Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Sci Rep ; 13(1): 15515, 2023 09 19.
Artigo em Inglês | MEDLINE | ID: mdl-37726294

RESUMO

Oxidative stress has been shown to induce cell death in a wide range of human diseases including cardiac ischemia/reperfusion injury, drug induced cardiotoxicity, and heart failure. However, the mechanism of cell death induced by oxidative stress remains incompletely understood. Here we provide new evidence that oxidative stress primarily induces ferroptosis, but not apoptosis, necroptosis, or mitochondria-mediated necrosis, in cardiomyocytes. Intriguingly, oxidative stress induced by organic oxidants such as tert-butyl hydroperoxide (tBHP) and cumene hydroperoxide (CHP), but not hydrogen peroxide (H2O2), promoted glutathione depletion and glutathione peroxidase 4 (GPX4) degradation in cardiomyocytes, leading to increased lipid peroxidation. Moreover, elevated oxidative stress is also linked to labile iron overload through downregulation of the transcription suppressor BTB and CNC homology 1 (Bach1), upregulation of heme oxygenase 1 (HO-1) expression, and enhanced iron release via heme degradation. Strikingly, oxidative stress also promoted HO-1 translocation to mitochondria, leading to mitochondrial iron overload and lipid reactive oxygen species (ROS) accumulation. Targeted inhibition of mitochondrial iron overload or ROS accumulation, by overexpressing mitochondrial ferritin (FTMT) or mitochondrial catalase (mCAT), respectively, markedly inhibited oxidative stress-induced ferroptosis. The levels of mitochondrial iron and lipid peroxides were also markedly increased in cardiomyocytes subjected to simulated ischemia and reperfusion (sI/R) or the chemotherapeutic agent doxorubicin (DOX). Overexpressing FTMT or mCAT effectively prevented cardiomyocyte death induced by sI/R or DOX. Taken together, oxidative stress induced by organic oxidants but not H2O2 primarily triggers ferroptotic cell death in cardiomyocyte through GPX4 and Bach1/HO-1 dependent mechanisms. Our results also reveal mitochondrial iron overload via HO-1 mitochondrial translocation as a key mechanism as well as a potential molecular target for oxidative stress-induced ferroptosis in cardiomyocytes.


Assuntos
Sobrecarga de Ferro , Estresse Oxidativo , Humanos , Espécies Reativas de Oxigênio , Morte Celular , Ferro , Miócitos Cardíacos
2.
J Mol Cell Cardiol ; 145: 59-73, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32553594

RESUMO

Deubiquitinating enzymes (DUBs) appear to be a new class of regulators of cardiac homeostasis and disease. However, DUB-mediated signaling in the heart is not well understood. Herein we report a novel mechanism by which cylindromatosis (CYLD), a DUB mediates cardiac pathological remodeling and dysfunction. Cardiomyocyte-restricted (CR) overexpression of CYLD (CR-CYLD) did not cause gross health issues and hardly affected cardiac function up to age of one year in both female and male mice at physiological conditions. However, CR-CYLD overexpression exacerbated pressure overload (PO)-induced cardiac dysfunction associated with suppressed cardiac hypertrophy and increased myocardial apoptosis in mice independent of the gender. At the molecular level, CR-CYLD overexpression enhanced PO-induced increases in poly-ubiquitinated proteins marked by lysine (K)48-linked ubiquitin chains and autophagic vacuoles containing undegraded contents while suppressing autophagic flux. Augmentation of cardiac autophagy via CR-ATG7 overexpression protected against PO-induced cardiac pathological remodeling and dysfunction in both female and male mice. Intriguingly, CR-CYLD overexpression switched the CR-ATG7 overexpression-dependent cardiac protection into myocardial damage and dysfunction associated with increased accumulation of autophagic vacuoles containing undegraded contents in the heart. Genetic manipulation of Cyld in combination with pharmacological modulation of autophagic functional status revealed that CYLD suppressed autolysosomal degradation and promoted cell death in cardiomyocytes. In addition, Cyld gene gain- and/or loss-of-function approaches in vitro and in vivo demonstrated that CYLD mediated cardiomyocyte death associated with impaired reactivation of mechanistic target of rapamycin complex 1 (mTORC1) and upregulated Ras genes from rat brain 7 (Rab7), two key components for autolysosomal degradation. These results demonstrate that CYLD serves as a novel mediator of cardiac pathological remodeling and dysfunction by suppressing autolysosome efflux in cardiomyocytes. Mechanistically, it is most likely that CYLD suppresses autolysosome efflux via impairing mTORC1 reactivation and interrupting Rab7 release from autolysosomes in cardiomyocytes.


Assuntos
Cardiomiopatias/metabolismo , Enzima Desubiquitinante CYLD/metabolismo , Lisossomos/metabolismo , Miócitos Cardíacos/metabolismo , Pressão , Animais , Autofagia , Proteína 7 Relacionada à Autofagia/metabolismo , Encéfalo/metabolismo , Fibroblastos/metabolismo , Genes ras , Lisina/metabolismo , Lisossomos/ultraestrutura , Masculino , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Camundongos Transgênicos , Miocárdio/patologia , Miócitos Cardíacos/patologia , Ratos , Ubiquitinação , Regulação para Cima , Vacúolos/metabolismo , Vacúolos/ultraestrutura
3.
Physiol Rev ; 99(4): 1765-1817, 2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31364924

RESUMO

Twelve regulated cell death programs have been described. We review in detail the basic biology of nine including death receptor-mediated apoptosis, death receptor-mediated necrosis (necroptosis), mitochondrial-mediated apoptosis, mitochondrial-mediated necrosis, autophagy-dependent cell death, ferroptosis, pyroptosis, parthanatos, and immunogenic cell death. This is followed by a dissection of the roles of these cell death programs in the major cardiac syndromes: myocardial infarction and heart failure. The most important conclusion relevant to heart disease is that regulated forms of cardiomyocyte death play important roles in both myocardial infarction with reperfusion (ischemia/reperfusion) and heart failure. While a role for apoptosis in ischemia/reperfusion cannot be excluded, regulated forms of necrosis, through both death receptor and mitochondrial pathways, are critical. Ferroptosis and parthanatos are also likely important in ischemia/reperfusion, although it is unclear if these entities are functioning as independent death programs or as amplification mechanisms for necrotic cell death. Pyroptosis may also contribute to ischemia/reperfusion injury, but potentially through effects in non-cardiomyocytes. Cardiomyocyte loss through apoptosis and necrosis is also an important component in the pathogenesis of heart failure and is mediated by both death receptor and mitochondrial signaling. Roles for immunogenic cell death in cardiac disease remain to be defined but merit study in this era of immune checkpoint cancer therapy. Biology-based approaches to inhibit cell death in the various cardiac syndromes are also discussed.


Assuntos
Morte Celular , Citotoxicidade Imunológica , Cardiopatias/patologia , Mitocôndrias Cardíacas/patologia , Miocárdio/patologia , Animais , Apoptose , Proteínas Reguladoras de Apoptose/metabolismo , Autofagia , Proteínas Relacionadas à Autofagia/metabolismo , Cardiopatias/imunologia , Cardiopatias/metabolismo , Cardiopatias/fisiopatologia , Humanos , Mitocôndrias Cardíacas/imunologia , Mitocôndrias Cardíacas/metabolismo , Miocárdio/imunologia , Miocárdio/metabolismo , Necrose , Piroptose , Transdução de Sinais
4.
Pathol Res Pract ; 215(2): 265-271, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30466764

RESUMO

Although dose intensification strategies achieve a favorable prognosis for pediatric patients of T-lmphoblastic lymphoma/leukemia (T-LBL/ALL), numerous side effects have been followed. Molecular targeted therapies will be needed to optimize the current treatment strategy for T-LBL/ALL. The aim of this study was to analyse expression and significance of CD47, PD1 and PDL1 in. T-LBL/ALL. We performed immunohistochemistry staining and real time fluorescence quantitative PCR (qRT-PCR) on FFPE tissues. Immunohistochemistry results showed that the high expression rate of CD47 protein was 46.4% (26/56) and the positive expression rate of PDL1 protein was 37.5% (21/56). PD1 expression was observed in tumor infiltrating lymphocytes in approximately 20% of T-LBL/ALL patients, but not expressed on tumor cells of T-LBL/ALL. And the results of qRT-PCR showed that the relative expression levels of CD47, PDL1 and PD1 mRNA in 56 cases of T LBL/ALL were significantly higher than those in control group (6.915 vs 4.050, 12.255 vs 2.575, 37.990 vs 3.615), and the differences were all statistically significant (p all <0.05). Univariate analysis showed that age, CD47 protein, CD47 mRNA,PDL1 protein and PDL1 mRNA expression were closely correlated with prognosis (P all <0.05). We found that the overall one-year survival rates of patients with a high expression (≥M) of CD47 and PDL1 mRNA were higher than in patients with low expression (25 years old. Multivariate Cox regression analysis showed that the high expression of CD47 and PDL1 protein were independent prognostic factors (both p < 0.05). In a word, PD1/PDL1 and CD47 may be involved in the disease progression and prognosis of T-LBL/ALL, and detection and targeting of CD47 and PD1/PDL1 may provide a rational basis to for treatment of T-LBL/ALL.


Assuntos
Antígeno B7-H1/biossíntese , Biomarcadores Tumorais/análise , Antígeno CD47/biossíntese , Leucemia-Linfoma Linfoblástico de Células T Precursoras/patologia , Receptor de Morte Celular Programada 1/biossíntese , Adolescente , Adulto , Idoso , Criança , Pré-Escolar , Progressão da Doença , Feminino , Perfilação da Expressão Gênica , Humanos , Estimativa de Kaplan-Meier , Masculino , Pessoa de Meia-Idade , Leucemia-Linfoma Linfoblástico de Células T Precursoras/mortalidade , Prognóstico , Modelos de Riscos Proporcionais , Transcriptoma , Adulto Jovem
6.
Circulation ; 136(8): 729-742, 2017 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-28572508

RESUMO

BACKGROUND: Programmed cell death, including apoptosis, mitochondria-mediated necrosis, and necroptosis, is critically involved in ischemic cardiac injury, pathological cardiac remodeling, and heart failure progression. Whereas apoptosis and mitochondria-mediated necrosis signaling is well established, the regulatory mechanisms of necroptosis and its significance in the pathogenesis of heart failure remain elusive. METHODS: We examined the role of tumor necrosis factor receptor-associated factor 2 (Traf2) in regulating myocardial necroptosis and remodeling using genetic mouse models. We also performed molecular and cellular biology studies to elucidate the mechanisms by which Traf2 regulates necroptosis signaling. RESULTS: We identified a critical role for Traf2 in myocardial survival and homeostasis by suppressing necroptosis. Cardiac-specific deletion of Traf2 in mice triggered necroptotic cardiac cell death, pathological remodeling, and heart failure. Plasma tumor necrosis factor α level was significantly elevated in Traf2-deficient mice, and genetic ablation of TNFR1 largely abrogated pathological cardiac remodeling and dysfunction associated with Traf2 deletion. Mechanistically, Traf2 critically regulates receptor-interacting proteins 1 and 3 and mixed lineage kinase domain-like protein necroptotic signaling with the adaptor protein tumor necrosis factor receptor-associated protein with death domain as an upstream regulator and transforming growth factor ß-activated kinase 1 as a downstream effector. It is important to note that genetic deletion of RIP3 largely rescued the cardiac phenotype triggered by Traf2 deletion, validating a critical role of necroptosis in regulating pathological remodeling and heart failure propensity. CONCLUSIONS: These results identify an important Traf2-mediated, NFκB-independent, prosurvival pathway in the heart by suppressing necroptotic signaling, which may serve as a new therapeutic target for pathological remodeling and heart failure.


Assuntos
Apoptose/fisiologia , Miócitos Cardíacos/metabolismo , Fator 2 Associado a Receptor de TNF/deficiência , Remodelação Ventricular/fisiologia , Animais , Animais Recém-Nascidos , Cardiotônicos/metabolismo , Morte Celular/fisiologia , Células Cultivadas , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Miócitos Cardíacos/patologia , Necrose/patologia , Necrose/prevenção & controle , Ratos , Ratos Sprague-Dawley , Fator 2 Associado a Receptor de TNF/genética
7.
Cell Death Dis ; 7(9): e2381, 2016 09 29.
Artigo em Inglês | MEDLINE | ID: mdl-27685625

RESUMO

Necroptosis has emerged as a new form of programmed cell death implicated in a number of pathological conditions such as ischemic injury, neurodegenerative disease, and viral infection. Recent studies indicate that TGFß-activated kinase 1 (TAK1) is nodal regulator of necroptotic cell death, although the underlying molecular regulatory mechanisms are not well defined. Here we reported that TAK1 regulates necroptotic signaling as well as caspase 8-mediated apoptotic signaling through both NFκB-dependent and -independent mechanisms. Inhibition of TAK1 promoted TNFα-induced cell death through the induction of RIP1 phosphorylation/activation and necrosome formation. Further, inhibition of TAK1 triggered two caspase 8 activation pathways through the induction of RIP1-FADD-caspase 8 complex as well as FLIP cleavage/degradation. Mechanistically, our data uncovered an essential role for the adaptor protein TNF receptor-associated protein with death domain (TRADD) in caspase 8 activation and necrosome formation triggered by TAK1 inhibition. Moreover, ablation of the deubiqutinase CYLD prevented both apoptotic and necroptotic signaling induced by TAK1 inhibition. Finally, blocking the ubiquitin-proteasome pathway prevented the degradation of key pro-survival signaling proteins and necrosome formation. Thus, we identified new regulatory mechanisms underlying the critical role of TAK1 in cell survival through regulation of multiple cell death checkpoints. Targeting key components of the necroptotic pathway (e.g., TRADD and CYLD) and the ubiquitin-proteasome pathway may represent novel therapeutic strategies for pathological conditions driven by necroptosis.


Assuntos
Apoptose , Caspase 8/metabolismo , Pontos de Checagem do Ciclo Celular , MAP Quinase Quinase Quinases/metabolismo , Transdução de Sinais , Animais , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD/metabolismo , Cisteína Endopeptidases/metabolismo , Enzima Desubiquitinante CYLD , Ativação Enzimática , Proteína de Domínio de Morte Associada a Fas/metabolismo , Humanos , Camundongos , NF-kappa B/metabolismo , Necrose , Fosforilação , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteólise , Proteína de Domínio de Morte Associada a Receptor de TNF/metabolismo , Ubiquitina/metabolismo
8.
Asia Pac J Clin Oncol ; 12(3): 308-13, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27220635

RESUMO

AIMS: Esophageal squamous cell carcinoma (ESCC) is associated with a short median survival and low cure rates. The postoperative survival time of some patients with ESCC is extremely short. It is important to understand risk factors in subsets of patients associated with extremely short-term survival. The standard factors such as T and N stage, which are predictive of actuarial survival, become less important as patients live for ≤1 year. However, the prevalence of these factors in these patient populations has not been well documented. We evaluated factors predictive of ≤1 year survival in this research. METHODS: We analyzed 1596 patients underwent esophagectomy for ESCC retrospectively. The demographic and clinicopathologic characteristics were compared between patients who died within 1 year of esophagectomy and patients who survived more than 1 year after esophagectomy. RESULTS: Univariate analysis showed significant differences between the two groups regarding gender, weight loss, comorbidity, neoadjuvant treatment, completeness of resection, pathological T stage, pathological N stage, histologic grade, the number of metastatic lymph nodes, postoperative complications, postoperative pulmonary infection and postoperative hospital stay. Based on logistic regression analysis, significant factors associated with extremely short-term survival were male gender, incomplete tumor resection, higher pathological T stage, higher pathological N stage and postoperative pulmonary infection. CONCLUSION: The independent positive predictors for extremely short-term survival are male gender, incomplete tumor resection and postoperative pulmonary infection besides higher pathological T stage and higher pathological N stage.


Assuntos
Carcinoma de Células Escamosas/cirurgia , Neoplasias Esofágicas/cirurgia , Esofagectomia , Idoso , Carcinoma de Células Escamosas/mortalidade , Carcinoma de Células Escamosas/patologia , Neoplasias Esofágicas/mortalidade , Neoplasias Esofágicas/patologia , Carcinoma de Células Escamosas do Esôfago , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Estudos Retrospectivos
9.
J Mol Cell Cardiol ; 93: 32-43, 2016 04.
Artigo em Inglês | MEDLINE | ID: mdl-26902968

RESUMO

Microtubules (MTs) have many roles in ventricular myocytes, including structural stability, morphological integrity, and protein trafficking. However, despite their functional importance, dynamic MTs had never been visualized in living adult myocytes. Using adeno-associated viral vectors expressing the MT-associated protein plus end binding protein 3 (EB3) tagged with EGFP, we were able to perform live imaging and thus capture and quantify MT dynamics in ventricular myocytes in real time under physiological conditions. Super-resolution nanoscopy revealed that EB1 associated in puncta along the length of MTs in ventricular myocytes. The vast (~80%) majority of MTs grew perpendicular to T-tubules at a rate of 0.06µm∗s(-1) and growth was preferentially (82%) confined to a single sarcomere. Microtubule catastrophe rate was lower near the Z-line than M-line. Hydrogen peroxide increased the rate of catastrophe of MTs ~7-fold, suggesting that oxidative stress destabilizes these structures in ventricular myocytes. We also quantified MT dynamics after myocardial infarction (MI), a pathological condition associated with increased production of reactive oxygen species (ROS). Our data indicate that the catastrophe rate of MTs increases following MI. This contributed to decreased transient outward K(+) currents by decreasing the surface expression of Kv4.2 and Kv4.3 channels after MI. On the basis of these data, we conclude that, under physiological conditions, MT growth is directionally biased and that increased ROS production during MI disrupts MT dynamics, decreasing K(+) channel trafficking.


Assuntos
Ventrículos do Coração/metabolismo , Microtúbulos/metabolismo , Miócitos Cardíacos/metabolismo , Estresse Oxidativo , Animais , Camundongos , Microscopia de Fluorescência , Proteínas Associadas aos Microtúbulos/metabolismo , Infarto do Miocárdio/diagnóstico , Infarto do Miocárdio/genética , Infarto do Miocárdio/metabolismo , Ligação Proteica , Transporte Proteico , Tubulina (Proteína)/metabolismo
10.
Circulation ; 130(24): 2162-72, 2014 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-25278099

RESUMO

BACKGROUND: Programmed necrosis (necroptosis) plays an important role in development, tissue homeostasis, and disease pathogenesis. The molecular mechanisms that regulate necroptosis in the heart and its physiological relevance in myocardial remodeling and heart failure remain largely unknown. METHODS AND RESULTS: Here, we identified an obligate function for TAK1 (transforming growth factor ß-activated kinase 1, gene name Map3k7) in regulating necroptotic myocyte death, myocardial remodeling, and heart failure propensity. Cardiac-specific ablation of Map3k7 in mice induced spontaneous apoptosis and necroptosis that led to adverse remodeling and heart failure, and these effects were abolished by ablation of tumor necrosis factor receptor-1. Mechanistically, TAK1 functions as a molecular switch in tumor necrosis factor receptor-1 signaling by regulating the formation of 2 cell death complexes, RIP 1 (receptor-interacting protein 1)-FADD (Fas-associated protein with death domain)-caspase 8 and RIP1-RIP3, a process that is dependent on FADD and caspase 8 as scaffolding molecules. Importantly, inhibition of RIP1 or RIP3 largely blocked necroptotic cell death, adverse remodeling, and heart failure in TAK1-deficient mice. CONCLUSIONS: These results indicate that TAK1 functions as a key survival factor in the heart by directly antagonizing necroptosis, which is critical for the maintenance of myocardial homeostasis and the prevention of adverse myocardial remodeling.


Assuntos
Insuficiência Cardíaca/patologia , Insuficiência Cardíaca/fisiopatologia , MAP Quinase Quinase Quinases/fisiologia , Transdução de Sinais/fisiologia , Remodelação Ventricular/fisiologia , Animais , Apoptose/fisiologia , Caspase 8/fisiologia , Linhagem Celular , Modelos Animais de Doenças , Proteína de Domínio de Morte Associada a Fas/fisiologia , Insuficiência Cardíaca/mortalidade , Homeostase/fisiologia , MAP Quinase Quinase Quinases/deficiência , MAP Quinase Quinase Quinases/genética , Camundongos , Camundongos Knockout , Miócitos Cardíacos/patologia , Miócitos Cardíacos/fisiologia , Necrose/fisiopatologia , Proteína Serina-Treonina Quinases de Interação com Receptores/fisiologia
11.
Environ Sci Technol ; 48(20): 12199-205, 2014 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-25229460

RESUMO

The slip ammonia from selective catalytic reduction (SCR) of NOx in coal-fired flue gas can result in deterioration of the utilities or even the environmental issues. To achieve selective catalytic oxidation (SCO) of slip ammonia, Ru-modified Ce-Zr solid solution catalysts were prepared and evaluated under various conditions. It was found that the Ru/Ce(0.6)Zr(0.4)O2(polyvinylpyrrolidone (PVP)) catalyst displayed significant catalytic activity and the slip ammonia was almost completely removed with the coexistence of NOx and SO2. Interestingly, the effect of SO2 on NH3 oxidation was bifacial, and the N2 selectivity of the resulting products was as high as 100% in the presence of SO2 and NH3. The mechanism of the SCO of NH3 over Ru/Ce(0.6)Zr(0.4)O2(PVP) was studied using various techniques, and the results showed that NH3 oxidation follows an internal SCR (iSCR) mechanism. The adsorbed ammonia was first activated and reacted with lattice oxygen atoms to form an -HNO intermediate. Then, the -HNO mainly reacted with atomic oxygen from O2 to form NO. Meanwhile, the formed NO interacted with -NH2 to N2 with N2O as the byproduct, but the presence of SO2 can effectively inhibit the production of N2O.


Assuntos
Poluentes Atmosféricos/química , Amônia/química , Rubídio/química , Adsorção , Catálise , Cério/química , Óxido Nítrico/química , Oxirredução , Óxidos/química , Oxigênio/química , Povidona , Espectroscopia de Infravermelho com Transformada de Fourier , Dióxido de Enxofre/química , Zircônio/química
12.
Circ Res ; 110(8): 1077-86, 2012 Apr 13.
Artigo em Inglês | MEDLINE | ID: mdl-22403241

RESUMO

RATIONALE: Both nuclear factors of activated T cells (NFAT) and nuclear factor-κB (NFκB) are Rel homology domain (RHD)-containing transcription factors whose independent activities are critically involved in regulating cardiac hypertrophy and failure. OBJECTIVE: To determine the potential functional interaction between NFAT and NFκB signaling pathways in cardiomyocytes and its role in cardiac hypertrophy and remodeling. METHODS AND RESULTS: We identified a novel transcriptional regulatory mechanism whereby NFκB and NFAT directly interact and synergistically promote transcriptional activation in cardiomyocytes. We show that the p65 subunit of NFκB coimmunoprecipitates with NFAT in cardiomyocytes, and this interaction maps to the RHD within p65. Overexpression of the p65-RHD disrupts the association between endogenous p65 and NFATc1, leading to reduced transcriptional activity. Overexpression of IκB kinase ß (IKKß) or p65-RHD causes nuclear translocation of NFATc1, and expression of a constitutively nuclear NFATc1-SA mutant similarly facilitated p65 nuclear translocation. Combined overexpression of p65 and NFATc1 promotes synergistic activation of NFAT transcriptional activity in cardiomyocytes, whereas inhibition of NFκB with IκBαM or dominant negative IKKß reduces NFAT activity. Importantly, agonist-induced NFAT activation is reduced in p65 null mouse embryonic fibroblasts (MEFs) compared with wild-type MEFs. In vivo, cardiac-specific deletion of p65 using a Cre-loxP system causes a ≈50% reduction in NFAT activity in luciferase reporter mice. Moreover, ablation of p65 in the mouse heart decreases the hypertrophic response after pressure overload stimulation, reduces the degree of pathological remodeling, and preserves contractile function. CONCLUSIONS: Our results suggest a direct interaction between NFAT and NFκB that effectively integrates 2 disparate signaling pathways in promoting cardiac hypertrophy and ventricular remodeling.


Assuntos
Cardiomegalia/metabolismo , Miocárdio/metabolismo , Fatores de Transcrição NFATC/metabolismo , Fator de Transcrição RelA/metabolismo , Função Ventricular Esquerda , Remodelação Ventricular , Animais , Animais Recém-Nascidos , Cardiomegalia/diagnóstico por imagem , Cardiomegalia/genética , Cardiomegalia/patologia , Cardiomegalia/fisiopatologia , Cardiomegalia/prevenção & controle , Células Cultivadas , Modelos Animais de Doenças , Ecocardiografia Doppler , Regulação da Expressão Gênica , Genes Reporter , Quinase I-kappa B/metabolismo , Imunoprecipitação , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Contração Miocárdica , Miocárdio/patologia , Fatores de Transcrição NFATC/genética , Ligação Proteica , Ratos , Ratos Sprague-Dawley , Transdução de Sinais , Fator de Transcrição RelA/deficiência , Fator de Transcrição RelA/genética , Ativação Transcricional
13.
Mol Cell Biol ; 26(10): 3785-97, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16648474

RESUMO

The calcium-calmodulin-activated protein phosphatase calcineurin functions as a key mediator of diverse biologic processes, including differentiation, apoptosis, growth, and adaptive responses, in part through dephosphorylation and activation of nuclear factor of activated T-cell (NFAT) transcription factors. Apoptosis signal-regulating kinase 1 (ASK1) is an upstream component of the mitogen-activated protein kinases that serves as a pivotal regulator of cytokine-, oxidative-, and stress-induced cell death. Here, we performed a yeast two-hybrid screen with calcineurin B as bait, which identified ASK1 as a direct physical interacting partner. The C-terminal 218 amino acids of ASK1 were sufficient to mediate interaction with calcineurin B in yeast, as well as in mammalian cell lysates. Importantly, endogenous calcium binding B subunit (CnB) protein interacted with endogenous ASK1 protein in cardiomyocytes at baseline, suggesting that the interaction observed in yeast was of potential biologic relevance. Indeed, calcineurin directly dephosphorylated ASK1 at serine 967 using purified proteins or mammalian cell lysates. Dephosphorylation of ASK1 serine 967 by calcineurin promoted its disassociation from 14-3-3 proteins, resulting in ASK1 activation. Calcineurin and ASK1 cooperatively enhanced cardiomyocyte apoptosis, while expression of a dominant negative ASK1 blocked calcineurin-induced apoptosis. Mouse embryonic fibroblasts deficient in ask1 were also partially resistant to calcineurin- or ionomycin-induced apoptosis. Finally, ASK1 negatively regulated calcineurin-NFAT signaling indirectly through c-Jun NH2-terminal kinase (JNK)- and p38-mediated phosphorylation of NFAT, which blocked calcineurin- and agonist-dependent hypertrophic growth of cardiomyocytes. Thus, ASK1 and calcineurin-NFAT constitute a feedback regulatory circuit in which calcineurin positively regulates ASK1 through direct dephosphorylation, while ASK1 negatively regulates calcineurin-NFAT signaling through p38- and JNK-mediated NFAT phosphorylation.


Assuntos
Calcineurina/metabolismo , Proliferação de Células , Regulação Enzimológica da Expressão Gênica , MAP Quinase Quinase Quinase 5/metabolismo , Miócitos Cardíacos/metabolismo , Fatores de Transcrição NFATC/metabolismo , Adenoviridae/genética , Animais , Animais Recém-Nascidos , Western Blotting , Cardiomiopatia Hipertrófica/metabolismo , Cardiomiopatia Hipertrófica/patologia , Técnicas de Cultura de Células , Morte Celular , Células Cultivadas , Fragmentação do DNA , Eletroforese em Gel de Poliacrilamida , Retroalimentação Fisiológica , Glutationa Transferase/metabolismo , Imuno-Histoquímica , MAP Quinase Quinase Quinase 5/química , MAP Quinase Quinase Quinase 5/genética , Microscopia de Fluorescência , Modelos Biológicos , Miócitos Cardíacos/fisiologia , Testes de Precipitina , Estrutura Terciária de Proteína , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes de Fusão/metabolismo , Técnicas do Sistema de Duplo-Híbrido
14.
Am J Physiol Heart Circ Physiol ; 285(6): H2578-86, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12881212

RESUMO

Evidence suggests that p38 mitogen-activated protein kinase (MAPK) activation influences cardiac function on an acute basis. The characterization and mechanisms by which this occurs were investigated in the present study. Adult rat ventricular myocytes treated with 1 mM arsenite for 30 min had a 16-fold increase in p38 MAPK phosphorylation that was attenuated by SB-203580 (a p38 MAPK inhibitor). Extracellular signal-regulated protein kinase (ERK) and c-Jun NH2-terminal kinase (JNK) were also minimally activated, but this activation was not sensitive to SB-203580. In addition, arsenite caused a p38 MAPK-independent translocation/activation of protein phosphatase 2a (PP2a) and decrease in phosphorylation of myosin light chain 2 (LC2). Arsenite-p38 MAPK activation led to translocation of heat shock protein 27 but not alpha B-crystallin to the myofilaments. Using isolated cardiomyocytes, we determined that arsenite reduces isometric tension without a change in Ca2+ sensitivity of tension via p38 MAPK and lowers myofibrillar actomyosin Mg2+-ATPase activity in a p38 MAPK-independent manner. Thus arsenite induces a p38 MAPK-independent change in PP2a and LC2 that may account for the arsenite-dependent decrease in ATPase and a p38 MAPK-dependent modification of the myofilaments that decreases myocardial force development.


Assuntos
Citoesqueleto de Actina/fisiologia , Proteínas de Choque Térmico , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Contração Miocárdica/fisiologia , Miócitos Cardíacos/enzimologia , Animais , Arsenitos/farmacologia , Miosinas Cardíacas/metabolismo , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/fisiologia , Proteínas de Choque Térmico HSP27 , Ventrículos do Coração/citologia , Miócitos Cardíacos/efeitos dos fármacos , Cadeias Leves de Miosina/metabolismo , Proteínas de Neoplasias/metabolismo , Fosfoproteínas Fosfatases/metabolismo , Fosforilação , Proteína Fosfatase 2 , Ratos , Cadeia B de alfa-Cristalina/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno
15.
Am J Physiol Heart Circ Physiol ; 285(1): H97-103, 2003 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12649078

RESUMO

Adenosine A1 receptor activation causes protein phosphatase 2a (PP2a) activation in ventricular myocytes. This attenuates beta-adrenergic functional effects in the heart (Liu Q and Hofmann PA. Am J Physiol Heart Circ Physiol 283: H1314-H1321, 2002). The purpose of the present study was to identify the signaling pathway involved in the translocation/activation of PP2a by adenosine A1 receptors in ventricular myocytes. We found that N6-cyclopentyladenosine (CPA; an adenosine A1 receptor agonist)-induced PP2a translocation was blocked by p38 MAPK inhibition but not by JNK inhibition. CPA increased phosphorylation of p38 MAPK, and this effect was abolished by pertussis toxin and inhibitors of the cGMP pathway. Moreover, CPA-induced PP2a translocation was blocked by inhibition of the cGMP pathway. Guanylyl cyclase activation mimicked the effects of CPA and caused p38 MAPK phosphorylation and PP2a translocation. Finally, CPA-induced dephosphorylations of troponin I and phospholamban were blocked by pertussis toxin and attenuated by p38 MAPK inhibition. These results suggest that adenosine A1 receptor-mediated PP2a activation uses a pertussis toxin-sensitive Gi protein-guanylyl cyclase-p38 MAPK pathway. This proposed, novel pathway may play a role in acute modulation of cardiac function.


Assuntos
Proteínas Quinases Ativadas por Mitógeno/metabolismo , Células Musculares/metabolismo , Miocárdio/metabolismo , Fosfoproteínas Fosfatases/metabolismo , Receptores Purinérgicos P1/metabolismo , Animais , Bucladesina/farmacologia , Proteínas de Ligação ao Cálcio/metabolismo , Ativação Enzimática/efeitos dos fármacos , Feminino , Guanilato Ciclase/metabolismo , Técnicas In Vitro , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Miocárdio/citologia , Proteína Fosfatase 2 , Antagonistas de Receptores Purinérgicos P1 , Ratos , Ratos Wistar , Transdução de Sinais/efeitos dos fármacos , Frações Subcelulares/efeitos dos fármacos , Frações Subcelulares/metabolismo , Translocação Genética/genética , Troponina/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno
16.
Am J Physiol Heart Circ Physiol ; 283(4): H1314-21, 2002 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12234781

RESUMO

The ability of adenosine A(1) receptors to activate type 2a protein phosphatase (PP2a) and account for antiadrenergic effects was investigated in rat myocardial preparations. We observed that the adenosine A(1) receptor agonist N(6)-cyclopentyladenosine (CPA) significantly reduces the isoproterenol-induced increase in left ventricular developed pressure of isolated heats, and this effect is blocked by pretreatment of hearts with the PP2a inhibitor cantharidin. CPA alone or given in conjunction with isoproterenol stimulation decreases phosphorylation of phospholamban and troponin I in ventricular myocytes. These dephosphorylations are blocked by an adenosine A(1) receptor antagonist and by PP2a inhibition with okadaic acid. Adenosine A(1) receptor activation was also shown to increase carboxymethylation of the PP2a catalytic subunit (PP2a-C) and cause translocation of PP2a-C to the particulate fraction in ventricular myocytes. These results support the hypothesis that adenosine A(1) receptor activation leads to methylation of PP2a-C and subsequent translocation of the PP2a holoenzyme. Increases in localized PP2a activity lead to dephosphorylation of key cardiac proteins responsible for the positive inotropic effects of beta-adrenergic stimulation.


Assuntos
Adenosina/análogos & derivados , Miocárdio/enzimologia , Fosfoproteínas Fosfatases/metabolismo , Receptores Purinérgicos P1/metabolismo , Adenosina/farmacologia , Agonistas Adrenérgicos beta/farmacologia , Animais , Proteínas de Ligação ao Cálcio/metabolismo , Cantaridina/farmacologia , Inibidores Enzimáticos/farmacologia , Feminino , Técnicas In Vitro , Isoproterenol/farmacologia , Metilação , Fibras Musculares Esqueléticas/enzimologia , Miocárdio/citologia , Ácido Okadáico/farmacologia , Fosfoproteínas Fosfatases/antagonistas & inibidores , Fosforilação , Proteína Fosfatase 2 , Ratos , Ratos Wistar , Troponina I/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA