria by the addition of glutamate/malate resulted in a 10-fold decrease in the ratio of oxidized to reduced Trx2. This shift in redox state was accompanied by an increase in NAD(P)H and was dependent on TrxR2 activity. Inhibition of TrxR2 in isolated mitochondria by auranofin resulted in increased H(2)O(2) emission, an effect that was seen under both forward and reverse electron transport. This effect was independent of changes in NAD(P)H or membrane potential. The effects of auranofin were reproduced in cardiomyocytes; superoxide and H(2)O(2) levels increased, but similarly, there was no effect on NAD(P)H or membrane potential. These data show that energization of mitochondria increases the antioxidant potential of the TrxR2/Trx2 system and that inhibition of TrxR2 results in increased H(2)O(2) emission through a mechanism that is independent of changes in other redox couples.


Assuntos
Peróxido de Hidrogênio/metabolismo , Mitocôndrias Cardíacas/enzimologia , Tiorredoxina Redutase 2/metabolismo , Animais , Auranofina/farmacologia , Dinitroclorobenzeno/farmacologia , Transporte de Elétrons/efeitos dos fármacos , Metabolismo Energético/efeitos dos fármacos , Ensaios Enzimáticos , Glutationa/metabolismo , Cobaias , Camundongos , Mitocôndrias Cardíacas/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Oxirredução/efeitos dos fármacos , Peroxirredoxina III/metabolismo , Tiorredoxina Redutase 2/antagonistas & inibidores , Tiorredoxinas/metabolismo
4.
BJU Int ; 110(10): 1455-62, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22508007

RESUMO

OBJECTIVES: To examine the acute effects of sunitinib on inotropic function, intracellular Ca(2+) transients, myofilament Ca(2+) sensitivity and generation of reactive oxygen species (ROS) in human multicellular myocardium and isolated mouse cardiomyocytes. To search for microRNAs as suitable biomarkers for indicating toxic cardiac effects. PATIENTS AND METHODS: After exposure to sunitinib (0.1-10 µg/mL) developed force, diastolic tension and kinetic variables were assessed in isolated human myocardium. Changes in myocyte sarcomere length, whole-cell calcium transients, myofilament force-Ca(2+) relationship, and ROS generation were examined in isolated ventricular mouse cardiomyocytes. Microarray and realtime-PCR were used to screen for differentially expressed microRNAs in cultured cardiomyocytes that were exposed for 24 h to sunitinib. RESULTS: We found that higher concentrations of sunitinib (1 and 10 µg/mL) decreased developed force at 30 minutes 76.9 + 2.8 and 54.5 + 6.3%, compared to 96.1 + 2.6% in controls (P < 0.01). Sunitinib exposure significantly decreased sarcomere shortening and Ca2+ transients. Myofilament Ca(2+) sensitivity was not altered, while ROS levels were significantly increased after exposure to the drug. MicroRNA expression patterns were not altered by sunitinib. CONCLUSIONS: Sunitinib elicits a dose-dependent negative inotropic effect in myocardium, accompanied by a decline in intracellular Ca(2+) and increased ROS generation. In clinical practice, these cardiotoxic effects should be considered in cases where cardiac concentrations of sunitinib could be increased.


Assuntos
Antineoplásicos/efeitos adversos , Indóis/efeitos adversos , Contração Miocárdica/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Inibidores de Proteínas Quinases/efeitos adversos , Pirróis/efeitos adversos , Idoso , Animais , Cálcio/metabolismo , Relação Dose-Resposta a Droga , Espectroscopia de Ressonância de Spin Eletrônica , Feminino , Coração/efeitos dos fármacos , Humanos , Técnicas In Vitro , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Miócitos Cardíacos/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Sarcômeros/efeitos dos fármacos , Sunitinibe
5.
J Pharmacol Exp Ther ; 339(3): 825-31, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21865439

RESUMO

Isoflurane (ISO) is known to depress cardiac contraction. Here, we hypothesized that decreasing myofilament Ca(2+) responsiveness is central to ISO-induced reduction in cardiac force development. Moreover, we also tested whether the nitroxyl (HNO) donor 1-nitrosocyclohexyl acetate (NCA), acting as a myofilament Ca(2+) sensitizer, restores force in the presence of ISO. Trabeculae from the right ventricles of LBN/F1 rats were superfused with Krebs-Henseleit solution at room temperature, and force and intracellular Ca(2+) ([Ca(2+)](i)) were measured. Steady-state activations were achieved by stimulating the muscles at 10 Hz in the presence of ryanodine. The same muscles were chemically skinned with 1% Triton X-100, and the force-Ca(2+) relation measurements were repeated. ISO depressed force in a dose-dependent manner without significantly altering [Ca(2+)](i). At 1.5%, force was reduced over 50%, whereas [Ca(2+)](i) remained unaffected. At 3%, contraction was decreased by ∼75% with [Ca(2+)](i) reduced by only 15%. During steady-state activation, 1.5% ISO depressed maximal Ca(2+)-activated force (F(max)) and increased the [Ca(2+)](i) required for 50% activation (Ca(50)) without affecting the Hill coefficient. After skinning, the same muscles showed similar decreases in F(max) and increases in Ca(50) in the presence of ISO. NCA restored force in the presence of ISO without affecting [Ca(2+)](i). These results show that 1) ISO depresses cardiac force development by decreasing myofilament Ca(2+) responsiveness, and 2) myofilament Ca(2+) sensitization by NCA can effectively restore force development without further increases in [Ca(2+)](i). The present findings have potential translational value because of the efficiency and efficacy of HNO on ISO-induced myocardial contractile dysfunction.


Assuntos
Anestésicos Inalatórios/toxicidade , Cálcio/fisiologia , Ventrículos do Coração/efeitos dos fármacos , Isoflurano/toxicidade , Contração Miocárdica/efeitos dos fármacos , Miofibrilas/efeitos dos fármacos , Óxidos de Nitrogênio/metabolismo , Acetatos/farmacologia , Anestésicos Inalatórios/farmacologia , Animais , Cardiotônicos/antagonistas & inibidores , Avaliação Pré-Clínica de Medicamentos , Radicais Livres/metabolismo , Glucose , Isoflurano/farmacologia , Contração Miocárdica/fisiologia , Miofibrilas/fisiologia , Compostos Nitrosos/farmacologia , Ratos , Rianodina/farmacologia , Canal de Liberação de Cálcio do Receptor de Rianodina/fisiologia , Trometamina , Função Ventricular/efeitos dos fármacos
6.
J Am Heart Assoc ; 5(3): e003208, 2016 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-27001967

RESUMO

BACKGROUND: Nitric oxide synthase uncoupling occurs under conditions of oxidative stress modifying the enzyme's function so it generates superoxide rather than nitric oxide. Nitric oxide synthase uncoupling occurs with chronic pressure overload, and both are ameliorated by exogenous tetrahydrobiopterin (BH4)-a cofactor required for normal nitric oxide synthase function-supporting a pathophysiological link. Genetically augmenting BH4 synthesis in endothelial cells fails to replicate this benefit, indicating that other cell types dominate the effects of exogenous BH4 administration. We tested whether the primary cellular target of BH4 is the cardiomyocyte or whether other novel mechanisms are invoked. METHODS AND RESULTS: Mice with cardiomyocyte-specific overexpression of GTP cyclohydrolase 1 (mGCH1) and wild-type littermates underwent transverse aortic constriction. The mGCH1 mice had markedly increased myocardial BH4 and, unlike wild type, maintained nitric oxide synthase coupling after transverse aortic constriction; however, the transverse aortic constriction-induced abnormalities in cardiac morphology and function were similar in both groups. In contrast, exogenous BH4 supplementation improved transverse aortic constricted hearts in both groups, suppressed multiple inflammatory cytokines, and attenuated infiltration of inflammatory macrophages into the heart early after transverse aortic constriction. CONCLUSIONS: BH4 protection against adverse remodeling in hypertrophic cardiac disease is not driven by its prevention of myocardial nitric oxide synthase uncoupling, as presumed previously. Instead, benefits from exogenous BH4 are mediated by a protective effect coupled to suppression of inflammatory pathways and myocardial macrophage infiltration.


Assuntos
Anti-Inflamatórios/farmacologia , Biopterinas/análogos & derivados , Fármacos Cardiovasculares/farmacologia , Hipertrofia Ventricular Esquerda/prevenção & controle , Miócitos Cardíacos/efeitos dos fármacos , Óxido Nítrico Sintase/metabolismo , Função Ventricular Esquerda/efeitos dos fármacos , Remodelação Ventricular/efeitos dos fármacos , Animais , Biopterinas/farmacologia , Citocinas/metabolismo , Citoproteção , Modelos Animais de Doenças , GTP Cicloidrolase/genética , GTP Cicloidrolase/metabolismo , Humanos , Hipertrofia Ventricular Esquerda/enzimologia , Hipertrofia Ventricular Esquerda/patologia , Hipertrofia Ventricular Esquerda/fisiopatologia , Mediadores da Inflamação/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos Transgênicos , Miócitos Cardíacos/enzimologia , Miócitos Cardíacos/patologia , Óxido Nítrico/metabolismo , Oxirredução , Transdução de Sinais , Superóxidos/metabolismo , Fatores de Tempo , Disfunção Ventricular Esquerda/enzimologia , Disfunção Ventricular Esquerda/fisiopatologia , Disfunção Ventricular Esquerda/prevenção & controle
7.
Antioxid Redox Signal ; 20(2): 267-80, 2014 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-23581564

RESUMO

AIMS: Monoamine oxidases (MAOs) are mitochondrial flavoenzymes responsible for neurotransmitter and biogenic amines catabolism. MAO-A contributes to heart failure progression via enhanced norepinephrine catabolism and oxidative stress. The potential pathogenetic role of the isoenzyme MAO-B in cardiac diseases is currently unknown. Moreover, it is has not been determined yet whether MAO activation can directly affect mitochondrial function. RESULTS: In wild type mice, pressure overload induced by transverse aortic constriction (TAC) resulted in enhanced dopamine catabolism, left ventricular (LV) remodeling, and dysfunction. Conversely, mice lacking MAO-B (MAO-B(-/-)) subjected to TAC maintained concentric hypertrophy accompanied by extracellular signal regulated kinase (ERK)1/2 activation, and preserved LV function, both at early (3 weeks) and late stages (9 weeks). Enhanced MAO activation triggered oxidative stress, and dropped mitochondrial membrane potential in the presence of ATP synthase inhibitor oligomycin both in neonatal and adult cardiomyocytes. The MAO-B inhibitor pargyline completely offset this change, suggesting that MAO activation induces a latent mitochondrial dysfunction, causing these organelles to hydrolyze ATP. Moreover, MAO-dependent aldehyde formation due to inhibition of aldehyde dehydrogenase 2 activity also contributed to alter mitochondrial bioenergetics. INNOVATION: Our study unravels a novel role for MAO-B in the pathogenesis of heart failure, showing that both MAO-driven reactive oxygen species production and impaired aldehyde metabolism affect mitochondrial function. CONCLUSION: Under conditions of chronic hemodynamic stress, enhanced MAO-B activity is a major determinant of cardiac structural and functional disarrangement. Both increased oxidative stress and the accumulation of aldehyde intermediates are likely liable for these adverse morphological and mechanical changes by directly targeting mitochondria.


Assuntos
Mitocôndrias Cardíacas/metabolismo , Monoaminoxidase/metabolismo , Miócitos Cardíacos/metabolismo , Disfunção Ventricular/metabolismo , Disfunção Ventricular/fisiopatologia , Aldeídos/metabolismo , Animais , Apoptose/genética , Pressão Sanguínea , Cardiomegalia/genética , Cardiomegalia/metabolismo , Cardiomegalia/patologia , Cardiomegalia/fisiopatologia , Dopamina/metabolismo , Ativação Enzimática , Fibrose , Camundongos , Camundongos Knockout , Mitocôndrias Cardíacas/genética , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Monoaminoxidase/genética , Miócitos Cardíacos/patologia , Oxirredução , Estresse Oxidativo/genética , Fosforilação , Ratos , Espécies Reativas de Oxigênio/metabolismo , Disfunção Ventricular/genética , Função Ventricular Esquerda
8.
Antioxid Redox Signal ; 18(9): 1078-99, 2013 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-22871241

RESUMO

SIGNIFICANCE: The regulation of myocardial function by constitutive nitric oxide synthases (NOS) is important for the maintenance of myocardial Ca(2+) homeostasis, relaxation and distensibility, and protection from arrhythmia and abnormal stress stimuli. However, sustained insults such as diabetes, hypertension, hemodynamic overload, and atrial fibrillation lead to dysfunctional NOS activity with superoxide produced instead of NO and worse pathophysiology. RECENT ADVANCES: Major strides in understanding the role of normal and abnormal constitutive NOS in the heart have revealed molecular targets by which NO modulates myocyte function and morphology, the role and nature of post-translational modifications of NOS, and factors controlling nitroso-redox balance. Localized and differential signaling from NOS1 (neuronal) versus NOS3 (endothelial) isoforms are being identified, as are methods to restore NOS function in heart disease. CRITICAL ISSUES: Abnormal NOS signaling plays a key role in many cardiac disorders, while targeted modulation may potentially reverse this pathogenic source of oxidative stress. FUTURE DIRECTIONS: Improvements in the clinical translation of potent modulators of NOS function/dysfunction may ultimately provide a powerful new treatment for many hearts diseases that are fueled by nitroso-redox imbalance.


Assuntos
Insuficiência Cardíaca/enzimologia , Óxido Nítrico Sintase/fisiologia , Animais , Arginase/antagonistas & inibidores , Arginase/metabolismo , Comunicação Autócrina , Biopterinas/análogos & derivados , Biopterinas/fisiologia , Sinalização do Cálcio , GMP Cíclico/fisiologia , Diabetes Mellitus/metabolismo , Progressão da Doença , Ativação Enzimática , Indução Enzimática , Cardiopatias/complicações , Cardiopatias/enzimologia , Cardiopatias/fisiopatologia , Humanos , Hipertensão/complicações , Hipertensão/metabolismo , Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Óxido Nítrico/fisiologia , Óxido Nítrico Sintase/química , Comunicação Parácrina , Processamento de Proteína Pós-Traducional , Estrutura Terciária de Proteína , Transporte Proteico , Transdução de Sinais/fisiologia , Superóxidos/metabolismo
9.
Antioxid Redox Signal ; 19(11): 1185-97, 2013 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-23919584

RESUMO

AIMS: Nitroxyl (HNO) interacts with thiols to act as a redox-sensitive modulator of protein function. It enhances sarcoplasmic reticular Ca(2+) uptake and myofilament Ca(2+) sensitivity, improving cardiac contractility. This activity has led to clinical testing of HNO donors for heart failure. Here we tested whether HNO alters the inhibitory interaction between phospholamban (PLN) and the sarcoplasmic reticulum Ca(2+)-ATPase (SERCA2a) in a redox-dependent manner, improving Ca(2+) handling in isolated myocytes/hearts. RESULTS: Ventriculocytes, sarcoplasmic reticulum (SR) vesicles, and whole hearts were isolated from control (wildtype [WT]) or PLN knockout (pln(-/-)) mice. Compared to WT, pln(-/-) myocytes displayed enhanced resting sarcomere shortening, peak Ca(2+) transient, and blunted ß-adrenergic responsiveness. HNO stimulated shortening, relaxation, and Ca(2+) transient in WT cardiomyocytes, and evoked positive inotropy/lusitropy in intact hearts. These changes were markedly blunted in pln(-/-) cells/hearts. HNO enhanced SR Ca(2+) uptake in WT but not pln(-/-) SR-vesicles. Spectroscopic studies in insect cell microsomes expressing SERCA2a±PLN showed that HNO increased Ca(2+)-dependent SERCA2a conformational flexibility but only when PLN was present. In cardiomyocytes, HNO achieved this effect by stabilizing PLN in an oligomeric disulfide bond-dependent configuration, decreasing the amount of free inhibitory monomeric PLN available. INNOVATION: HNO-dependent redox changes in myocyte PLN oligomerization relieve PLN inhibition of SERCA2a. CONCLUSIONS: PLN plays a central role in HNO-induced enhancement of SERCA2a activity, leading to increased inotropy/lusitropy in intact myocytes and hearts. PLN remains physically associated with SERCA2a; however, less monomeric PLN is available resulting in decreased inhibition of the enzyme. These findings offer new avenues to improve Ca(2+) handling in failing hearts.


Assuntos
Antioxidantes/farmacologia , Proteínas de Ligação ao Cálcio/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Óxidos de Nitrogênio/farmacologia , Multimerização Proteica/efeitos dos fármacos , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Cálcio/metabolismo , Sinalização do Cálcio/efeitos dos fármacos , Proteínas de Ligação ao Cálcio/química , Proteínas de Ligação ao Cálcio/genética , Cardiotônicos/farmacologia , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Dissulfetos , Ventrículos do Coração/efeitos dos fármacos , Ventrículos do Coração/metabolismo , Técnicas In Vitro , Camundongos , Camundongos Knockout , Microssomos/metabolismo , Oxirredução/efeitos dos fármacos , Fosforilação , Ligação Proteica , Conformação Proteica/efeitos dos fármacos , Domínios e Motivos de Interação entre Proteínas , Estabilidade Proteica/efeitos dos fármacos , Retículo Sarcoplasmático/metabolismo , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/química
10.
Int J Biochem Cell Biol ; 44(12): 2106-15, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22939972

RESUMO

Previously we reported that the sesquiterpene lactone parthenolide induces oxidative stress in cardiac myocytes, which blocks Janus kinase (JAK) activation by the interleukin 6 (IL-6)-type cytokines. One implication suggested by this finding is that IL-6 signaling is dependent upon cellular anti-oxidant defenses or redox status. Therefore, the present study was undertaken to directly test the hypothesis that JAK1 signaling by the IL-6-type cytokines in cardiac myocytes is impaired by glutathione (GSH) depletion, since this tripeptide is one of the major anti-oxidant molecules and redox-buffers in cells. Cardiac myocytes were pretreated for 6h with l-buthionine-sulfoximine (BSO) to inhibit GSH synthesis. After 24h, cells were dosed with the IL-6-like cytokine, leukemia inhibitory factor (LIF). BSO treatment decreased GSH levels and dose-dependently attenuated activation of JAK1, Signal Transducer and Activator of Transcription 3 (STAT3), and extracellular signal regulated kinases 1 and 2 (ERK1/2). Addition of glutathione monoethyl ester, which is cleaved intracellularly to GSH, prevented attenuation of LIF-induced JAK1 and STAT3 activation, as did the reductant N-acetyl-cysteine. Unexpectedly, LIF-induced STAT1 activation was unaffected by GSH depletion. Evidence was found that STAT3 is more resistant than STAT1 to intermolecular disulfide bond formation under oxidizing conditions and more likely to retain the monomeric form, suggesting that conformational differences explain the differential effect of GSH depletion on STAT1 and STAT3. Overall, our findings indicate that activation of both JAK1 and STAT3 is redox-sensitive and the character of IL-6 type cytokine signaling in cardiac myocytes is sensitive to changes in the cellular redox status. In cardiac myocytes, activation of STAT1 may be favored over STAT3 under oxidizing conditions due to GSH depletion and/or augmented reactive oxygen species production, such as in ischemia-reperfusion and heart failure.


Assuntos
Glutationa/metabolismo , Janus Quinase 1/metabolismo , Fator Inibidor de Leucemia/metabolismo , Miócitos Cardíacos/metabolismo , Fator de Transcrição STAT3/metabolismo , Acetilcisteína/farmacologia , Animais , Butionina Sulfoximina/farmacologia , Células Cultivadas , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Sequestradores de Radicais Livres/farmacologia , Glutationa/fisiologia , Fator Inibidor de Leucemia/fisiologia , Sistema de Sinalização das MAP Quinases , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Miócitos Cardíacos/efeitos dos fármacos , Estresse Oxidativo , Ligação Proteica , Ratos , Ratos Sprague-Dawley , Fator de Transcrição STAT1/metabolismo
11.
J Gen Physiol ; 139(6): 479-91, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22585969

RESUMO

The net emission of hydrogen peroxide (H(2)O(2)) from mitochondria results from the balance between reactive oxygen species (ROS) continuously generated in the respiratory chain and ROS scavenging. The relative contribution of the two major antioxidant systems in the mitochondrial matrix, glutathione (GSH) and thioredoxin (Trx), has not been assessed. In this paper, we examine this key question via combined experimental and theoretical approaches, using isolated heart mitochondria from mouse, rat, and guinea pig. As compared with untreated control mitochondria, selective inhibition of Trx reductase with auranofin along with depletion of GSH with 2,4-dinitrochlorobenzene led to a species-dependent increase in H(2)O(2) emission flux of 17, 11, and 6 fold in state 4 and 15, 7, and 8 fold in state 3 for mouse, rat, and guinea pig mitochondria, respectively. The maximal H(2)O(2) emission as a percentage of the total O(2) consumption flux was 11%/2.3% for mouse in states 4 and 3 followed by 2%/0.25% and 0.74%/0.29% in the rat and guinea pig, respectively. A minimal computational model accounting for the kinetics of GSH/Trx systems was developed and was able to simulate increase in H(2)O(2) emission fluxes when both scavenging systems were inhibited separately or together. Model simulations suggest that GSH/Trx systems act in concert. When the scavenging capacity of either one of them saturates during H(2)O(2) overload, they relieve each other until complete saturation, when maximal ROS emission occurs. Quantitatively, these results converge on the idea that GSH/Trx scavenging systems in mitochondria are both essential for keeping minimal levels of H(2)O(2) emission, especially during state 3 respiration, when the energetic output is maximal. This suggests that the very low levels of H(2)O(2) emission observed during forward electron transport in the respiratory chain are a result of the well-orchestrated actions of the two antioxidant systems working continuously to offset ROS production.


Assuntos
Antioxidantes/metabolismo , Glutationa/metabolismo , Peróxido de Hidrogênio/metabolismo , Mitocôndrias Cardíacas/metabolismo , Tiorredoxinas/metabolismo , Animais , Respiração Celular/fisiologia , Transporte de Elétrons/fisiologia , Cobaias , Cinética , Camundongos , Modelos Teóricos , Ratos , Espécies Reativas de Oxigênio/metabolismo
12.
J Am Coll Cardiol ; 59(22): 1979-87, 2012 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-22624839

RESUMO

OBJECTIVES: The aim of this study was to determine whether activation of ß3-adrenergic receptor (AR) and downstream signaling of nitric oxide synthase (NOS) isoforms protects the heart from failure and hypertrophy induced by pressure overload. BACKGROUND: ß3-AR and its downstream signaling pathways are recognized as novel modulators of heart function. Unlike ß1- and ß2-ARs, ß3-ARs are stimulated at high catecholamine concentrations and induce negative inotropic effects, serving as a "brake" to protect the heart from catecholamine overstimulation. METHODS: C57BL/6J and neuronal NOS (nNOS) knockout mice were assigned to receive transverse aortic constriction (TAC), BRL37344 (ß3 agonist, BRL 0.1 mg/kg/h), or both. RESULTS: Three weeks of BRL treatment in wild-type mice attenuated left ventricular dilation and systolic dysfunction, and partially reduced cardiac hypertrophy induced by TAC. This effect was associated with increased nitric oxide production and superoxide suppression. TAC decreased endothelial NOS (eNOS) dimerization, indicating eNOS uncoupling, which was not reversed by BRL treatment. However, nNOS protein expression was up-regulated 2-fold by BRL, and the suppressive effect of BRL on superoxide generation was abrogated by acute nNOS inhibition. Furthermore, BRL cardioprotective effects were actually detrimental in nNOS(-/-) mice. CONCLUSIONS: These results are the first to show in vivo cardioprotective effects of ß3-AR-specific agonism in pressure overload hypertrophy and heart failure, and support nNOS as the primary downstream NOS isoform in maintaining NO and reactive oxygen species balance in the failing heart.


Assuntos
Agonistas de Receptores Adrenérgicos beta 3/farmacologia , Insuficiência Cardíaca/prevenção & controle , Hipertrofia Ventricular Esquerda/prevenção & controle , Contração Miocárdica/efeitos dos fármacos , Miocárdio/enzimologia , Óxido Nítrico Sintase Tipo I/biossíntese , Remodelação Ventricular/fisiologia , Animais , Western Blotting , Catecolaminas/sangue , Modelos Animais de Doenças , Seguimentos , Insuficiência Cardíaca/sangue , Insuficiência Cardíaca/fisiopatologia , Hipertrofia Ventricular Esquerda/sangue , Hipertrofia Ventricular Esquerda/fisiopatologia , Espectroscopia de Ressonância Magnética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miocárdio/patologia , Estresse Oxidativo/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Superóxidos/metabolismo , Remodelação Ventricular/efeitos dos fármacos
13.
Antioxid Redox Signal ; 14(9): 1687-98, 2011 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-21235349

RESUMO

The nitric oxide (NO(•)) sibling, nitroxyl or nitrosyl hydride (HNO), is emerging as a molecule whose pharmacological properties include providing functional support to failing hearts. HNO also preconditions myocardial tissue, protecting it against ischemia-reperfusion injury while exerting vascular antiproliferative actions. In this review, HNO's peculiar cardiovascular assets are discussed in light of its unique chemistry that distinguish HNO from NO(•) as well as from reactive oxygen and nitrogen species such as the hydroxyl radical and peroxynitrite. Included here is a discussion of the possible routes of HNO formation in the myocardium and its chemical targets in the heart. HNO has been shown to have positive inotropic/lusitropic effects under normal and congestive heart failure conditions in animal models. The mechanistic intricacies of the beneficial cardiac effects of HNO are examined in cellular models. In contrast to ß-receptor/cyclic adenosine monophosphate/protein kinase A-dependent enhancers of myocardial performance, HNO uses its "thiophylic" nature as a vehicle to interact with redox switches such as cysteines, which are located in key components of the cardiac electromechanical machinery ruling myocardial function. Here, we will briefly review new features of HNO's cardiovascular effects that when combined with its positive inotropic/lusitropic action may render HNO donors an attractive addition to the current therapeutic armamentarium for treating patients with acutely decompensated congestive heart failure.


Assuntos
Óxidos de Nitrogênio/metabolismo , Animais , Humanos , Modelos Biológicos , Miocárdio/metabolismo , Miocárdio/patologia , Óxido Nítrico Sintase/metabolismo , Oxirredução , Espécies Reativas de Oxigênio/metabolismo , Traumatismo por Reperfusão/metabolismo , Compostos de Sulfidrila/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA