Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Basic Res Cardiol ; 118(1): 42, 2023 10 05.
Artigo em Inglês | MEDLINE | ID: mdl-37798455

RESUMO

Mitochondrial function is maintained by several strictly coordinated mechanisms, collectively termed mitochondrial quality control mechanisms, including fusion and fission, degradation, and biogenesis. As the primary source of energy in cardiomyocytes, mitochondria are the central organelle for maintaining cardiac function. Since adult cardiomyocytes in humans rarely divide, the number of dysfunctional mitochondria cannot easily be diluted through cell division. Thus, efficient degradation of dysfunctional mitochondria is crucial to maintaining cellular function. Mitophagy, a mitochondria specific form of autophagy, is a major mechanism by which damaged or unnecessary mitochondria are targeted and eliminated. Mitophagy is active in cardiomyocytes at baseline and in response to stress, and plays an essential role in maintaining the quality of mitochondria in cardiomyocytes. Mitophagy is mediated through multiple mechanisms in the heart, and each of these mechanisms can partially compensate for the loss of another mechanism. However, insufficient levels of mitophagy eventually lead to mitochondrial dysfunction and the development of heart failure. In this review, we discuss the molecular mechanisms of mitophagy in the heart and the role of mitophagy in cardiac pathophysiology, with the focus on recent findings in the field.


Assuntos
Cardiopatias , Mitofagia , Humanos , Adulto , Mitofagia/fisiologia , Autofagia/fisiologia , Mitocôndrias/metabolismo , Cardiopatias/metabolismo , Miócitos Cardíacos/metabolismo , Dinâmica Mitocondrial
2.
J Mol Cell Cardiol ; 165: 1-8, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-34919896

RESUMO

Autophagy mediates cellular quality control mechanisms and energy homeostasis through lysosomal degradation. Autophagy is typically viewed as an adaptive process that allows cells to survive against stress, such as nutrient deprivation and hypoxia. However, autophagy also mediates cell death during development and in response to stress. Cell death accompanied by autophagy activation and accumulation of autophagosomes has been classified as type II programmed cell death. Compared to the wealth of knowledge regarding the adaptive role of autophagy, however, the molecular mechanisms through which autophagy induces cell death and its functional significance are poorly understood. Autophagy is activated excessively under some conditions, causing uncontrolled degradation of cellular materials and cell death. An imbalance between autophagosome formation and lysosomal degradation causes a massive accumulation of autophagosomes, which subsequently causes cellular dysfunction and death. Dysregulation of autophagy induces a unique form of cell death, termed autosis, with defined morphological and biochemical features distinct from other forms of programmed cell death, such as apoptosis and necrosis. In the heart, dysregulated autophagy induces death of cardiomyocytes and actively mediates cardiac injury and dysfunction in some conditions, including reperfusion injury, doxorubicin cardiomyopathy, and lysosomal storage disorders. The goal in this review is to introduce the concept of autophagic cell death and discuss its functional significance in various cardiac conditions.


Assuntos
Autofagia , Miócitos Cardíacos , Apoptose , Autofagossomos/metabolismo , Autofagia/fisiologia , Lisossomos/metabolismo , Miócitos Cardíacos/metabolismo
3.
J Mol Cell Cardiol ; 173: 16-24, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36084743

RESUMO

Cardiomyocytes undergo various forms of cell death during heart disease such as myocardial infarction and heart failure. Understanding the mechanisms of cell death in cardiomyocytes is one of the most fundamental issues in the treatment of heart failure. Among the several kinds of cell death mechanisms, this review will focus on autophagy-related cardiomyocyte cell death. Although autophagy plays an essential role in mediating cellular quality control mechanisms for cell survival, dysregulation of autophagy can cause cell death, referred to as autophagy-dependent cell death or type II programmed cell death. The recent discovery of autosis as a modality of autophagy-dependent cell death with unique morphological and biochemical features has allowed us to broaden our understanding of the mechanistic role of autophagy in cell death. Here, we discuss autophagy-dependent cardiomyocyte cell death, including autosis, in pathophysiological conditions of the heart.


Assuntos
Morte Celular Autofágica , Cardiopatias , Insuficiência Cardíaca , Humanos , Autofagia/fisiologia , Miócitos Cardíacos/metabolismo , Cardiopatias/metabolismo , Insuficiência Cardíaca/metabolismo
4.
Annu Rev Physiol ; 80: 1-26, 2018 02 10.
Artigo em Inglês | MEDLINE | ID: mdl-29068766

RESUMO

Autophagy is an evolutionarily conserved mechanism by which cytoplasmic elements are degraded intracellularly. Autophagy has also emerged as a major regulator of cardiac homeostasis and function. Autophagy preserves cardiac structure and function under baseline conditions and is activated during stress, limiting damage under most conditions. It reduces injury and preserves cardiac function during ischemia. It also reduces chronic ischemic remodeling and mediates the cardiac adaptation to pressure overload by restricting misfolded protein accumulation, mitochondrial dysfunction, and oxidative stress. Impairment of autophagy is involved in the development of diabetes and aging-induced cardiac abnormalities. Autophagy defects contribute to the development of cardiac proteinopathy and doxorubicin-induced cardiomyopathy. However, massive activation of autophagy may be detrimental for the heart in certain stress conditions, such as reperfusion injury. In this review, we discuss recent evidence supporting the important role of autophagy and mitophagy in the regulation of cardiac homeostasis and adaptation to stress.


Assuntos
Autofagia/fisiologia , Lisossomos/metabolismo , Miocárdio/metabolismo , Envelhecimento/metabolismo , Animais , Homeostase/fisiologia , Humanos , Mitofagia/fisiologia , Estresse Oxidativo/fisiologia
5.
J Mol Cell Cardiol ; 142: 138-145, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32302592

RESUMO

Maintenance of mitochondrial function and integrity is critical for normal cell survival, particularly in non-dividing cells with a high-energy demand such as cardiomyocytes. Well-coordinated quality control mechanisms in cardiomyocytes, involving mitochondrial biogenesis, mitochondrial dynamics-fission and fusion, and mitophagy, act to protect against mitochondrial dysfunction. Mitochondrial fission, which requires dynamin-related protein 1 (Drp1), is essential for segregation of damaged mitochondria for degradation. Alterations in this process have been linked to cardiomyocyte apoptosis and cardiomyopathy. In this review, we discuss the role of Drp1 in mitophagy and apoptosis in the context of cardiac pathology, including myocardial ischemia and heart failure.


Assuntos
Dinaminas/genética , Mitocôndrias Cardíacas/genética , Mitocôndrias Cardíacas/metabolismo , Mitofagia , Animais , Apoptose/genética , Cardiomiopatias/etiologia , Cardiomiopatias/metabolismo , Morte Celular , Suscetibilidade a Doenças , Dinaminas/metabolismo , Regulação da Expressão Gênica , Humanos , Dinâmica Mitocondrial , Mitofagia/genética , Miócitos Cardíacos/metabolismo , Necroptose/genética , Processamento de Proteína Pós-Traducional , Transdução de Sinais
6.
J Mol Cell Cardiol ; 126: 1-12, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30408466

RESUMO

Muscle atrophy F-box (MAFbx/atrogin-1), an E3 ubiquitin ligase, is a crucial mediator of skeletal muscle atrophy and cardiac hypertrophy in response to pressure overload and exercise. The role of MAFbx in the regulation of cardiac remodeling after myocardial infarction (MI) remains unclear. Permanent coronary ligation of the left coronary artery was performed on MAFbx knockout (KO) and wild-type (WT) mice and MAFbx expression in the WT mice was shown to be significantly increased in the left ventricles after MI. The mortality rate due to post-MI cardiac rupture was significantly decreased in MAFbx KO mice compared to that in the WT mice. DNA microarray and mRNA expression analyses revealed that the upregulation of genes involved in inflammatory processes and cell motility of leukocytes and neutrophils, including Mmp9, Il1b, Cxcl2, and Nlrp3, was significantly attenuated in MAFbx KO mice 1 day after MI. MAFbx downregulation inhibited nuclear factor-κB (Nfkb) activation after MI. Flow cytometry results demonstrated that the myocardial infiltration of neutrophils was suppressed in MAFbx KO mice 1 day after MI. Nlrp3 and Il1b protein levels were decreased in MAFbx KO mice compared with those in the WT mice. MAFbx downregulation significantly attenuated Tnfa-induced Cxcl2, Il1b, and Nlrp3 expression in cardiomyocytes. We conclude that MAFbx plays an important role in the mediation of excessive inflammation, including neutrophil infiltration, inflammasome formation, and production of proinflammatory cytokines through the activation of Nfkb, promoting cardiac rupture after MI.


Assuntos
Ruptura Cardíaca Pós-Infarto/metabolismo , Proteínas Musculares/metabolismo , Proteínas Ligases SKP Culina F-Box/metabolismo , Animais , Deleção de Genes , Regulação da Expressão Gênica , Ruptura Cardíaca Pós-Infarto/genética , Ventrículos do Coração/patologia , Inflamassomos/metabolismo , Inflamação/genética , Inflamação/patologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miocárdio/metabolismo , Miocárdio/patologia , Miócitos Cardíacos/metabolismo , Infiltração de Neutrófilos , Ratos
7.
Circ Res ; 118(10): 1563-76, 2016 05 13.
Artigo em Inglês | MEDLINE | ID: mdl-27174950

RESUMO

The aging population is increasing in developed countries. Because the incidence of cardiac disease increases dramatically with age, it is important to understand the molecular mechanisms through which the heart becomes either more or less susceptible to stress. Cardiac aging is characterized by the presence of hypertrophy, fibrosis, and accumulation of misfolded proteins and dysfunctional mitochondria. Macroautophagy (hereafter referred to as autophagy) is a lysosome-dependent bulk degradation mechanism that is essential for intracellular protein and organelle quality control. Autophagy and autophagic flux are generally decreased in aging hearts, and murine autophagy loss-of-function models develop exacerbated cardiac dysfunction that is accompanied by the accumulation of misfolded proteins and dysfunctional organelles. On the contrary, stimulation of autophagy generally improves cardiac function in mouse models of protein aggregation by removing accumulated misfolded proteins, dysfunctional mitochondria, and damaged DNA, thereby improving the overall cellular environment and alleviating aging-associated pathology in the heart. Increasing lines of evidence suggest that autophagy is required for many mechanisms that mediate lifespan extension, such as caloric restriction, in various organisms. These results raise the exciting possibility that autophagy may play an important role in combating the adverse effects of aging in the heart. In this review, we discuss the role of autophagy in the heart during aging, how autophagy alleviates age-dependent changes in the heart, and how the level of autophagy in the aging heart can be restored.


Assuntos
Envelhecimento/metabolismo , Autofagia , Miocárdio/metabolismo , Animais , Humanos , Mitofagia , Estresse Oxidativo , Transdução de Sinais
8.
Am J Physiol Heart Circ Physiol ; 313(3): H584-H596, 2017 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-28646024

RESUMO

Studies using omics-based approaches have advanced our knowledge of metabolic remodeling in cardiac hypertrophy and failure. Metabolomic analysis of the failing heart has revealed global changes in mitochondrial substrate metabolism. Peroxisome proliferator-activated receptor-α (PPARα) plays a critical role in synergistic regulation of cardiac metabolism through transcriptional control. Metabolic reprogramming via PPARα signaling in heart failure ultimately propagates into myocardial energetics. However, emerging evidence suggests that the expression level of PPARα per se does not always explain the energetic state in the heart. The transcriptional activities of PPARα are dynamic, yet highly coordinated. An additional level of complexity in the PPARα regulatory mechanism arises from its ability to interact with various partners, which ultimately determines the metabolic phenotype of the diseased heart. This review summarizes our current knowledge of the PPARα regulatory mechanisms in cardiac metabolism and the possible role of PPARα in epigenetic modifications in the diseased heart. In addition, we discuss how metabolomics can contribute to a better understanding of the role of PPARα in the progression of cardiac hypertrophy and failure.


Assuntos
Cardiomegalia/metabolismo , Metabolismo Energético , Epigênese Genética , Insuficiência Cardíaca/metabolismo , Metabolômica , Miocárdio/metabolismo , PPAR gama/metabolismo , Transcrição Gênica , Animais , Cardiomegalia/genética , Cardiomegalia/patologia , Cardiomegalia/fisiopatologia , Progressão da Doença , Epigenômica , Insuficiência Cardíaca/genética , Insuficiência Cardíaca/patologia , Insuficiência Cardíaca/fisiopatologia , Humanos , Metabolômica/métodos , Miocárdio/patologia , PPAR gama/genética , Transdução de Sinais
9.
J Mol Cell Cardiol ; 78: 116-22, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25305175

RESUMO

Mitochondria are essential organelles that produce the cellular energy source, ATP. Dysfunctional mitochondria are involved in the pathophysiology of heart disease, which is associated with reduced levels of ATP and excessive production of reactive oxygen species. Mitochondria are dynamic organelles that change their morphology through fission and fusion in order to maintain their function. Fusion connects neighboring depolarized mitochondria and mixes their contents to maintain membrane potential. In contrast, fission segregates damaged mitochondria from intact ones, where the damaged part of mitochondria is subjected to mitophagy whereas the intact part to fusion. It is generally believed that mitochondrial fusion is beneficial for the heart, especially under stress conditions, because it consolidates the mitochondria's ability to supply energy. However, both excessive fusion and insufficient fission disrupt the mitochondrial quality control mechanism and potentiate cell death. In this review, we discuss the role of mitochondrial dynamics and mitophagy in the heart and the cardiomyocytes therein, with a focus on their roles in cardiovascular disease. This article is part of a Special Issue entitled "Mitochondria: From Basic Mitochondrial Biology to Cardiovascular Disease".


Assuntos
Fenômenos Fisiológicos Cardiovasculares , Mitocôndrias/fisiologia , Dinâmica Mitocondrial , Mitofagia , Animais , Autofagia , Doenças Cardiovasculares/etiologia , Humanos
10.
Circ Res ; 113(11): 1192-5, 2013 Nov 08.
Artigo em Inglês | MEDLINE | ID: mdl-24201113

RESUMO

A series of studies conducted 20 years ago, documenting the cardiac hypertrophy phenotype and its underlying signaling mechanism induced by angiotensin II (Ang II) and mechanical stress, showed a remarkable similarity between the effect of the Gαq agonist and that of mechanical forces on cardiac hypertrophy. Subsequent studies confirmed the involvement of autocrine/paracrine mechanisms, including stretch-induced release of Ang II in load-induced cardiac hypertrophy. Recent studies showed that the Ang II type 1 (AT1) receptor is also directly activated by mechanical forces, suggesting that AT1 receptors play an important role in mediating load-induced cardiac hypertrophy through both ligand- and mechanical stress-dependent mechanisms.


Assuntos
Angiotensina II/fisiologia , Cardiomegalia/fisiopatologia , Transdução de Sinais/fisiologia , Estresse Mecânico , Animais , Cardiomegalia/patologia , Humanos , Hiperplasia , Miócitos Cardíacos/patologia , Proteínas Proto-Oncogênicas c-fos/fisiologia , Receptor Tipo 1 de Angiotensina/fisiologia
11.
Circ Res ; 113(11): 1253-64, 2013 Nov 08.
Artigo em Inglês | MEDLINE | ID: mdl-24081881

RESUMO

RATIONALE: Autophagy is an essential survival mechanism during energy stress in the heart. Oxidative stress is activated by energy stress, but its role in mediating autophagy is poorly understood. NADPH oxidase (Nox) 4 is an enzyme that generates reactive oxygen species (ROS) at intracellular membranes. Whether Nox4 acts as a sensor of energy stress to mediate activation of autophagy is unknown. OBJECTIVE: We investigated whether Nox4 is involved in the regulation of autophagy and cell survival during energy stress in cardiomyocytes. METHODS AND RESULTS: Production of ROS in cardiomyocytes was increased during glucose deprivation (GD) in a Nox4-dependent manner. Protein levels and the ROS-producing activity of Nox4 were increased in the endoplasmic reticulum (ER), but not in mitochondria, in response to GD. Selective knockdown of Nox4, but not Nox2, or selective reduction of ROS in the ER with ER-targeted catalase, but not mitochondria-targeted perioxiredoxin 3, abrogated GD-induced autophagy. Nox4 promoted autophagy during GD through activation of the protein kinase RNA-activated-like ER kinase pathway by suppression of prolyl hydroxylase 4. The decrease in cell survival during GD in the presence of Nox4 knockdown was rescued by reactivation of autophagy by Atg7 overexpression, indicating that the effect of Nox4 on cell survival is critically mediated through regulation of autophagy. Nox4 was activated during fasting and prolonged ischemia in the mouse heart, where Nox4 is also required for autophagy activation and cardioprotection. CONCLUSIONS: Nox4 critically mediates autophagy in response to energy stress in cardiomyocytes by eliciting ROS in the ER and stimulating the protein kinase RNA-activated-like ER kinase signaling pathway.


Assuntos
Fator 4 Ativador da Transcrição/fisiologia , Autofagia/fisiologia , Retículo Endoplasmático/fisiologia , Fator de Iniciação 2 em Eucariotos/fisiologia , Miócitos Cardíacos/fisiologia , NADPH Oxidases/fisiologia , Estresse Fisiológico/fisiologia , eIF-2 Quinase/fisiologia , Animais , Sobrevivência Celular/fisiologia , Metabolismo Energético/fisiologia , Técnicas In Vitro , Camundongos , Camundongos Knockout , Modelos Animais , Miócitos Cardíacos/citologia , NADPH Oxidase 4 , NADPH Oxidases/deficiência , NADPH Oxidases/genética , Estresse Oxidativo/fisiologia , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/fisiologia , Regulação para Cima/fisiologia
12.
Circ Res ; 109(2): 161-71, 2011 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-21617130

RESUMO

RATIONALE: Overexpression of muscle atrophy F-box (MAFbx/atrogin-1), an E3 ubiquitin ligase, induces proteasomal degradation in cardiomyocytes. The role of endogenous MAFbx in regulating cardiac hypertrophy and failure remains unclear. OBJECTIVE: We investigated the role of MAFbx in regulating cardiac hypertrophy and function in response to pressure overload. Transverse aortic constriction (TAC) was applied to MAFbx knockout (KO) and wild-type (WT) mice. METHODS AND RESULTS: Expression of MAFbx in WT mice was significantly increased by TAC. TAC-induced increases in cardiac hypertrophy were significantly smaller in MAFbx KO than in WT mice. There was significantly less lung congestion and interstitial fibrosis in MAFbx KO than in WT mice. MAFbx KO also inhibited ß-adrenergic cardiac hypertrophy. DNA microarray analysis revealed that activation of genes associated with the transcription factor binding site for the nuclear factor-κB family were inhibited in MAFbx KO mice compared with WT mice after TAC. Although the levels of IκB-α were significantly decreased after TAC in WT mice, they were increased in MAFbx KO mice. MAFbx regulates ubiquitination and proteasomal degradation of IκB-α in cardiomyocytes. In primary cultured rat cardiomyocytes, phenylephrine-induced activation of nuclear factor-κB and hypertrophy were significantly suppressed by MAFbx knockdown but were partially rescued by overexpression of nuclear factor-κB p65. CONCLUSIONS: MAFbx plays an essential role in mediating cardiac hypertrophy in response to pressure overload. Downregulation of MAFbx inhibits cardiac hypertrophy in part through stabilization of IκB-α and inactivation of nuclear factor-κB. Taken together, inhibition of MAFbx attenuates pathological hypertrophy, thereby protecting the heart from progression into heart failure.


Assuntos
Cardiomegalia/metabolismo , Proteínas Musculares/fisiologia , NF-kappa B/metabolismo , Proteínas Ligases SKP Culina F-Box/fisiologia , Animais , Cardiomegalia/etiologia , Células Cultivadas , Constrição Patológica , Expressão Gênica , Regulação da Expressão Gênica/fisiologia , Proteínas I-kappa B/metabolismo , Camundongos , Camundongos Knockout , Proteínas Musculares/deficiência , Proteínas Musculares/metabolismo , Inibidor de NF-kappaB alfa , Substâncias Protetoras , Ratos , Proteínas Ligases SKP Culina F-Box/deficiência , Proteínas Ligases SKP Culina F-Box/metabolismo
13.
Cardiovasc Res ; 118(17): 3320-3330, 2023 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-35150237

RESUMO

The Hippo pathway, an evolutionarily conserved signalling mechanism, controls organ size and tumourigenesis. Increasing lines of evidence suggest that autophagy, an important mechanism of lysosome-mediated cellular degradation, is regulated by the Hippo pathway, which thereby profoundly affects cell growth and death responses in various cell types. In the heart, Mst1, an upstream component of the Hippo pathway, not only induces apoptosis but also inhibits autophagy through phosphorylation of Beclin 1. YAP/TAZ, transcription factor co-factors and the terminal effectors of the Hippo pathway, affect autophagy through transcriptional activation of TFEB, a master regulator of autophagy and lysosomal biogenesis. The cellular abundance of YAP is negatively regulated by autophagy and suppression of autophagy induces accumulation of YAP, which, in turn, acts as a feedback mechanism to induce autophagosome formation. Thus, the Hippo pathway and autophagy regulate each other, thereby profoundly affecting cardiomyocyte survival and death. This review discusses the interaction between the Hippo pathway and autophagy and its functional significance during stress conditions in the heart and the cardiomyocytes therein.


Assuntos
Via de Sinalização Hippo , Proteínas Serina-Treonina Quinases , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas de Ciclo Celular/metabolismo , Fatores de Transcrição/metabolismo , Miócitos Cardíacos/metabolismo , Autofagia
14.
Redox Biol ; 59: 102561, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36512915

RESUMO

Nicotinamide adenine dinucleotide (NAD+) kinase (NADK) phosphorylates NAD+, thereby producing nicotinamide adenine dinucleotide phosphate (NADP). Both NADK genes and the NADP(H)-producing mechanism are evolutionarily conserved among archaea, bacteria, plants and mammals. In mammals, NADK is activated by phosphorylation and protein-protein interaction. Recent studies conducted using genetically altered models validate the essential role of NADK in cellular redox homeostasis and metabolism in multicellular organisms. Here, we describe the evolutionary conservation, molecular properties, and signaling mechanisms and discuss the pathophysiological significance of NADK.


Assuntos
NAD , Plantas , Animais , NAD/metabolismo , NADP/metabolismo , Plantas/metabolismo , Transdução de Sinais , Mamíferos/metabolismo
15.
Circ Res ; 107(5): 642-9, 2010 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-20595653

RESUMO

RATIONALE: The function of PKN, a stress-activated protein kinase, in the heart is poorly understood. OBJECTIVE: We investigated the functional role of PKN during myocardial ischemia/reperfusion (I/R). METHODS AND RESULTS: PKN is phosphorylated at Thr774 in hearts subjected to ischemia and reperfusion. Myocardial infarction/area at risk (MI/AAR) produced by 45 minutes of ischemia and 24 hours of reperfusion was significantly smaller in transgenic mice with cardiac-specific overexpression of constitutively active (CA) PKN (Tg-CAPKN) than in nontransgenic (NTg) mice (15+/-5 versus 38+/-5%, P<0.01). The number of TUNEL-positive nuclei was significantly lower in Tg-CAPKN (0.3+/-0.2 versus 1.0+/-0.2%, P<0.05). Both MI/AAR (63+/-9 versus 45+/-8%, P<0.05) and the number of TUNEL-positive cells (7.9+/-1.0 versus 1.3+/-0.9%, P<0.05) were greater in transgenic mice with cardiac-specific overexpression of dominant negative PKN (Tg-DNPKN) than in NTg mice. Thr774 phosphorylation of PKN was also observed in response to H(2)O(2) in cultured cardiac myocytes. Stimulation of PKN prevented, whereas inhibition of PKN aggravated, cell death induced by H(2)O(2), suggesting that the cell-protective effect of PKN is cell-autonomous in cardiac myocytes. PKN induced phosphorylation of alpha B crystallin and increased cardiac proteasome activity. The infarct reducing effect in Tg-CAPKN mice was partially inhibited by epoxomicin, a proteasome inhibitor. CONCLUSIONS: PKN is activated by I/R and inhibits apoptosis of cardiac myocytes, thereby protecting the heart from I/R injury. PKN mediates phosphorylation of alpha B crystallin and stimulation of proteasome activity, which, in part, mediates the protective effect of PKN in the heart.


Assuntos
Precondicionamento Isquêmico Miocárdico , Infarto do Miocárdio/prevenção & controle , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Reperfusão Miocárdica/efeitos adversos , Miócitos Cardíacos/enzimologia , Proteína Quinase C/metabolismo , Animais , Animais Recém-Nascidos , Apoptose , Sobrevivência Celular , Células Cultivadas , Modelos Animais de Doenças , Ativação Enzimática , Peróxido de Hidrogênio/farmacologia , Camundongos , Camundongos Transgênicos , Infarto do Miocárdio/enzimologia , Infarto do Miocárdio/genética , Infarto do Miocárdio/patologia , Traumatismo por Reperfusão Miocárdica/enzimologia , Traumatismo por Reperfusão Miocárdica/genética , Traumatismo por Reperfusão Miocárdica/patologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/patologia , Oligopeptídeos/farmacologia , Fosforilação , Inibidores de Proteases/farmacologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Inibidores de Proteassoma , Proteína Quinase C/genética , Transporte Proteico , Ratos , Ratos Wistar , Transdução de Sinais , Treonina , Fatores de Tempo , Cadeia B de alfa-Cristalina/metabolismo
16.
JMA J ; 5(4): 407-415, 2022 Oct 17.
Artigo em Inglês | MEDLINE | ID: mdl-36407069

RESUMO

One of the major complications of diabetes mellitus is diabetic cardiomyopathy. One of the mechanisms that initiates the irreversible deterioration of cardiac function in diabetic cardiomyopathy is mitochondrial dysfunction. Functionally impaired mitochondria result in greater levels of oxidative stress and lipotoxicity, both of which exacerbate mitochondrial damage. Mitochondrial health is constantly monitored by mitochondrial quality control mechanisms. Mitophagy selectively degrades damaged mitochondria, thereby maintaining the healthy pool of mitochondria and preserving myocardial function. Mitophagy in diabetic cardiomyopathy is mediated by multiple mechanisms in a time-dependent manner. Potential targets for the treatment of diabetic cardiomyopathy include increased oxidative stress, mitochondrial dynamics, and mitochondrial clearance. Thus, stimulation of mitophagy represents a promising strategy for the alleviation of diabetic cardiomyopathy.

17.
Cells ; 11(2)2022 01 13.
Artigo em Inglês | MEDLINE | ID: mdl-35053374

RESUMO

Autosis is a unique form of cell death with characteristic morphological and biochemical features caused by dysregulated autophagy. Autosis is observed in the heart during the late phase of ischemia/reperfusion (I/R), when marked accumulation of autophagosomes is induced. We previously showed that the excessive accumulation of autophagosomes promotes autosis in cardiomyocytes. Although the inhibition of autophagic flux via the upregulation of Rubicon induces the accumulation of autophagosomes during I/R, it appears that additional mechanisms exacerbating autophagosome accumulation are required for the induction of autosis. Here, we show that Tfeb contributes to the induction of autosis during the late phase of I/R in the heart. During myocardial reperfusion, Tfeb is activated and translocated into the nucleus, which in turn upregulates genes involved in autophagy and lysosomal function. The overexpression of Tfeb enhanced cardiomyocyte death induced by a high dose of TAT-Beclin 1, an effect that was inhibited by the downregulation of Atg7. Conversely, the knockdown of Tfeb attenuated high-dose TAT-Beclin1-induced death in cardiomyocytes. Although the downregulation of Tfeb in the heart significantly decreased the number of autophagic vacuoles and inhibited autosis during I/R, the activation of Tfeb activity via 3,4-dimethoxychalcone, an activator of Tfeb, aggravated myocardial injury during I/R. These findings suggest that Tfeb promotes cardiomyocyte autosis during the late phase of reperfusion in the heart.


Assuntos
Autofagia/genética , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Regulação da Expressão Gênica , Traumatismo por Reperfusão Miocárdica/genética , Animais , Animais Recém-Nascidos , Proteína Beclina-1/metabolismo , Chalconas , Regulação para Baixo/genética , Produtos do Gene tat/metabolismo , Lisossomos/metabolismo , Camundongos Endogâmicos C57BL , Traumatismo por Reperfusão Miocárdica/patologia , Miócitos Cardíacos/metabolismo , Transcrição Gênica , Regulação para Cima/genética , Vacúolos/metabolismo
18.
J Mol Cell Cardiol ; 51(4): 570-3, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20955713

RESUMO

Thioredoxin 1 (Trx1) is a small molecule with reactive cysteines that reduces proteins with disulfide bonds through a thiol disulfide exchange reaction. Accumulating lines of evidence suggest that Trx1 protects the heart from ischemia/reperfusion injury, pathological hypertrophy, and inflammation; induces preconditioning effects and angiogenesis; and upregulates mitochondrial genes. Exogenously given recombinant Trx1 (r-Trx1) may protect the heart through its actions in both extracellular and intracellular spaces. In this brief review, the potential of Trx1 therapy for heart disease is discussed. This article is part of a special issue entitled "Key Signaling Molecules in Hypertrophy and Heart Failure."


Assuntos
Cardiopatias/tratamento farmacológico , Tiorredoxinas/uso terapêutico , Animais , Cardiomegalia/prevenção & controle , Coração/efeitos dos fármacos , Coração/fisiopatologia , Humanos , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Proteínas Recombinantes/farmacologia , Proteínas Recombinantes/uso terapêutico , Transdução de Sinais , Tiorredoxinas/farmacologia , Tiorredoxinas/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA