Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Biol Chem ; 297(1): 100850, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34087234

RESUMO

Reperfusion therapy, the standard treatment for acute myocardial infarction, can trigger necrotic death of cardiomyocytes and provoke ischemia/reperfusion (I/R) injury. However, signaling pathways that regulate cardiomyocyte necrosis remain largely unknown. Our recent genome-wide RNAi screen has identified a potential necrosis suppressor gene PRKAR1A, which encodes PKA regulatory subunit 1α (R1α). R1α is primarily known for regulating PKA activity by sequestering PKA catalytic subunits in the absence of cAMP. Here, we showed that depletion of R1α augmented cardiomyocyte necrosis in vitro and in vivo, resulting in exaggerated myocardial I/R injury and contractile dysfunction. Mechanistically, R1α loss downregulated the Nrf2 antioxidant transcription factor and aggravated oxidative stress following I/R. Degradation of the endogenous Nrf2 inhibitor Keap1 through p62-dependent selective autophagy was blocked by R1α depletion. Phosphorylation of p62 at Ser349 by mammalian target of rapamycin complex 1 (mTORC1), a critical step in p62-Keap1 interaction, was induced by I/R, but diminished by R1α loss. Activation of PKA by forskolin or isoproterenol almost completely abolished hydrogen-peroxide-induced p62 phosphorylation. In conclusion, R1α loss induces unrestrained PKA activation and impairs the mTORC1-p62-Keap1-Nrf2 antioxidant defense system, leading to aggravated oxidative stress, necrosis, and myocardial I/R injury. Our findings uncover a novel role of PKA in oxidative stress and necrosis, which may be exploited to develop new cardioprotective therapies.


Assuntos
Complexo de Carney/genética , Subunidade RIalfa da Proteína Quinase Dependente de AMP Cíclico/genética , Proteína 1 Associada a ECH Semelhante a Kelch/genética , Traumatismo por Reperfusão Miocárdica/genética , Fator 2 Relacionado a NF-E2/genética , Adenilil Ciclases/genética , Animais , Complexo de Carney/patologia , Complexo de Carney/terapia , Catecolaminas/metabolismo , Modelos Animais de Doenças , Humanos , Camundongos , Necrose Dirigida por Permeabilidade Transmembrânica da Mitocôndria/genética , Traumatismo por Reperfusão Miocárdica/patologia , Traumatismo por Reperfusão Miocárdica/terapia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Estresse Oxidativo/genética , Fosforilação/genética , Proteínas de Ligação a RNA/genética , Ratos , Receptores Adrenérgicos/genética , Transdução de Sinais/efeitos dos fármacos
2.
J Biol Chem ; 295(13): 4265-4276, 2020 03 27.
Artigo em Inglês | MEDLINE | ID: mdl-32075913

RESUMO

Recent clinical investigations indicate that anthracycline-based chemotherapies induce early decline in heart mass in cancer patients. Heart mass decline may be caused by a decrease in cardiac cell number because of increased cell death or by a reduction in cell size because of atrophy. We previously reported that an anthracycline, doxorubicin (DOX), induces apoptotic death of cardiomyocytes by activating cyclin-dependent kinase 2 (CDK2). However, the signaling pathway downstream of CDK2 remains to be characterized, and it is also unclear whether the same pathway mediates cardiac atrophy. Here we demonstrate that DOX exposure induces CDK2-dependent phosphorylation of the transcription factor forkhead box O1 (FOXO1) at Ser-249, leading to transcription of its proapoptotic target gene, Bcl-2-interacting mediator of cell death (Bim). In cultured cardiomyocytes, treatment with the FOXO1 inhibitor AS1842856 or transfection with FOXO1-specific siRNAs protected against DOX-induced apoptosis and mitochondrial damage. Oral administration of AS1842856 in mice abrogated apoptosis and prevented DOX-induced cardiac dysfunction. Intriguingly, pharmacological FOXO1 inhibition also attenuated DOX-induced cardiac atrophy, likely because of repression of muscle RING finger 1 (MuRF1), a proatrophic FOXO1 target gene. In conclusion, DOX exposure induces CDK2-dependent FOXO1 activation, resulting in cardiomyocyte apoptosis and atrophy. Our results identify FOXO1 as a promising drug target for managing DOX-induced cardiotoxicity. We propose that FOXO1 inhibitors may have potential as cardioprotective therapeutic agents during cancer chemotherapy.


Assuntos
Atrofia/genética , Cardiotoxicidade/genética , Quinase 2 Dependente de Ciclina/genética , Proteína Forkhead Box O1/genética , Proteínas Musculares/genética , Proteínas com Motivo Tripartido/genética , Ubiquitina-Proteína Ligases/genética , Animais , Apoptose/efeitos dos fármacos , Atrofia/induzido quimicamente , Atrofia/patologia , Cardiotoxicidade/etiologia , Cardiotoxicidade/patologia , Modelos Animais de Doenças , Doxorrubicina/efeitos adversos , Proteína Forkhead Box O1/antagonistas & inibidores , Regulação da Expressão Gênica/efeitos dos fármacos , Coração/efeitos dos fármacos , Coração/fisiopatologia , Humanos , Camundongos , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/patologia , Neoplasias/complicações , Neoplasias/tratamento farmacológico , Neoplasias/genética , Quinolonas/farmacologia , Transdução de Sinais/efeitos dos fármacos
3.
FASEB J ; 34(2): 3347-3358, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31919902

RESUMO

Radiation therapy (RT) is commonly used to treat solid tumors of the breast, lung, and esophagus; however, the heart is an unintentional target of ionizing radiation (IR). IR exposure to the heart results in chronic toxicities including heart failure. We hypothesize that the circadian system plays regulatory roles in minimizing the IR-induced cardiotoxicity. We treated mice in control (Day Shift), environmentally disrupted (Rotating Shift), and genetically disrupted (Per 1/2 mutant) circadian conditions with 18 Gy of IR to the heart. Compared to control mice, circadian clock disruption significantly exacerbated post-IR systolic dysfunction (by ultrasound echocardiography) and increased fibrosis in mice. At the cellular level, Bmal1 protein bound to Atm, Brca1, and Brca2 promoter regions and its expression level was inversely correlated with the DNA damage levels based on the state of the clock. Further studies with circadian synchronized cardiomyocytes revealed that Bmal1 depletion increased the IR-induced DNA damage and apoptosis. Collectively, these findings suggest that the circadian clock protects from IR-induced toxicity and potentially impacts RT treatment outcome in cancer patients through IR-induced DNA damage responses.


Assuntos
Miócitos Cardíacos/metabolismo , Proteínas Circadianas Period/genética , Lesões Experimentais por Radiação/genética , Animais , Apoptose , Proteínas Mutadas de Ataxia Telangiectasia/genética , Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Proteína BRCA1/genética , Proteína BRCA1/metabolismo , Proteína BRCA2/genética , Proteína BRCA2/metabolismo , Linhagem Celular , Dano ao DNA , Camundongos , Camundongos Endogâmicos C57BL , Mutação , Miócitos Cardíacos/fisiologia , Miócitos Cardíacos/efeitos da radiação , Regiões Promotoras Genéticas , Lesões Experimentais por Radiação/metabolismo , Radiação Ionizante , Ratos , Sístole
4.
J Biol Chem ; 293(51): 19672-19685, 2018 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-30361442

RESUMO

With the rapid increase in cancer survival because of improved diagnosis and therapy in the past decades, cancer treatment-related cardiotoxicity is becoming an urgent healthcare concern. The anthracycline doxorubicin (DOX), one of the most effective chemotherapeutic agents to date, causes cardiomyopathy by inducing cardiomyocyte apoptosis. We demonstrated previously that overexpression of the cyclin-dependent kinase (CDK) inhibitor p21 promotes resistance against DOX-induced cardiomyocyte apoptosis. Here we show that DOX exposure provokes cardiac CDK2 activation and cardiomyocyte cell cycle S phase reentry, resulting in enhanced cellular sensitivity to DOX. Genetic or pharmacological inhibition of CDK2 markedly suppressed DOX-induced cardiomyocyte apoptosis. Conversely, CDK2 overexpression augmented DOX-induced apoptosis. We also found that DOX-induced CDK2 activation in the mouse heart is associated with up-regulation of the pro-apoptotic BCL2 family member BCL2-like 11 (Bim), a BH3-only protein essential for triggering Bax/Bak-dependent mitochondrial outer membrane permeabilization. Further experiments revealed that DOX induces cardiomyocyte apoptosis through CDK2-dependent expression of Bim. Inhibition of CDK2 with roscovitine robustly repressed DOX-induced mitochondrial depolarization. In a cardiotoxicity model of chronic DOX exposure (5 mg/kg weekly for 4 weeks), roscovitine administration significantly attenuated DOX-induced contractile dysfunction and ventricular remodeling. These findings identify CDK2 as a key determinant of DOX-induced cardiotoxicity. CDK2 activation is necessary for DOX-induced Bim expression and mitochondrial damage. Our results suggest that pharmacological inhibition of CDK2 may be a cardioprotective strategy for preventing anthracycline-induced heart damage.


Assuntos
Apoptose/efeitos dos fármacos , Quinase 2 Dependente de Ciclina/metabolismo , Doxorrubicina/efeitos adversos , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/patologia , Inibidores de Proteínas Quinases/farmacologia , Animais , Proteína 11 Semelhante a Bcl-2/metabolismo , Cardiotoxicidade/etiologia , Cardiotoxicidade/metabolismo , Cardiotoxicidade/patologia , Cardiotoxicidade/prevenção & controle , Linhagem Celular , Ativação Enzimática/efeitos dos fármacos , Camundongos , Mitocôndrias/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Ratos , Roscovitina/farmacologia , Fase S/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos
5.
Yale J Biol Med ; 92(4): 641-650, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31866779

RESUMO

Cell cycle progression in dividing cells, characterized by faithful replication of the genomic materials and duplication of the original cell, is fundamental for growth and reproduction of all mammalian organisms. Functional maturation of postmitotic cells, however, requires cell cycle exit and terminal differentiation. In mature postmitotic cells, many cell cycle proteins remain to be expressed, or can be induced and reactivated in pathological conditions such as traumatic injury and degenerative diseases. Interestingly, elevated levels of cell cycle proteins in postmitotic cells often do not induce proliferation, but result in aberrant cell cycle reentry and cell death. At present, the cell cycle machinery is known predominantly for regulating cell cycle progression and cell proliferation, albeit accumulating evidence indicates that cell cycle proteins may also control cell death, especially in postmitotic tissues. Herein, we provide a brief summary of these findings and hope to highlight the connection between cell cycle reentry and postmitotic cell death. In addition, we also outline the signaling pathways that have been identified in cell cycle-related cell death. Advanced understanding of the molecular mechanisms underlying cell cycle-related death is of paramount importance because this knowledge can be applied to develop protective strategies against pathologies in postmitotic tissues. Moreover, a full-scope understanding of the cell cycle machinery will allow fine tuning to favor cell proliferation over cell death, thereby potentially promoting tissue regeneration.


Assuntos
Apoptose , Proteínas de Ciclo Celular/metabolismo , Mitose , Animais , Humanos , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Neurônios/citologia , Neurônios/metabolismo
6.
J Biol Chem ; 292(6): 2065-2079, 2017 02 10.
Artigo em Inglês | MEDLINE | ID: mdl-27994061

RESUMO

Autophagy is an evolutionarily conserved intracellular degradation/recycling system that is essential for cellular homeostasis but is dysregulated in a number of diseases, including myocardial hypertrophy. Although it is clear that limiting or accelerating autophagic flux can result in pathological cardiac remodeling, the physiological signaling pathways that fine-tune cardiac autophagy are poorly understood. Herein, we demonstrated that stimulation of cardiomyocytes with phenylephrine (PE), a well known hypertrophic agonist, suppresses autophagy and that activation of focal adhesion kinase (FAK) is necessary for PE-stimulated autophagy suppression and subsequent initiation of hypertrophic growth. Mechanistically, we showed that FAK phosphorylates Beclin1, a core autophagy protein, on Tyr-233 and that this post-translational modification limits Beclin1 association with Atg14L and reduces Beclin1-dependent autophagosome formation. Remarkably, although ectopic expression of wild-type Beclin1 promoted cardiomyocyte atrophy, expression of a Y233E phosphomimetic variant of Beclin1 failed to affect cardiomyocyte size. Moreover, genetic depletion of Beclin1 attenuated PE-mediated/FAK-dependent initiation of myocyte hypertrophy in vivo Collectively, these findings identify FAK as a novel negative regulator of Beclin1-mediated autophagy and indicate that this pathway can facilitate the promotion of compensatory hypertrophic growth. This novel mechanism to limit Beclin1 activity has important implications for treating a variety of pathologies associated with altered autophagic flux.


Assuntos
Autofagia , Proteína Beclina-1/metabolismo , Cardiomegalia/patologia , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Miócitos Cardíacos/patologia , Animais , Proteína Beclina-1/genética , Camundongos , Camundongos Endogâmicos C57BL , Fosforilação , Receptores Adrenérgicos alfa/metabolismo , Transdução de Sinais
7.
Physiol Rev ; 91(3): 1023-70, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21742795

RESUMO

One of the greatest examples of integrated signal transduction is revealed by examination of effects mediated by AKT kinase in myocardial biology. Positioned at the intersection of multiple afferent and efferent signals, AKT exemplifies a molecular sensing node that coordinates dynamic responses of the cell in literally every aspect of biological responses. The balanced and nuanced nature of homeostatic signaling is particularly essential within the myocardial context, where regulation of survival, energy production, contractility, and response to pathological stress all flow through the nexus of AKT activation or repression. Equally important, the loss of regulated AKT activity is primarily the cause or consequence of pathological conditions leading to remodeling of the heart and eventual decompensation. This review presents an overview compendium of the complex world of myocardial AKT biology gleaned from more than a decade of research. Summarization of the widespread influence that AKT exerts upon myocardial responses leaves no doubt that the participation of AKT in molecular signaling will need to be reckoned with as a seemingly omnipresent regulator of myocardial molecular biological responses.


Assuntos
Miocárdio/enzimologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Animais , Sinalização do Cálcio/fisiologia , Cardiomiopatias/fisiopatologia , Sobrevivência Celular/fisiologia , Ativação Enzimática , Humanos , MicroRNAs/metabolismo , Mitocôndrias/enzimologia , Contração Miocárdica/fisiologia , Neovascularização Fisiológica/fisiologia , Proteínas Quinases/metabolismo , Proteínas Proto-Oncogênicas c-pim-1/metabolismo , Caracteres Sexuais , Transdução de Sinais/fisiologia
8.
J Mol Cell Cardiol ; 67: 1-11, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24342076

RESUMO

Clinical application of potent anthracycline anticancer drugs, especially doxorubicin (DOX), is limited by a toxic cardiac side effect that is not fully understood and preventive strategies are yet to be established. Studies in genetically modified mice have demonstrated that focal adhesion kinase (FAK) plays a key role in regulating adaptive responses of the adult myocardium to pathological stimuli through activation of intracellular signaling cascades that facilitate cardiomyocyte growth and survival. The objective of this study was to determine if targeted myocardial FAK activation could protect the heart from DOX-induced de-compensation and to characterize the underlying mechanisms. To this end, mice with myocyte-restricted FAK knock-out (MFKO) or myocyte-specific expression of an active FAK variant (termed SuperFAK) were subjected to DOX treatment. FAK depletion enhanced susceptibility to DOX-induced myocyte apoptosis and cardiac dysfunction, while elevated FAK activity provided remarkable cardioprotection. Our mec6hanistic studies reveal a heretofore unappreciated role for the protective cyclin-dependent kinase inhibitor p21 in the repression of the pro-apoptotic BH3-only protein Bim and the maintenance of mitochondrial integrity and myocyte survival. DOX treatment induced proteasomal degradation of p21, which exacerbated mitochondrial dysfunction and cardiomyocyte apoptosis. FAK was both necessary and sufficient for maintaining p21 levels following DOX treatment and depletion of p21 compromised FAK-dependent protection from DOX. These findings identify p21 as a key determinant of DOX resistance downstream of FAK in cardiomyocytes and indicate that cardiac-restricted enhancement of the FAK/p21 signaling axis might be an effective strategy to preserve myocardial function in patients receiving anthracycline chemotherapy.


Assuntos
Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Doxorrubicina/antagonistas & inibidores , Doxorrubicina/toxicidade , Quinase 1 de Adesão Focal/metabolismo , Miócitos Cardíacos/patologia , Animais , Apoptose/efeitos dos fármacos , Proteínas Reguladoras de Apoptose/metabolismo , Proteína 11 Semelhante a Bcl-2 , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , Mitocôndrias/efeitos dos fármacos , Miocárdio/enzimologia , Miocárdio/patologia , Miócitos Cardíacos/enzimologia , Proteínas Proto-Oncogênicas/metabolismo , Transdução de Sinais/efeitos dos fármacos
9.
Proc Natl Acad Sci U S A ; 108(15): 6145-50, 2011 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-21444791

RESUMO

Nucleolar stress, characterized by loss of nucleolar integrity, has not been described in the cardiac context. In addition to ribosome biogenesis, nucleoli are critical for control of cell proliferation and stress responses. Our group previously demonstrated induction of the nucleolar protein nucleostemin (NS) in response to cardiac pathological insult. NS interacts with nucleophosmin (NPM), a marker of nucleolar stress with cytoprotective properties. The dynamic behavior of NS and NPM reveal that nucleolar disruption is an early event associated with stress response in cardiac cells. Rapid translocation of NS and NPM to the nucleoplasm and suppression of new preribosomal RNA synthesis occurs in both neonatal rat cardiomyocytes (NRCM) and cardiac progenitor cells (CPC) upon exposure to doxorubicin or actinomycin D. Silencing of NS significantly increases cell death resulting from doxorubicin treatment in CPC, whereas NPM knockdown alone induces cell death. Overexpression of either NS or NPM significantly decreases caspase 8 activity in cultured cardiomyocytes challenged with doxorubicin. The presence of altered nucleolar structures resulting from myocardial infarction in mice supports the model of nucleolar stress as a general response to pathological injury. Collectively, these findings serve as the initial description of myocardial nucleolar stress and establish the postulate that nucleoli acts as sensors of stress, regulating the cellular response to pathological insults.


Assuntos
Proteínas de Transporte/metabolismo , Nucléolo Celular/metabolismo , Miocárdio/metabolismo , Proteínas Nucleares/metabolismo , Estresse Fisiológico , Animais , Aorta/metabolismo , Aorta/patologia , Apoptose , Nucléolo Celular/patologia , Células Cultivadas , Constrição Patológica/metabolismo , Constrição Patológica/patologia , Proteínas de Ligação ao GTP , Humanos , Camundongos , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/patologia , Miocárdio/patologia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Nucleofosmina , RNA Ribossômico/biossíntese , Proteínas de Ligação a RNA , Ratos
10.
Cardiovasc Res ; 2024 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-38646672

RESUMO

AIMS: The anthracycline family of anticancer agents such as doxorubicin (DOX) can induce apoptotic death of cardiomyocytes and cause cardiotoxicity. We previously reported that DOX-induced apoptosis is accompanied by cardiomyocyte cell cycle-reentry. Cell cycle progression requires cyclin-dependent kinase 7 (CDK7)-mediated activation of downstream cell cycle CDKs. This study aims to determine whether CDK7 can be targeted for cardioprotection during anthracycline chemotherapy. METHODS AND RESULTS: DOX exposure induced CDK7 activation in mouse heart and isolated cardiomyocytes. Cardiac-specific ablation of Cdk7 attenuated DOX-induced cardiac dysfunction and fibrosis. Treatment with the covalent CDK7 inhibitor THZ1 also protected against DOX-induced cardiomyopathy and apoptosis. DOX treatment induced activation of the proapoptotic CDK2-FOXO1-Bim axis in a CDK7-dependent manner. In response to DOX, endogenous CDK7 directly bound and phosphorylated CDK2 at Thr160 in cardiomyocytes, leading to full CDK2 kinase activation. Importantly, inhibition of CDK7 further suppressed tumor growth when used in combination with DOX in an immunocompetent mouse model of breast cancer. CONCLUSIONS: Activation of CDK7 is necessary for DOX-induced cardiomyocyte apoptosis and cardiomyopathy. Our findings uncover a novel proapoptotic role for CDK7 in cardiomyocytes. Moreover, this study suggests that inhibition of CDK7 attenuates DOX-induced cardiotoxicity, but augments the anticancer efficacy of DOX. Therefore, combined administration of CDK7 inhibitor and DOX may exhibit diminished cardiotoxicity but superior anticancer activity.

11.
Environ Pollut ; 345: 123454, 2024 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-38286259

RESUMO

As typical antibiotics, tetracycline (TC) and sulfadiazine (SDZ) enter the human body through the food chain. Therefore, it is necessary to understand their individual and combined toxicity. In this study, the effects of TC, SDZ, and their mixture on cell viability, cell membrane damage, liver cell damage, and oxidative damage were evaluated in in vitro assays with human liver cells Huh-7. The results showed cytotoxicity of TC, SDZ, and their mixture, which induced oxidative stress and caused membrane and cell damage. The effect of antibiotics on Huh-7 cells increased with increasing concentration, except for lactate dehydrogenase (LDH) activity that commonly showed a threshold concentration response and cell viability, which commonly showed a biphasic trend, suggesting the possibility of hormetic responses where proper doses are included. The toxicity of TC was commonly higher than that of SDZ when applied at the same concentration. These findings shed light on the individual and joint effects of these major antibiotics on liver cells, providing a scientific basis for the evaluation of antibiotic toxicity and associated risks.


Assuntos
Antibacterianos , Sulfadiazina , Humanos , Sulfadiazina/toxicidade , Antibacterianos/toxicidade , Tetraciclina/toxicidade , Fígado , Hepatócitos
12.
Arterioscler Thromb Vasc Biol ; 32(4): 924-33, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22383703

RESUMO

OBJECTIVE: We previously reported that cardiac-restricted deletion of focal adhesion kinase (FAK) exacerbated myocyte death following ischemia/reperfusion (I/R). Here, we interrogated whether targeted elevation of myocardial FAK activity could protect the heart from I/R injury. METHODS AND RESULTS: Transgenic mice were generated with myocyte-specific expression of a FAK variant (termed SuperFAK) that conferred elevated allosteric activation. FAK activity in unstressed transgenic hearts was modestly elevated, but this had no discernable effect on anabolic heart growth or cardiac function. Importantly, SuperFAK hearts exhibited a dramatic increase in FAK activity and a reduction in myocyte apoptosis and infarct size 24 to 72 hours following I/R. Moreover, serial echocardiography revealed that the transgenic mice were protected from cardiac decompensation for up to 8 weeks following surgery. Mechanistic studies revealed that elevated FAK activity protected cardiomyocytes from I/R-induced apoptosis by enhancing nuclear factor-κB (NF-κB)-dependent survival signaling during the early period of reperfusion (30 and 60 minutes). Moreover, adenoviral-mediated expression of SuperFAK in cultured cardiomyocytes attenuated H(2)O(2) or hypoxia/reoxygenation-induced apoptosis, whereas blockade of the NF-κB pathway using a pharmacological inhibitor or small interfering RNAs completely abolished the beneficial effect of SuperFAK. CONCLUSIONS: Enhancing cardiac FAK activity attenuates I/R-induced myocyte apoptosis through activation of the prosurvival NF-κB pathway and may represent a novel therapeutic strategy for ischemic heart diseases.


Assuntos
Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Terapia Genética , Infarto do Miocárdio/prevenção & controle , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Miócitos Cardíacos/enzimologia , Animais , Apoptose , Células Cultivadas , Modelos Animais de Doenças , Ativação Enzimática , Proteína-Tirosina Quinases de Adesão Focal/genética , Peróxido de Hidrogênio/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Infarto do Miocárdio/enzimologia , Infarto do Miocárdio/genética , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Traumatismo por Reperfusão Miocárdica/enzimologia , Traumatismo por Reperfusão Miocárdica/genética , Traumatismo por Reperfusão Miocárdica/patologia , Traumatismo por Reperfusão Miocárdica/fisiopatologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/patologia , NF-kappa B/antagonistas & inibidores , NF-kappa B/genética , NF-kappa B/metabolismo , Interferência de RNA , Ratos , Fatores de Tempo , Transfecção , Função Ventricular Esquerda , Remodelação Ventricular
13.
JACC CardioOncol ; 5(3): 360-373, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37397090

RESUMO

Background: Anthracycline chemotherapies cause heart failure in a subset of cancer patients. We previously reported that the anthracycline doxorubicin (DOX) induces cardiotoxicity through the activation of cyclin-dependent kinase 2 (CDK2). Objectives: The aim of this study was to determine whether retinoblastoma-like 2 (RBL2/p130), an emerging CDK2 inhibitor, regulates anthracycline sensitivity in the heart. Methods: Rbl2-/- mice and Rbl2+/+ littermates received DOX (5 mg/kg/wk for 4 weeks intraperitoneally, 20 mg/kg cumulative). Heart function was monitored with echocardiography. The association of RBL2 genetic variants with anthracycline cardiomyopathy was evaluated in the SJLIFE (St. Jude Lifetime Cohort Study) and CPNDS (Canadian Pharmacogenomics Network for Drug Safety) studies. Results: The loss of endogenous Rbl2 increased basal CDK2 activity in the mouse heart. Mice lacking Rbl2 were more sensitive to DOX-induced cardiotoxicity, as evidenced by rapid deterioration of heart function and loss of heart mass. The disruption of Rbl2 exacerbated DOX-induced mitochondrial damage and cardiomyocyte apoptosis. Mechanistically, Rbl2 deficiency enhanced CDK2-dependent activation of forkhead box O1 (FOXO1), leading to up-regulation of the proapoptotic protein Bim. The inhibition of CDK2 desensitized Rbl2-depleted cardiomyocytes to DOX. In wild-type cardiomyocytes, DOX exposure induced Rbl2 expression in a FOXO1-dependent manner. Importantly, the rs17800727 G allele of the human RBL2 gene was associated with reduced anthracycline cardiotoxicity in childhood cancer survivors. Conclusions: Rbl2 is an endogenous CDK2 inhibitor in the heart and represses FOXO1-mediated proapoptotic gene expression. The loss of Rbl2 increases sensitivity to DOX-induced cardiotoxicity. Our findings suggest that RBL2 could be used as a biomarker to predict the risk of cardiotoxicity before the initiation of anthracycline-based chemotherapy.

14.
Arterioscler Thromb Vasc Biol ; 31(10): 2193-202, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21757658

RESUMO

OBJECTIVE: The investment of newly formed endothelial cell tubes with differentiated smooth muscle cells (SMC) is critical for appropriate vessel formation, but the underlying mechanisms remain unknown. We previously showed that depletion of focal adhesion kinase (FAK) in the nkx2.5 expression domain led to aberrant outflow tract (OFT) morphogenesis and strove herein to determine the cell types and mechanisms involved. METHODS AND RESULTS: We crossed fak(loxp) targeted mice with available Cre drivers to deplete FAK in OFT SMC (FAK(wnt) and FAK(nk)) or coronary SMC (FAK(cSMC)). In each case, depletion of FAK led to defective vasculogenesis that was incompatible with postnatal life. Immunohistochemical analysis of the mutant vascular structures revealed that FAK was not required for progenitor cell proliferation, survival, or differentiation into SMC but was necessary for subsequent SMC recruitment to developing vasculature. Using a novel FAK-null SMC culture model, we found that depletion of FAK did not influence SMC growth or survival, but blocked directional SMC motility and invasion toward the potent endothelial-derived chemokine, platelet-derived growth factor PDGFBB. FAK depletion resulted in unstable lamellipodial protrusions due to defective spatial-temporal activation of the small GTPase, Rac-1, and lack of Rac1-dependent recruitment of cortactin (an actin stabilizing protein) to the leading edge. Moreover, FAK null SMC exhibited a significant reduction in stimulated extracellular matrix degradation. CONCLUSIONS: FAK drives PDGFBB-stimulated SMC chemotaxis/invasion and is essential for SMC to appropriately populate the aorticopulmonary septum and the coronary vascular plexus.


Assuntos
Quimiotaxia , Quinase 1 de Adesão Focal/metabolismo , Músculo Liso Vascular/enzimologia , Miócitos de Músculo Liso/enzimologia , Neovascularização Fisiológica , Animais , Aorta/embriologia , Aorta/enzimologia , Apoptose , Becaplermina , Proliferação de Células , Sobrevivência Celular , Células Cultivadas , Quimiotaxia/genética , Vasos Coronários/embriologia , Vasos Coronários/enzimologia , Cortactina/metabolismo , Células Endoteliais/metabolismo , Matriz Extracelular/metabolismo , Quinase 1 de Adesão Focal/deficiência , Quinase 1 de Adesão Focal/genética , Regulação da Expressão Gênica no Desenvolvimento , Proteína Homeobox Nkx-2.5 , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Knockout , Músculo Liso Vascular/embriologia , Neovascularização Fisiológica/genética , Neuropeptídeos/metabolismo , Fator de Crescimento Derivado de Plaquetas/metabolismo , Proteínas Proto-Oncogênicas c-sis , Pseudópodes/enzimologia , Artéria Pulmonar/embriologia , Artéria Pulmonar/enzimologia , Codorniz/embriologia , Interferência de RNA , Transdução de Sinais , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transfecção , Proteína Wnt1/genética , Proteína Wnt1/metabolismo , Proteínas rac de Ligação ao GTP/metabolismo , Proteínas rac1 de Ligação ao GTP
15.
Eur Heart J ; 32(17): 2179-88, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21228009

RESUMO

AIMS: The cascade of events leading to compromised mitochondrial integrity in response to stress is mediated by various combinatorial interactions of pro- and anti-apoptotic molecules. Nur77, an immediate early gene that encodes a nuclear orphan receptor, translocates from the nucleus to mitochondria to induce cytochrome c release and apoptosis in cancer cells in response to various pro-apoptotic treatments. However, the role of Nur77 in the cardiac setting is still unclear. The objective of this study is to determine the physiological relevance and pathophysiological importance of Nur77 in cardiomyocytes. METHODS AND RESULTS: Myocardial Nur77 is upregulated following cardiomyopathic injury and, while expressed in the postnatal myocardium, declines in level within weeks after birth. Nur77 is localized predominantly in cardiomyocyte nuclei under normal conditions where it is not apoptotic, but translocates to mitochondria in response to oxidative stress both in vitro and in vivo. Mitochondrial localization of Nur77 induces cytochrome c release and typical morphological features of apoptosis, including chromatin condensation and DNA fragmentation. Knockdown of Nur77 rescued hydrogen peroxide-induced cardiomyocyte apoptosis. CONCLUSION: Translocation of Nur77 from the nucleus to the mitochondria in cardiomyocytes results in the loss of mitochondrial integrity and subsequent apoptosis in response to ischaemia/reperfusion injury. Our findings identify Nur77 as a novel mediator of cardiomyocyte apoptosis and warrants further investigation of mitochondrial Nur77 translocation as a mechanism to control cell death in the treatment of ischaemic heart diseases.


Assuntos
Apoptose/fisiologia , Mitocôndrias Cardíacas/fisiologia , Isquemia Miocárdica/patologia , Miócitos Cardíacos/patologia , Membro 1 do Grupo A da Subfamília 4 de Receptores Nucleares/fisiologia , Animais , Constrição , Feminino , Masculino , Camundongos , Traumatismo por Reperfusão Miocárdica/patologia , Ratos , Ratos Sprague-Dawley , Transfecção , Regulação para Cima
16.
J Am Heart Assoc ; 11(19): e024764, 2022 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-36129061

RESUMO

Background The postmitotic state of adult cardiomyocytes, maintained by the cell cycle repressor Rbl2 (retinoblastoma-like 2), is associated with considerable resistance to apoptosis. However, whether Rbl2 regulates cardiomyocyte apoptosis remains unknown. Methods and Results Here, we show that ablation of Rbl2 increased cardiomyocyte apoptosis following acute myocardial ischemia/reperfusion injury, leading to diminished cardiac function and exaggerated ventricular remodeling in the long term. Mechanistically, ischemia/reperfusion induced expression of the proapoptotic protein BCL2 interacting protein 3 (Bnip3), which was augmented by deletion of Rbl2. Because the Bnip3 promoter contains an adenoviral early region 2 binding factor (E2F)-binding site, we further showed that loss of Rbl2 upregulated the transcriptional activator E2F1 but downregulated the transcriptional repressor E2F4. In cultured cardiomyocytes, treatment with H2O2 markedly increased the levels of E2F1 and Bnip3, resulting in mitochondrial depolarization and apoptosis. Depletion of Rbl2 significantly augmented H2O2-induced mitochondrial damage and apoptosis in vitro. Conclusions Rbl2 deficiency enhanced E2F1-mediated Bnip3 expression, resulting in aggravated cardiomyocyte apoptosis and ischemia/reperfusion injury. Our results uncover a novel antiapoptotic role for Rbl2 in cardiomyocytes, suggesting that the cell cycle machinery may directly regulate apoptosis in postmitotic cardiomyocytes. These findings may be exploited to develop new strategies to limit ischemia/reperfusion injury in the treatment of acute myocardial infarction.


Assuntos
Traumatismo por Reperfusão Miocárdica , Neoplasias da Retina , Retinoblastoma , Apoptose/fisiologia , Humanos , Peróxido de Hidrogênio/metabolismo , Traumatismo por Reperfusão Miocárdica/genética , Traumatismo por Reperfusão Miocárdica/metabolismo , Miócitos Cardíacos/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Neoplasias da Retina/metabolismo , Retinoblastoma/metabolismo , Proteína p130 Retinoblastoma-Like/metabolismo
17.
Cardiovasc Res ; 118(2): 386-398, 2022 01 29.
Artigo em Inglês | MEDLINE | ID: mdl-33483740

RESUMO

Protein kinase A (PKA) is a central regulator of cardiac performance and morphology. Myocardial PKA activation is induced by a variety of hormones, neurotransmitters, and stress signals, most notably catecholamines secreted by the sympathetic nervous system. Catecholamines bind ß-adrenergic receptors to stimulate cAMP-dependent PKA activation in cardiomyocytes. Elevated PKA activity enhances Ca2+ cycling and increases cardiac muscle contractility. Dynamic control of PKA is essential for cardiac homeostasis, as dysregulation of PKA signalling is associated with a broad range of heart diseases. Specifically, abnormal PKA activation or inactivation contributes to the pathogenesis of myocardial ischaemia, hypertrophy, heart failure, as well as diabetic, takotsubo, or anthracycline cardiomyopathies. PKA may also determine sex-dependent differences in contractile function and heart disease predisposition. Here, we describe the recent advances regarding the roles of PKA in cardiac physiology and pathology, highlighting previous study limitations and future research directions. Moreover, we discuss the therapeutic strategies and molecular mechanisms associated with cardiac PKA biology. In summary, PKA could serve as a promising drug target for cardioprotection. Depending on disease types and mechanisms, therapeutic intervention may require either inhibition or activation of PKA. Therefore, specific PKA inhibitors or activators may represent valuable drug candidates for the treatment of heart diseases.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Cardiopatias/enzimologia , Contração Miocárdica , Miocárdio/enzimologia , Animais , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Ativação Enzimática , Cardiopatias/tratamento farmacológico , Cardiopatias/patologia , Cardiopatias/fisiopatologia , Humanos , Terapia de Alvo Molecular , Miocárdio/patologia , Fosforilação , Inibidores de Proteínas Quinases/uso terapêutico , Transdução de Sinais
18.
Front Cardiovasc Med ; 8: 817977, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-35111832

RESUMO

Anthracycline antineoplastic agents such as doxorubicin are widely used and highly effective component of adjuvant chemotherapy for breast cancer and curative regimens for lymphomas, leukemias, and sarcomas. The primary dose-limiting adverse effect of anthracyclines is cardiotoxicity that typically manifests as cardiomyopathy and can progress to the potentially fatal clinical syndrome of heart failure. Decades of pre-clinical research have explicated the complex and multifaceted mechanisms of anthracycline-induced cardiotoxicity. It is well-established that oxidative stress contributes to the pathobiology and recent work has elucidated important central roles for direct mitochondrial injury and iron overload. Here we focus instead on emerging aspects of anthracycline-induced cardiotoxicity that may have received less attention in other recent reviews: thrombosis, myocardial atrophy, and non-apoptotic programmed cell death.

19.
Ying Yong Sheng Tai Xue Bao ; 32(4): 1345-1351, 2021 Apr.
Artigo em Zh | MEDLINE | ID: mdl-33899403

RESUMO

Atmospheric haze pollution is a popular environmental issue in recent years. The aerosols reduce solar radiation reaching land surface, with consequences on the growth of crops. In order to examine the effects of low solar radiation intensity on the physiological characteristics and mineral nutrition of grain crops, the random designed field experiment of rice cultivar 'Nanjing 5055' planted under different shading degrees (CK, natural sunlight control; Y1 and Y2 were treatments with shading rates of 19% and 45%, respectively) were conducted. The response of chlorophyll content (SPAD), leaf area index (LAI), net photosynthetic rate of leaves, grain yields and secondary/micro element contents (Ca, Mg, Fe, Zn, Mn, Cu) in rice were measured during key growth stages (jointing, heading, and grain filling stages). Results showed that, shading treatments inhibited the synthesis of photosynthetic products and reduced the LAI during the whole growing period, but at the early stage it did not affect the chlorophyll content, which was significantly increased at the late growth stage. Compared with CK, the 1000-grain weight of rice was decreased by 14.4% and 18.4%, and seed setting rate was decreased by 4.3% and 12.9%, which resulted in rice yield reduction. With the increases of shading rates, rice yield was decreased by 58.5% and 66.4%, respectively. The nutrient concentrations, especially for the micro-elements, in brown rice and glume were increased. Shading had a negative effect on rice growth, which would eventually reduce the crop production. The higher contents of heavy metals such as Cu and Mn would be a pollution risk for human health. Therefore, the impacts of weakened solar radiation on quantity and quality of crops need comprehensive evaluation.


Assuntos
Oryza , Clorofila , Grão Comestível , Humanos , Fotossíntese , Folhas de Planta
20.
Physiol Rep ; 8(6): e14405, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32212257

RESUMO

Protein kinase A (PKA) activity is pivotal for proper functioning of the human heart, and its dysregulation has been implicated in a variety of cardiac pathologies. PKA regulatory subunit 1α (R1α, encoded by the PRKAR1A gene) is highly expressed in the heart, and controls PKA kinase activity by sequestering PKA catalytic subunits. Patients with PRKAR1A mutations are often diagnosed with Carney complex (CNC) in early adulthood, and may die later in life from cardiac complications such as heart failure. However, it remains unknown whether PRKAR1A deficiency interferes with normal heart development. Here, we showed that left ventricular mass was reduced in young CNC patients with PRKAR1A mutations or deletions. Cardiac-specific heterozygous ablation of PRKAR1A in mice increased cardiac PKA activity, and reduced heart weight and cardiomyocyte size without altering contractile function at 3 months of age. Silencing of PRKAR1A, or stimulation with the PKA activator forskolin completely abolished α1-adrenergic receptor-mediated cardiomyocyte hypertrophy. Mechanistically, depletion of PRKAR1A provoked PKA-dependent inactivating phosphorylation of Drp1 at S637, leading to impaired mitochondrial fission. Pharmacologic inhibition of Drp1 with Mdivi 1 diminished hypertrophic growth of cardiomyocytes. In conclusion, PRKAR1A deficiency suppresses cardiomyocyte hypertrophy and impedes heart growth, likely through inhibiting Drp1-mediated mitochondrial fission. These findings provide a potential novel mechanism for the cardiac manifestations associated with CNC.


Assuntos
Subunidade RIalfa da Proteína Quinase Dependente de AMP Cíclico/deficiência , Subunidade RIalfa da Proteína Quinase Dependente de AMP Cíclico/fisiologia , Ventrículos do Coração/patologia , Miócitos Cardíacos/patologia , Adolescente , Adulto , Animais , Subunidade RIalfa da Proteína Quinase Dependente de AMP Cíclico/genética , Feminino , Humanos , Hipertrofia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação , Tamanho do Órgão , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA