Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Cell Res ; 33(7): 546-561, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37156877

RESUMO

Genetic information is generally transferred from RNA to protein according to the classic "Central Dogma". Here, we made a striking discovery that post-translational modification of a protein specifically regulates the editing of its own mRNA. We show that S-nitrosylation of cathepsin B (CTSB) exclusively alters the adenosine-to-inosine (A-to-I) editing of its own mRNA. Mechanistically, CTSB S-nitrosylation promotes the dephosphorylation and nuclear translocation of ADD1, leading to the recruitment of MATR3 and ADAR1 to CTSB mRNA. ADAR1-mediated A-to-I RNA editing enables the binding of HuR to CTSB mRNA, resulting in increased CTSB mRNA stability and subsequently higher steady-state levels of CTSB protein. Together, we uncovered a unique feedforward mechanism of protein expression regulation mediated by the ADD1/MATR3/ADAR1 regulatory axis. Our study demonstrates a novel reverse flow of information from the post-translational modification of a protein back to the post-transcriptional regulation of its own mRNA precursor. We coined this process as "Protein-directed EDiting of its Own mRNA by ADAR1 (PEDORA)" and suggest that this constitutes an additional layer of protein expression control. "PEDORA" could represent a currently hidden mechanism in eukaryotic gene expression regulation.


Assuntos
Catepsina B , Edição de RNA , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Catepsina B/genética , Catepsina B/metabolismo , Regulação da Expressão Gênica , Precursores de RNA/metabolismo , RNA/metabolismo , Adenosina Desaminase/genética , Adenosina Desaminase/metabolismo
2.
Redox Biol ; 52: 102290, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35334246

RESUMO

Endothelial dysfunction is the initial process of atherosclerosis. Heat shock protein 90 (Hsp90), as a molecular chaperone, plays a crucial role in various cardiovascular diseases. Hsp90 function is regulated by S-nitrosylation (SNO). However, the precise role of SNO-Hsp90 in endothelial dysfunction during atherosclerosis remains unclear. We here identified Hsp90 as a highly S-nitrosylated target in endothelial cells (ECs) by biotin switch assay combined with liquid chromatography-tandem mass spectrometry (LC-MS/MS). The elevation of SNO-Hsp90 was observed in atherosclerotic human and rodent aortas as well as in oxidized LDL (oxLDL)-treated ECs. Inhibition of inducible nitric oxide synthase (iNOS) or transfection with Hsp90 cysteine 521 (Cys521) mutation plasmid decreased the level of SNO-Hsp90 in oxLDL-cultured ECs. Coimmunoprecipitation and proximity ligation assay demonstrated that SNO-Hsp90 at Cys521 suppressed the interaction between Hsp90 and activator of Hsp90 ATPase activity 1 (AHA1), but promoted the association of Hsp90 and cell division cycle 37 (CDC37). Hsp90 Cys521 mutation increased endothelial nitric oxide synthase (eNOS) activity and inhibited nuclear factor kappa-B (NF-κB) signaling, thereby increasing nitric oxide (NO) bioavailability and alleviating endothelial adhesion, inflammation and oxidative stress in oxLDL-treated ECs. Also, administration of endothelial-specific adeno-associated viruses of Cys521-mutated Hsp90 significantly mitigated vascular oxidative stress, macrophage infiltration and atherosclerosis lesion areas in high fat diet-fed ApoE-/- mice. In conclusion, SNO-Hsp90 at Cys521, that serves as a conformational switch, disrupts Hsp90/AHA1 interaction but promotes recruitment of CDC37 to exacerbate atherosclerosis.


Assuntos
Aterosclerose , Cisteína , Adenosina Trifosfatases , Animais , Aterosclerose/genética , Aterosclerose/metabolismo , Cromatografia Líquida , Cisteína/metabolismo , Células Endoteliais/metabolismo , Proteínas de Choque Térmico HSP90/genética , Proteínas de Choque Térmico HSP90/metabolismo , Camundongos , Chaperonas Moleculares/metabolismo , Óxido Nítrico Sintase Tipo III/genética , Óxido Nítrico Sintase Tipo III/metabolismo , Espectrometria de Massas em Tandem
3.
Acta Pharmacol Sin ; 43(3): 602-612, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-34011968

RESUMO

Cardiac fibrosis (CF) is an irreversible pathological process that occurs in almost all kinds of cardiovascular diseases. Phosphorylation-dependent activation of c-Jun N-terminal kinase (JNK) induces cardiac fibrosis. However, whether S-nitrosylation of JNK mediates cardiac fibrosis remains an open question. A biotin-switch assay confirmed that S-nitrosylation of JNK (SNO-JNK) increased significantly in the heart tissues of hypertrophic patients, transverse aortic constriction (TAC) mice, spontaneously hypertensive rats (SHRs), and neonatal rat cardiac fibroblasts (NRCFs) stimulated with angiotensin II (Ang II). Site to site substitution of alanine for cysteine in JNK was applied to determine the S-nitrosylated site. S-Nitrosylation occurred at both Cys116 and Cys163 and substitution of alanine for cysteine 116 and cysteine 163 (C116/163A) inhibited Ang II-induced myofibroblast transformation. We further confirmed that the source of S-nitrosylation was inducible nitric oxide synthase (iNOS). 1400 W, an inhibitor of iNOS, abrogated the profibrotic effects of Ang II in NRCFs. Mechanistically, SNO-JNK facilitated the nuclear translocation of JNK, increased the phosphorylation of c-Jun, and induced the transcriptional activity of AP-1 as determined by chromatin immunoprecipitation and EMSA. Finally, WT and iNOS-/- mice were subjected to TAC and iNOS knockout reduced SNO-JNK and alleviated cardiac fibrosis. Our findings demonstrate an alternative mechanism by which iNOS-induced SNO-JNK increases JNK pathway activity and accelerates cardiac fibrosis. Targeting SNO-JNK might be a novel therapeutic strategy against cardiac fibrosis.


Assuntos
Fibrose/patologia , Cardiopatias/patologia , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Angiotensina II/farmacologia , Animais , Aorta/efeitos dos fármacos , Fibroblastos/efeitos dos fármacos , Humanos , Iminas/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Óxido Nítrico Sintase Tipo II/efeitos dos fármacos , Ratos , Ratos Endogâmicos SHR , Transdução de Sinais/efeitos dos fármacos
4.
Front Nutr ; 8: 754235, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34708067

RESUMO

Melatonin (N-acetyl-5-methoxytryptamine) has been shown to have a cardioprotective effect against myocarditis. However, the mechanisms underlying the protective role of melatonin (MLT) in sepsis-induced myocarditis are yet to be revealed. In this study, MLT was administrated to mice, 14 days before cecal ligation puncture surgery. Echocardiography results showed that MLT alleviated cardiac dysfunction in sepsis-induced myocarditis. Furthermore, MLT reduced cardiac inflammation by inhibiting the expression of Il-1α, Il-1ß, Il-6, and Mcp-1 messenger RNA (mRNA) levels. The RNA sequencing (RNA-seq) assays with heart tissues showed that MLT maintains the mitochondrial function in sepsis-caused myocarditis. Additionally, the production of reactive oxygen species (ROS) in heart tissues was suppressed by MLT. Taken together, in evaluating the therapeutic effect of MLT on sepsis-induced myocarditis, the results showed that MLT alleviated cardiac damage by regulating mitochondrial function and mitochondrial ROS.

5.
Br J Pharmacol ; 178(23): 4608-4625, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34265086

RESUMO

BACKGROUND AND PURPOSE: Effective anti-fibrotic therapeutic solutions are unavailable so far. The heat shock protein 90 (HSP90) exerts deleterious effects in some fibrotic diseases. S-nitrosylation (SNO) of HSP90 affects its own function. However, little is known about its role in pathological stress. Here, we investigated the effect of SNO-HSP90 on cardiac fibrosis. EXPERIMENTAL APPROACH: SNO-HSP90 level was measured by biotin-switch. SNO sites were identified through mass spectrometry. S-nitrosylation site-mutated plasmids or adeno-associated virus, gene deletion, and pharmacological antagonists were used to identify the contribution of SNO-HSP90 to myocardial fibrosis. KEY RESULTS: SNO-HSP90 level was positively correlated with fibrosis marker expression in hearts from patients and significantly higher in fibrotic hearts from spontaneously hypertensive rats and mice subjected to transverse aortic constriction, as well as in angiotensin II- or isoprenaline-treated neonatal rat cardiac fibroblasts. S-nitrosylated site of HSP90 at cysteine 589 was identified. Inhibition of SNO-HSP90 by Cys589 mutation reduced fibrosis in angiotensin II- or isoprenaline-treated cardiac fibroblasts. Administration of recombinant adeno-associated virus of Cys589 mutation improved heart function and alleviated fibrosis in transverse aortic constriction mice. Mechanistically, SNO-HSP90 stimulated binding of TGFß receptor 2 to HSP90, in response to fibrotic stimuli, followed by increased phosphorylation and nuclear translocation of SMAD3. Additionally, inducible NO synthase (iNOS) deficiency or the iNOS inhibitor, 1400W, reduced SNO-HSP90 levels and activation of the TGFß/SMAD3 signalling pathway. CONCLUSIONS AND IMPLICATIONS: Genetic or pharmacological inhibition of SNO-HSP90 mitigates fibrosis through blocking the TGFß/SMAD3 signalling pathway, providing a potential therapy for cardiac remodelling.


Assuntos
Fibroblastos , Proteínas de Choque Térmico HSP90/metabolismo , Transdução de Sinais , Animais , Modelos Animais de Doenças , Fibroblastos/metabolismo , Fibrose , Proteínas de Choque Térmico HSP90/genética , Humanos , Camundongos , Ratos , Proteína Smad3/metabolismo , Fator de Crescimento Transformador beta/metabolismo
6.
Circulation ; 144(8): 638-654, 2021 08 24.
Artigo em Inglês | MEDLINE | ID: mdl-34098726

RESUMO

BACKGROUND: Cardiac hypertrophy is an important prepathology of, and will ultimately lead to, heart failure. However, the mechanisms underlying pathological cardiac hypertrophy remain largely unknown. This study aims to elucidate the effects and mechanisms of HINT1 (histidine triad nucleotide-binding protein 1) in cardiac hypertrophy and heart failure. METHODS: HINT1 was downregulated in human hypertrophic heart samples compared with nonhypertrophic samples by mass spectrometry analysis. Hint1 knockout mice were challenged with transverse aortic constriction surgery. Cardiac-specific overexpression of HINT1 mice by intravenous injection of adeno-associated virus 9 (AAV9)-encoding Hint1 under the cTnT (cardiac troponin T) promoter were subjected to transverse aortic construction. Unbiased transcriptional analyses were used to identify the downstream targets of HINT1. AAV9 bearing shRNA against Hoxa5 (homeobox A5) was administrated to investigate whether the effects of HINT1 on cardiac hypertrophy were HOXA5-dependent. RNA sequencing analysis was performed to recapitulate possible changes in transcriptome profile.Coimmunoprecipitation assays and cellular fractionation analyses were conducted to examine the mechanism by which HINT1 regulates the expression of HOXA5. RESULTS: The reduction of HINT1 expression was observed in the hearts of hypertrophic patients and pressure overloaded-induced hypertrophic mice, respectively. In Hint1-deficient mice, cardiac hypertrophy deteriorated after transverse aortic construction. Conversely, cardiac-specific overexpression of HINT1 alleviated cardiac hypertrophy and dysfunction. Unbiased profiler polymerase chain reaction array showed HOXA5 is 1 target for HINT1, and the cardioprotective role of HINT1 was abolished by HOXA5 knockdown in vivo. Hoxa5 was identified to affect hypertrophy through the TGF-ß (transforming growth factor ß) signal pathway. Mechanically, HINT1 inhibited PKCß1 (protein kinase C ß type 1) membrane translocation and phosphorylation via direct interaction, attenuating the MEK/ERK/YY1 (mitogen-activated protein kinase/extracellular signal-regulated kinase kinase/yin yang 1) signal pathway, downregulating HOXA5 expression, and eventually attenuating cardiac hypertrophy. CONCLUSIONS: HINT1 protects against cardiac hypertrophy through suppressing HOXA5 expression. These findings indicate that HINT1 may be a potential target for therapeutic interventions in cardiac hypertrophy and heart failure.


Assuntos
Cardiomegalia/etiologia , Cardiomegalia/metabolismo , Regulação da Expressão Gênica , Proteínas de Homeodomínio/genética , Proteínas do Tecido Nervoso/metabolismo , Animais , Biomarcadores , Cardiomegalia/diagnóstico , Células Cultivadas , Bases de Dados Genéticas , Modelos Animais de Doenças , Suscetibilidade a Doenças , Proteínas de Homeodomínio/metabolismo , Humanos , Imuno-Histoquímica , Sistema de Sinalização das MAP Quinases , Masculino , Camundongos , Camundongos Knockout , Modelos Biológicos , Miócitos Cardíacos/metabolismo , Proteínas do Tecido Nervoso/deficiência , Proteínas do Tecido Nervoso/genética , Especificidade de Órgãos , Ratos , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA