Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Acta Pharmacol Sin ; 44(3): 546-560, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-36042291

RESUMO

Sirtuin3 (SIRT3), a class III histone deacetylase, is implicated in various cardiovascular diseases as a novel therapeutic target. SIRT3 has been proven to be cardioprotective in a model of Ang II-induced cardiac hypertrophy. However, a few small-molecule compounds targeting deacetylases could activate SIRT3. In this study, we generated a novel SIRT3 activator, 3-(2-bromo-4-hydroxyphenyl)-7-hydroxy-2H-chromen-2-one (SZC-6), through structural optimization of the first SIRT3 agonist C12. We demonstrated that SZC-6 directly bound to SIRT3 with Kd value of 15 µM, and increased SIRT3 deacetylation activity with EC50 value of 23.2 ± 3.3 µM. In neonatal rat cardiomyocytes (NRCMs), pretreatment with SZC-6 (10, 20, 40 µM) dose-dependently attenuated isoproterenol (ISO)-induced hypertrophic responses. Administration of SZC-6 (20, 40 and 60 mg·kg-1·d-1, s.c.) for 2 weeks starting from one week prior ISO treatment dose-dependently reversed ISO-induced impairment of diastolic and systolic cardiac function in wild-type mice, but not in SIRT3 knockdown mice. We showed that SZC-6 (10, 20, 40 µM) dose-dependently inhibited cardiac fibroblast proliferation and differentiation into myofibroblasts, which was abolished in SIRT3-knockdown mice. We further revealed that activation of SIRT3 by SZC-6 increased ATP production and rate of mitochondrial oxygen consumption, and reduced ROS, improving mitochondrial function in ISO-treated NRCMs. We also found that SZC-6 dose-dependently enhanced LKB1 phosphorylation, thereby promoting AMPK activation to inhibit Drp1-dependent mitochondrial fragmentation. Taken together, these results demonstrate that SZC-6 is a novel SIRT3 agonist with potential value in the treatment of cardiac hypertrophy partly through activation of the LKB1-AMPK pathway.


Assuntos
Sirtuína 3 , Camundongos , Ratos , Animais , Sirtuína 3/metabolismo , Proteínas Quinases Ativadas por AMP/metabolismo , Cardiomegalia/induzido quimicamente , Miócitos Cardíacos/metabolismo , Isoproterenol
2.
J Cell Sci ; 132(10)2019 05 16.
Artigo em Inglês | MEDLINE | ID: mdl-31028181

RESUMO

The cancer clinical therapy of doxorubicin (Dox) treatment is limited by its life-threatening cardiotoxic effects. Dickkopf-1 (Dkk1), the founding and best-studied member of the Dkk family, functions as an antagonist of canonical Wnt/ß-catenin. Dkk1 is considered to play a broad role in a variety of biological processes, but its effects on Dox-induced cardiomyopathy are poorly understood. Here, we found that the level of Dkk1 was significantly increased in Dox-treated groups, and this increase exacerbated Dox-induced cardiomyocyte apoptosis and mitochondrial dysfunction. Overexpressing Dkk1 aggravated Dox-induced cardiotoxicity in H9C2 cells. Similar results were detected when adding active Dkk1 protein extracellularly. Conversely, adding specific antibody blocking extracellular Dkk1 attenuated the cardiotoxic response to Dox. Adenovirus encoding Dkk1 was transduced through intramyocardial injection and exacerbated Dox-induced cardiomyocyte apoptosis, mitochondrial damage and heart injury in vivo Furthermore, Wnt/ß-catenin signaling was inhibited during Dox-induced cardiotoxicity, and the re-activation of ß-catenin prevented the effect of overexpressed Dkk1 and Dox-induced cardiotoxicity. In conclusion, these results reveal the crucial role of the Dkk1-Wnt/ß-catenin signaling axis in the process of Dox-induced cardiotoxicity and provide novel insights into the potential mechanism of cardiomyopathy caused by clinical application of Dox.


Assuntos
Cardiotoxicidade/etiologia , Doxorrubicina/toxicidade , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Via de Sinalização Wnt/efeitos dos fármacos , Animais , Antibióticos Antineoplásicos/toxicidade , Apoptose/fisiologia , Cardiotoxicidade/genética , Cardiotoxicidade/metabolismo , Cardiotoxicidade/patologia , Linhagem Celular , Peptídeos e Proteínas de Sinalização Intercelular/genética , Masculino , Ratos , Ratos Sprague-Dawley , Regulação para Cima/efeitos dos fármacos , Proteínas Wnt/antagonistas & inibidores , Proteínas Wnt/metabolismo , beta Catenina/antagonistas & inibidores , beta Catenina/metabolismo
3.
Acta Pharmacol Sin ; 42(5): 715-725, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-32814818

RESUMO

Cardiac fibrosis is a typical pathological change in various cardiovascular diseases. Although it has been recognized as a crucial risk factor responsible for heart failure, there is still a lack of effective treatment. Recent evidence shows that microRNAs (miRNAs) play an important role in the development of cardiac fibrosis and represent novel therapeutic targets. In this study we tried to identify the cardiac fibrosis-associated miRNA and elucidate its regulatory mechanisms in mice. Cardiac fibrosis was induced by infusion of angiotensin II (Ang II, 2 mg·kg-1·d-1) for 2 weeks via osmotic pumps. We showed that Ang II infusion induced cardiac disfunction and fibrosis accompanied by markedly increased expression level of miR-99b-3p in heart tissues. Upregulation of miR-99b-3p and fibrotic responses were also observed in cultured rat cardiac fibroblasts (CFs) treated with Ang II (100 nM) in vitro. Transfection with miR-99b-3p mimic resulted in the overproduction of fibronectin, collagen I, vimentin and α-SMA, and facilitated the proliferation and migration of CFs. On the contrary, transfection with specific miR-99b-3p inhibitor attenuated Ang II-induced fibrotic responses. Similarly, intravenous injection of specific miR-99b-3p antagomir could prevent Ang II-infused mice from cardiac dysfunction and fibrosis. We identified glycogen synthase kinase-3 beta (GSK-3ß) as a direct target of miR-99b-3p. In CFs, miR-99b-3p mimic significantly reduced the expression of GSK-3ß, leading to activation of its downstream profibrotic effector Smad3, whereas miR-99b-3p inhibitor caused anti-fibrotic effects. GSK-3ß knockdown ameliorated the anti-fibrotic role of miR-99b-3p inhibitor. These results suggest that miR-99b-3p contributes to Ang II-induced cardiac fibrosis at least partially through GSK-3ß. The modulation of miR-99b-3p may provide a new approach for tackling fibrosis-related cardiomyopathy.


Assuntos
Doenças Cardiovasculares/metabolismo , Fibrose/metabolismo , Glicogênio Sintase Quinase 3 beta/metabolismo , MicroRNAs/metabolismo , Regiões 3' não Traduzidas , Angiotensina II , Animais , Antagomirs/farmacologia , Doenças Cardiovasculares/induzido quimicamente , Doenças Cardiovasculares/complicações , Doenças Cardiovasculares/patologia , Fibroblastos/efeitos dos fármacos , Fibrose/induzido quimicamente , Fibrose/complicações , Fibrose/patologia , Glicogênio Sintase Quinase 3 beta/genética , Masculino , Camundongos Endogâmicos C57BL , MicroRNAs/antagonistas & inibidores , Miocárdio/metabolismo , Miocárdio/patologia , Ratos Sprague-Dawley , Regulação para Cima/efeitos dos fármacos
4.
Acta Pharmacol Sin ; 42(9): 1422-1436, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-33247214

RESUMO

Reduction of expression and activity of sirtuin 3 (SIRT3) contributes to the pathogenesis of cardiomyopathy via inducing mitochondrial injury and energy metabolism disorder. However, development of effective ways and agents to modulate SIRT3 remains a big challenge. In this study we explored the upstream suppressor of SIRT3 in angiotensin II (Ang II)-induced cardiac hypertrophy in mice. We first found that SIRT3 deficiency exacerbated Ang II-induced cardiac hypertrophy, and resulted in the development of spontaneous heart failure. Since miRNAs play crucial roles in the pathogenesis of cardiac hypertrophy, we performed miRNA sequencing on myocardium tissues from Ang II-infused Sirt3-/- and wild type mice, and identified microRNA-214 (miR-214) was significantly up-regulated in Ang II-infused mice. Similar results were also obtained in Ang II-treated neonatal mouse cardiomyocytes (NMCMs). Using dual-luciferase reporter assay we demonstrated that SIRT3 was a direct target of miR-214. Overexpression of miR-214 in vitro and in vivo decreased the expression of SIRT3, which resulted in extensive mitochondrial damages, thereby facilitating the onset of hypertrophy. In contrast, knockdown of miR-214 counteracted Ang II-induced detrimental effects via restoring SIRT3, and ameliorated mitochondrial morphology and respiratory activity. Collectively, these results demonstrate that miR-214 participates in Ang II-induced cardiac hypertrophy by directly suppressing SIRT3, and subsequently leading to mitochondrial malfunction, suggesting the potential of miR-214 as a promising intervention target for antihypertrophic therapy.


Assuntos
Cardiomegalia/metabolismo , MicroRNAs/metabolismo , Mitocôndrias Cardíacas/metabolismo , Sirtuína 3/metabolismo , Angiotensina II/farmacologia , Animais , Cardiomegalia/induzido quimicamente , Cardiomegalia/genética , Cardiomegalia/patologia , Linhagem Celular , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , MicroRNAs/genética , MicroRNAs/fisiologia , Miocárdio/metabolismo , Miocárdio/patologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Ratos Sprague-Dawley , Sirtuína 3/genética
5.
Cell Commun Signal ; 18(1): 46, 2020 03 17.
Artigo em Inglês | MEDLINE | ID: mdl-32183816

RESUMO

BACKGROUND: Hepatocellular carcinoma (HCC) is the most common primary liver cancer and is a highly vascularized solid tumor. Angiopoietin-2 (ANGPT2) has been described as an attractive target for antiangiogenic therapy. Exosomes are small extracellular vesicles secreted by most cell types and contribute to cell-to-cell communication by delivering functional cargo to recipient cells. The expression of ANGPT2 in tumor-derived exosomes remains unknown. METHODS: We detected the ANGPT2 expression in HCC-derived exosomes by immunoblotting, enzyme-linked immunosorbent assay and immunogold labeling, then observed exosomal ANGPT2 internalization and recycling by confocal laser scanning microscopy, co-immunoprecipitation and immunoblotting. We used two HCC cell lines (Hep3B and MHCC97H) to overexpress ANGPT2 by lentivirus infection or knockdown ANGPT2 by the CRISPR/Cas system, then isolated exosomes to coculture with human umbilical vein endothelial cells (HUVECs) and observed the angiogenesis by Matrigel microtubule formation assay, transwell migration assay, wound healing assay, cell counting kit-8 assay, immunoblotting and in vivo tumorigenesis assay. RESULTS: We found that HCC-derived exosomes carried ANGPT2 and delivered it into HUVECs by exosome endocytosis, this delivery led to a notable increase in angiogenesis by a Tie2-independent pathway. Concomitantly, we observed that HCC cell-secreted exosomal ANGPT2 was recycled by recipient HUVECs and might be reused. In addition, the CRISPR-Cas systems to knock down ANGPT2 significantly inhibited the angiogenesis induced by HCC cell-secreted exosomal ANGPT2, and obviously suppressed the epithelial-mesenchymal transition activation in HCC. CONCLUSIONS: Taken together, these results reveal a novel pathway of tumor angiogenesis induced by HCC cell-secreted exosomal ANGPT2 that is different from the classic ANGPT2/Tie2 pathway. This way may be a potential therapeutic target for antiangiogenic therapy. Video Abstract.


Assuntos
Angiopoietina-2/fisiologia , Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/metabolismo , Linhagem Celular Tumoral , Transição Epitelial-Mesenquimal , Exossomos/metabolismo , Regulação Neoplásica da Expressão Gênica , Células Endoteliais da Veia Umbilical Humana , Humanos , Neovascularização Patológica
6.
Pharmacol Res ; 161: 105104, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32739429

RESUMO

BACKGROUND AND PURPOSE: Although histone lysine methylation has been extensively studied for their participation in pathological cardiac hypertrophy, the potential regulatory role of histone arginine methylation remains to be elucidated. The present study focused on H4R3 symmetric di-methylation (H4R3me2s) induced by protein arginine methyltransferase 5 (Prmt5), and explored its epigenetic regulation and underlying mechanisms in cardiomyocyte hypertrophy. METHODS AND RESULTS: 1. The expressions of Prmt5 and H4R3me2s were suppressed in cardiac hypertrophy models in vivo and in vitro; 2. Prmt5 silencing or its inhibitor EPZ, or knockdown of cooperator of Prmt5 (Copr5) to disrupt H4R3me2s, facilitated cardiomyocyte hypertrophy, whereas overexpression of wild type Prmt5 rather than the inactive mutant protected cardiomyocytes against hypertrophy; 3. ChIP-sequence analysis identified Filip1L as a target gene of Prmt5-induced H4R3me2s; 4. Knockdown or inhibition of Prmt5 impaired Filip1L transcription and subsequently prevented ß-catenin degradation, thus augmenting cardiomyocyte hypertrophy. CONCLUSIONS: The present study reveals that Prmt5-induced H4R3me2s ameliorates cardiomyocyte hypertrophy by transcriptional upregulation of Filip1L and subsequent enhancement of ß-catenin degradation. Deficiency of Prmt5 and the resulting suppression of H4R3me2s might facilitate the development of pathological cardiac hypertrophy. Prmt5 might serve as a key epigenetic regulator in pathological cardiac hypertrophy.


Assuntos
Histonas/metabolismo , Hipertrofia Ventricular Esquerda/enzimologia , Miócitos Cardíacos/enzimologia , Proteína-Arginina N-Metiltransferases/metabolismo , Função Ventricular Esquerda , Remodelação Ventricular , beta Catenina/metabolismo , Animais , Arginina , Células Cultivadas , Modelos Animais de Doenças , Epigênese Genética , Hipertrofia Ventricular Esquerda/genética , Hipertrofia Ventricular Esquerda/patologia , Hipertrofia Ventricular Esquerda/fisiopatologia , Masculino , Metilação , Camundongos Endogâmicos C57BL , Miócitos Cardíacos/patologia , Proteína-Arginina N-Metiltransferases/genética , Ratos Sprague-Dawley , Transdução de Sinais
7.
Acta Pharmacol Sin ; 41(9): 1150-1157, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32238888

RESUMO

Doxorubicin (Dox) is an effective chemotherapy drug against a wide range of cancers, including both hematological and solid tumors. However, the serious cardiotoxic effect restricted its clinical application. We previously have illuminated the protective role of canonical Wnt/ß-catenin signaling in Dox-induced cardiotoxicity. Secreted frizzled-related protein 1 (sFRP1) is one of the endogenous inhibitors of both canonical and noncanonical Wnt signaling. In this study, we investigated the relationship between sFRP1 and noncanonical Wnt/PCP-JNK (Wnt/planar cell polarity-c-Jun N-terminal kinase) pathway in Dox-induced cardiotoxicity in vitro and in vivo. We showed that treatment of H9c2 cardiac myoblasts with Dox (1 µM) time-dependently suppressed cell viability accompanied by significantly decreased sFRP1 protein level and increased Wnt/PCP-JNK signaling. Pretreatment with SP600125, the Wnt/PCP-JNK signaling inhibitor, attenuated Dox-induced apoptosis of H9c2 cells. Overexpression of sFRP1 protected H9c2 cells from Dox-induced apoptosis by inhibiting the Wnt/PCP-JNK pathway. After intraperitoneal injection of a cumulative dose of 15 mg/kg Dox, rats displayed significant cardiac dysfunction; their heart showed inhibited Wnt/ß-catenin signaling and activated Wnt/PCP-JNK signaling. These results suggest that sFRP1 may be a novel target for Dox-induced cardiotoxicity.


Assuntos
Antineoplásicos/efeitos adversos , Apoptose/efeitos dos fármacos , Cardiotoxicidade/metabolismo , Doxorrubicina/efeitos adversos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Proteínas de Membrana/metabolismo , Via de Sinalização Wnt/fisiologia , Animais , Antracenos/farmacologia , Linhagem Celular , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Masculino , Ratos Sprague-Dawley , Via de Sinalização Wnt/efeitos dos fármacos
8.
J Mol Cell Cardiol ; 133: 125-137, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31199952

RESUMO

The clinical application of doxorubicin (Dox) in cancer therapy is limited by its serious cardiotoxicity. Our previous studies and others have recognized that mitochondrial dysfunction is the common feature of Dox-induced cardiotoxicity. However, mechanisms underlying mitochondrial disorders remained largely unknown. SESN2, a highly conserved and stress-inducible protein, is involved in mitochondrial function and autophagy in cardiovascular diseases. This study aimed to investigate whether SESN2 affects Dox-induced cardiotoxicity and the underlying mechanisms. Sprague-Dawley rats and neonatal rat cardiomyocytes were treated with Dox. SESN2 expression was assessed. The effects of SESN2 on Dox-induced cardiotoxicity were assessed by functional gain and loss experiments. Echocardiographic parameters, morphological and histological analyses, transmission electron microscope and immunofluorescence assays were used to assess cardiac and mitochondrial function. The protein expression of SESN2 was significantly reduced following Dox stimulation. Both knockout of SESN2 by sgRNA and Dox treatment resulted in the inhibition of Parkin-mediated mitophagy, marked cardiomyocytes apoptosis and mitochondria dysfunction. Ectopic expression of SESN2 effectively protected against Dox-induced cardiomyocyte apoptosis, mitochondrial injury and cardiac dysfunction. Mechanistically, SESN2 interacted with Parkin and p62, promoted accumulation of Parkin to mitochondria and then alleviated Dox-caused inhibition of Parkin mediated mitophagy. Ultimately, the clearance of damaged mitochondria and mitochondrial function were improved following SESN2 overexpression. SESN2 protected against Dox-induced cardiotoxicity through improving mitochondria function and mitophagy. These results established SESN2 as a key player in mitochondrial function and provided a potential therapeutic approach to Dox-induced cardiomyopathy.


Assuntos
Cardiomiopatias/etiologia , Doxorrubicina/efeitos adversos , Mitocôndrias/genética , Mitocôndrias/metabolismo , Mitofagia/genética , Peroxidases/genética , Trifosfato de Adenosina/metabolismo , Animais , Apoptose/efeitos dos fármacos , Cardiomiopatias/metabolismo , Cardiomiopatias/patologia , Cardiotoxicidade , Modelos Animais de Doenças , Dosagem de Genes , Genes Mitocondriais , Masculino , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Potencial da Membrana Mitocondrial/genética , Camundongos Transgênicos , Mitocôndrias/ultraestrutura , Modelos Biológicos , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Peroxidases/metabolismo , Ratos , Ratos Sprague-Dawley
9.
Acta Pharmacol Sin ; 40(5): 589-598, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-30030529

RESUMO

High-mobility group box 1 (HMGB1) exhibits various functions according to its subcellular location, which is finely conditioned by diverse post-translational modifications, such as acetylation. The nuclear HMGB1 may prevent from cardiac hypertrophy, whereas its exogenous protein is proven to induce hypertrophic response. This present study sought to investigate the regulatory relationships between poly(ADP-ribose) polymerase 1 (PARP1) and HMGB1 in the process of pathological myocardial hypertrophy. Primary-cultured neonatal rat cardiomyocytes (NRCMs) were respectively incubated with three cardiac hypertrophic stimulants, including angiotensin II (Ang II), phenylephrine (PE), and isoproterenol (ISO), and cell surface area and the mRNA expression of hypertrophic biomarkers were measured. the catalytic activity of PARP1 was remarkably enhanced, meanwhile HMGB1 excluded from the nucleus. PARP1 overexpression by infecting with adenovirus PARP1 (Ad-PARP1) promoted the nuclear export of HMGB1, facilitated its secretion outside the cell, aggravated cardiomyocyte hypertrophy, which could be alleviated by HMGB1 overexpression. PE treatment led to the similar results, while that effect was widely depressed by PARP1 silencing or its specific inhibitor AG14361. Moreover, SD rats were intraperitoneally injected with 3-aminobenzamide (3AB, 20 mg/kg every day, a well-established PARP1 inhibitor) 7 days after abdominal aortic constriction (AAC) surgery for 6 weeks, echocardiography and morphometry of the hearts were measured. Pre-treatment of 3AB relieved AAC-caused the translocation of nuclear HMGB1 protein, cardiac hypertrophy, and heart dysfunction. Our research offers a novel evidence that PARP1 combines with HMGB1 and accelerates its translocation from nucleus to cytoplasm, and the course finally causes cardiac hypertrophy.


Assuntos
Cardiomegalia/etiologia , Núcleo Celular/metabolismo , Proteína HMGB1/metabolismo , Poli(ADP-Ribose) Polimerase-1/metabolismo , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Angiotensina II/farmacologia , Animais , Isoproterenol/farmacologia , Masculino , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Fenilefrina/farmacologia , Ratos Sprague-Dawley
10.
Arch Toxicol ; 93(2): 533-546, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30377735

RESUMO

Doxorubicin (Dox) is an effective anticancer drug, however, its clinical application is restricted by the life-threatening cardiotoxic effects. Secreted Frizzled-related protein 1 (sFRP1) has been reported to participate in both the cancer and cardiovascular diseases and was one of the differential expression genes in normal hearts compared with Dox-treated hearts. Thus, it is important to reveal the potential role of sFRP1 in Dox-induced cardiotoxicity. Here, we show that sFRP1 has a biphasic effect on Dox-induced cardiotoxicity in a location-dependent manner. The secretion of sFRP1 was significantly increased in Dox-treated neonatal rat cardiomyocytes (NRCMs) (1 µM) and SD rats (5 mg/kg/injection at day 1, 5, and 9, i.p.). Adding the anti-sFRP1 antibody (0.5 µg/ml) and inhibiting sFRP1 secretion by caffeine (5 mM) both relieved Dox-induced cardiotoxicity through activating Wnt/ß-catenin signaling, whereas increasing the secretion of sFRP1 by heparin (100 µg/ml) had the opposite effect. The intracellular level of sFRP1 was significantly decreased after Dox treatment both in vitro and in vivo. Knockdown of sFRP1 by sgRNA aggravated Dox-induced cardiotoxicity, while moderate overexpression of sFRP1 by Ad-sFRP1 exhibited protective effect. Besides, poly(ADP-ribosyl) polymerase-1 (PARP1) was screened as an interacting partner of sFRP1 in NRCMs by mass spectrometry. Our results suggested that the intracellular sFRP1 protected NRCMs from Dox-induced cardiotoxicity by interacting with PARP1. Thus, our results provide a novel evidence that sFRP1 has a biphasic effect on Dox-induced cardiotoxicity. In addition, the oversecretion of sFRP1 might be used as a biomarker to indicate the occurrence of cardiotoxicity induced by Dox treatment.


Assuntos
Proteínas Mutadas de Ataxia Telangiectasia/genética , Cardiotoxicidade/etiologia , Doxorrubicina/toxicidade , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Proteínas de Membrana/metabolismo , Animais , Antibióticos Antineoplásicos/efeitos adversos , Antibióticos Antineoplásicos/toxicidade , Apoptose/efeitos dos fármacos , Apoptose/genética , Cardiotoxicidade/metabolismo , Cardiotoxicidade/patologia , Estudos de Casos e Controles , Doxorrubicina/efeitos adversos , Humanos , Masculino , Poli(ADP-Ribose) Polimerase-1/metabolismo , Ratos Sprague-Dawley , Via de Sinalização Wnt/efeitos dos fármacos , Via de Sinalização Wnt/fisiologia , beta Catenina/metabolismo
11.
Acta Pharmacol Sin ; 38(5): 638-650, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28239158

RESUMO

We previously identified AG-690/11026014 (6014) as a novel poly(ADP-ribose) polymerase-1 (PARP-1) inhibitor that effectively prevented angiotensin II (Ang II)-induced cardiomyocyte hypertrophy. In the present study, we reported a new synthesis route for 6014, and investigated its protective effects on Ang II-induced cardiac remodeling and cardiac dysfunction and the underlying mechanisms in mice. We designed a new synthesis route to obtain a sufficient quantity of 6014 for this in vivo study. C57BL/6J mice were infused with Ang II and treated with 6014 (10, 30, 90 mg·kg-1·d-1, ig) for 4 weeks. Then two-dimensional echocardiography was performed to assess the cardiac function and structure. Histological changes of the hearts were examined with HE staining and Masson's trichrome staining. The protein expression was evaluated by Western blot, immunohistochemistry and immunofluorescence assays. The activities of sirtuin-1 (SIRT-1) and the content of NAD+ were detected with the corresponding test kits. Treatment with 6014 dose-dependently improved cardiac function, including LVEF, CO and SV and reversed the changes of cardiac structure in Ang II-infused mice: it significantly ameliorated Ang II-induced cardiac hypertrophy evidenced by attenuating the enlargement of cardiomyocytes, decreased HW/BW and LVW/BW, and decreased expression of hypertrophic markers ANF, BNP and ß-MHC; it also prevented Ang II-induced cardiac fibrosis, as implied by the decrease in excess accumulation of extracellular matrix (ECM) components collagen I, collagen III and FN. Further studies revealed that treatment with 6014 did not affect the expression levels of PARP-1, but dose-dependently inhibited the activity of PARP-1 and subsequently restored the activity of SIRT-1 in heart tissues due to the decreased consumption of NAD+ and attenuated Poly-ADP-ribosylation (PARylation) of SIRT-1. In conclusion, the novel PARP-1 inhibitor 6014 effectively protects mice against AngII-induced cardiac remodeling and improves cardiac function. Thus, 6014 might be a potential therapeutic agent for heart diseases..


Assuntos
Cardiomegalia/terapia , Cardiotônicos/uso terapêutico , Poli(ADP-Ribose) Polimerase-1/antagonistas & inibidores , Tioglicolatos/uso terapêutico , Remodelação Ventricular/efeitos dos fármacos , Xantinas/uso terapêutico , Angiotensina II/farmacologia , Animais , Cardiomegalia/induzido quimicamente , Cardiotônicos/síntese química , Fibrose/induzido quimicamente , Fibrose/terapia , Masculino , Camundongos Endogâmicos C57BL , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/patologia , Sirtuína 1/metabolismo , Tioglicolatos/síntese química , Xantinas/síntese química
12.
Acta Pharmacol Sin ; 38(9): 1257-1268, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28649129

RESUMO

Phosphodiesterase-9A (PDE9A) expression is upregulated during cardiac hypertrophy and heart failure. Accumulating evidence suggests that PDE9A might be a promising therapeutic target for heart diseases. The present study sought to investigate the effects and underlying mechanisms of C33(S), a novel selective PDE9A inhibitor, on cardiac hypertrophy in vitro and in vivo. Treatment of neonatal rat cardiomyocytes (NRCMs) with PE (100 µmol/L) or ISO (1 µmol/L) induced cardiac hypertrophy characterized by significantly increased cell surface areas and increased expression of fetal genes (ANF and BNP). Furthermore, PE or ISO significantly increased the expression of PDE9A in the cells; whereas knockdown of PDE9A significantly alleviated PE-induced hypertrophic responses. Moreover, pretreatment with PDE9A inhibitor C33(S) (50 and 500 nmol/L) or PF-7943 (2 µmol/L) also alleviated the cardiac hypertrophic responses in PE-treated NRCMs. Abdominal aortic constriction (AAC)-induced cardiac hypertrophy and ISO-induced heart failure were established in SD rats. In ISO-treated rats, oral administration of C33(S) (9, 3, and 1 mg·kg-1·d-1, for 3 consecutive weeks) significantly increased fractional shortening (43.55%±3.98%, 54.79%±1.95%, 43.98%±7.96% vs 32.18%±6.28%), ejection fraction (72.97%±4.64%, 84.29%±1.56%, 73.41%±9.37% vs 49.17%±4.20%) and cardiac output (60.01±9.11, 69.40±11.63, 58.08±8.47 mL/min vs 48.97±2.11 mL/min) but decreased the left ventricular internal diameter, suggesting that the transition to heart failure was postponed by C33(S). We further revealed that C33(S) significantly elevated intracellular cGMP levels, phosphorylation of phospholamban (PLB) and expression of SERCA2a in PE-treated NRCMs in vitro and in ISO-induced heart failure model in vivo. Our results demonstrate that C33(S) effectively protects against cardiac hypertrophy and postpones the transition to heart failure, suggesting that it is a promising agent in the treatment of cardiac diseases.


Assuntos
3',5'-AMP Cíclico Fosfodiesterases/antagonistas & inibidores , Cardiomegalia/tratamento farmacológico , GMP Cíclico/metabolismo , Inibidores Enzimáticos/farmacologia , Pirazóis/farmacologia , Pirimidinas/farmacologia , Transdução de Sinais/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos , 3',5'-AMP Cíclico Fosfodiesterases/metabolismo , Animais , Cardiomegalia/metabolismo , Cardiomegalia/patologia , Células Cultivadas , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/administração & dosagem , Inibidores Enzimáticos/química , Masculino , Miócitos Cardíacos/efeitos dos fármacos , Pirazóis/administração & dosagem , Pirazóis/química , Pirimidinas/administração & dosagem , Pirimidinas/química , Ratos , Ratos Sprague-Dawley , Relação Estrutura-Atividade
13.
J Pharmacol Sci ; 132(1): 15-23, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27094369

RESUMO

This study was aimed to investigate the crosstalk between protein kinase C ζ (PKCζ) and signal transducer and activator of transcription 3 (STAT3) in cardiomyocyte hypertrophy. In neonatal rat cardiomyocyte hypertrophic model induced by phenylephrine (PE), the levels of phosphorylated PKCζ and phosphorylated STAT3 were significantly increased, suggesting the activation of both PKCζ and STAT3 in cardiomyocyte hypertrophy. Overexpression of PKCζ by adenovirus infection elevated the expressions of hypertrophic markers atrial natriuretic factor (ANF) and brains natriuretic polypeptide (BNP), as well as the cell surface area; while genetic silencing of PKCζ inhibited PE-induced cardiomyocyte hypertrophy. An interaction between PKCζ and STAT3 in cardiomyocytes was shown by co-immunoprecipitation experiments. Overexpression of PKCζ increased the phosphorylated level of STAT3 at both Ser727 and Tyr705, promoted the nuclear translocation of STAT3, and enhanced the expression of STAT3 downstream target genes c-fos and angiotensinogen (aGT); whereas PKCζ knockdown prevented PE-induced STAT3 activation, nuclear shuttling and transcriptional activation. In conclusion, PKCζ interacts with STAT3 and promotes its activation in cardiomyocyte hypertrophy. Strategies targeting inhibition of PKCζ-STAT3 signaling pathway suggest a therapeutic potential for cardiac hypertrophy.


Assuntos
Cardiomegalia/metabolismo , Miócitos Cardíacos/metabolismo , Proteína Quinase C/metabolismo , Fator de Transcrição STAT3/metabolismo , Animais , Células Cultivadas , Proteínas de Fluorescência Verde/genética , Fenilefrina/farmacologia , Fosforilação , Plasmídeos , Proteína Quinase C/genética , RNA Interferente Pequeno/genética , Ratos , Ratos Sprague-Dawley , Fator de Transcrição STAT3/genética
14.
Chem Biol Interact ; 399: 111132, 2024 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-38964637

RESUMO

The clinical application of doxorubicin (DOX) was limited by the serious cardiotoxicity. The traditional Chinese medicine Andrographis paniculata and its principal active component (Dehydroandrographolide, DA) have been well known for their diverse cardiovascular protective effects. However, the effects of DA on DOX-induced cardiotoxicity (DIC) were still unknown. In this study, we evaluated the effects and revealed the potential mechanisms of DA on DIC both in vivo and in vitro. The effects of DA on DIC were systematically assessed by echocardiography and histological assays. Western blot and flow cytometry were used to measure apoptosis of cardiomyocytes. Transmission electron microscopy and StubRFP-SensGFP-LC3 lentivirus were further used to assay autophagic flux. Our results showed that DA administration significantly improved cardiac function and attenuated DOX-induced cardiomyocyte apoptosis. Mechanically, DA restored autophagic flux and lysosome functions via inhibiting DOX-induced mTOR signal pathway activation and increasing the translocation of TFEB to the nucleus. However, activation of mTOR or knockdown of TFEB significantly inhibited the protective effects of DA against DIC by impacting lysosomal functions and autophagic flux. In conclusion, our results revealed that DA might be a potential cardioprotective agent against DIC.


Assuntos
Autofagia , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos , Cardiotoxicidade , Diterpenos , Doxorrubicina , Miócitos Cardíacos , Transdução de Sinais , Serina-Treonina Quinases TOR , Animais , Doxorrubicina/toxicidade , Autofagia/efeitos dos fármacos , Diterpenos/farmacologia , Diterpenos/química , Serina-Treonina Quinases TOR/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Transdução de Sinais/efeitos dos fármacos , Cardiotoxicidade/prevenção & controle , Apoptose/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Ratos , Ratos Sprague-Dawley
15.
Cell Rep ; 43(10): 114839, 2024 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-39392752

RESUMO

Lysine acetylation is a conserved post-translational modification involved in energy metabolism in mitochondria and heart function. This study investigates the role of mitochondria-localized lysine acetyltransferase MOF (males absent on the first) in heart failure (HF). We find that MOF is upregulated in mitochondria during HF, and overexpression of mitochondria-targeted MOF (mtMOF) in mouse models results in mitochondria dysfunction, cardiac remodeling, and HF. Furthermore, sirtuin 3 (SIRT3) knockout aggravates mtMOF-induced damages, underscoring the role of MOF-catalyzed hyperacetylation in HF. Quantitative lysine acetylome analysis identifies ATP5B as a substrate of MOF. We demonstrate that the acetylation of ATP5B at K201, co-regulated by MOF and SIRT3, impairs mitochondrial respiration and energy metabolism both in vitro and in vivo. These findings suggest that the role of MOF in HF could be attributed to its regulation of ATP5B acetylation. Overall, our results highlight the disruptive impact of mitochondrial MOF on cardiac function and emphasize the significance of enzyme-catalyzed acetylation in mitochondria.

16.
Acta Pharm Sin B ; 13(12): 4875-4892, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38045054

RESUMO

The clinical utilization of doxorubicin (Dox) in various malignancies is restrained by its major adverse effect: irreversible cardiomyopathy. Extensive studies have been done to explore the prevention of Dox cardiomyopathy. Currently, ferroptosis has been shown to participate in the incidence and development of Dox cardiomyopathy. Sorting Nexin 3 (SNX3), the retromer-associated cargo binding protein with important physiological functions, was identified as a potent therapeutic target for cardiac hypertrophy in our previous study. However, few study has shown whether SNX3 plays a critical role in Dox-induced cardiomyopathy. In this study, a decreased level of SNX3 in Dox-induced cardiomyopathy was observed. Cardiac-specific Snx3 knockout (Snx3-cKO) significantly alleviated cardiomyopathy by downregulating Dox-induced ferroptosis significantly. SNX3 was further demonstrated to exacerbate Dox-induced cardiomyopathy via induction of ferroptosis in vivo and in vitro, and cardiac-specific Snx3 transgenic (Snx3-cTg) mice were more susceptible to Dox-induced ferroptosis and cardiomyopathy. Mechanistically, SNX3 facilitated the recycling of transferrin 1 receptor (TFRC) via direct interaction, disrupting iron homeostasis, increasing the accumulation of iron, triggering ferroptosis, and eventually exacerbating Dox-induced cardiomyopathy. Overall, these findings established a direct SNX3-TFRC-ferroptosis positive regulatory axis in Dox-induced cardiomyopathy and suggested that targeting SNX3 provided a new effective therapeutic strategy for Dox-induced cardiomyopathy through TFRC-dependent ferroptosis.

17.
Front Med (Lausanne) ; 9: 1075465, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36714100

RESUMO

Background: Acute lung injury (ALI) is a life-threatening inflammatory disease without effective therapeutic regimen. Macrophage polarization plays a key role in the initiation and resolution of pulmonary inflammation. Therefore, modulating macrophage phenotype is a potentially effective way for acute lung injury. Cryptotanshinone (CTS) is a lipophilic bioactive compound extracted from the root of Salvia miltiorrhiza with a variety of pharmacological effects, especially the anti-inflammatory role. In this study, we investigated the therapeutic and immunomodulatory effects of CTS on ALI. Materials and methods: The rat model of ALI was established by intratracheal instillation of LPS (5 mg/kg) to evaluate the lung protective effect of CTS in vivo and to explore the regulation of CTS on the phenotype of lung macrophage polarization. LPS (1 µg/mL) was used to stimulate RAW264.7 macrophages in vitro to further explore the effect of CTS on the polarization and metabolic reprogramming of RAW264.7 macrophages and to clarify the potential mechanism of CTS anti-ALI. Results: CTS significantly improved lung function, reduced pulmonary edema, effectively inhibited pulmonary inflammatory infiltration, and alleviated ALI. Both in vivo and in vitro results revealed that CTS inhibited the differentiation of macrophage into the M1 phenotype and promoted polarization into M2 phenotype during ALI. Further in vitro studies indicated that CTS significantly suppressed LPS-induced metabolic transition from aerobic oxidation to glycolysis in macrophages. Mechanistically, CTS blocked LPS-induced metabolic transformation of macrophages by activating AMPK. Conclusion: These findings demonstrated that CTS regulates macrophage metabolism by activating AMPK, and then induced M1-type macrophages to transform into M2-type macrophages, thereby alleviating the inflammatory response of ALI, suggesting that CTS might be a potential anti-ALI agent.

18.
Int J Biol Sci ; 18(2): 783-799, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35002525

RESUMO

Poly(ADP-ribosyl)ation (PARylation) and SUMO modification (SUMOylation) are novel post-translational modifications (PTMs) mainly induced by PARP1 and SUMO1. Growing evidence has revealed that C/EBPß plays multiple roles in biological processes and participates in cardiovascular diseases. However, the cross-talk between C/EBPß PARylation and SUMOylation during cardiovascular diseases is unknown. This study aims to investigate the effects of C/EBPß PTMs on cardiac hypertrophy and its underlying mechanism. Abdominal aortic constriction (AAC) and phenylephrine (PE) were conducted to induce cardiac hypertrophy. Intramyocardial delivery of recombinant adenovirus (Ad-PARP1) was taken to induce PARP1 overexpression. In this study, we found C/EBPß participates in PARP1-induced cardiac hypertrophy. C/EBPß K134 residue could be both PARylated and SUMOylated individually by PARP1 and SUMO1. Moreover, the accumulation of PARylation on C/EBPß at K134 site exhibits downregulation of C/EBPß SUMOylation at the same site. Importantly, C/EBPß K134 site SUMOylation could decrease C/EBPß protein stability and participates in PARP1-induced cardiac hypertrophy. Taken together, these findings highlight the importance of the cross-talk between C/EBPß PTMs at K134 site in determining its protein level and function, suggesting that multi-target pharmacological strategies inhibiting PARP1 and activating C/EBPß SUMOylation would be potential for treating pathological cardiac hypertrophy.


Assuntos
Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Cardiomegalia/metabolismo , Poli(ADP-Ribose) Polimerase-1/metabolismo , Proteína SUMO-1/metabolismo , Animais , Cardiomegalia/genética , Modelos Animais de Doenças , Células HEK293 , Humanos , Masculino , Poli(ADP-Ribose) Polimerase-1/antagonistas & inibidores , Poli(ADP-Ribose) Polimerase-1/genética , Ligação Proteica , Processamento de Proteína Pós-Traducional , Ratos , Ratos Sprague-Dawley , Proteína SUMO-1/genética , Sumoilação
19.
Front Pharmacol ; 13: 798436, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35237161

RESUMO

Doxorubicin (Dox) is a chemotherapeutic drug used to treat a wide range of cancers, but its clinical application is limited due to its cardiotoxicity. Protein kinase C-ζ (PKC-ζ) is a serine/threonine kinase belonging to atypical protein kinase C (PKC) subfamily, and is activated by its phosphorylation. We and others have reported that PKC-ζ induced cardiac hypertrophy by activating the inflammatory signaling pathway. This study focused on whether PKC-ζ played an important role in Dox-induced cardiotoxicity. We found that PKC-ζ phosphorylation was increased by Dox treatment in vivo and in vitro. PKC-ζ overexpression exacerbated Dox-induced cardiotoxicity. Conversely, knockdown of PKC-ζ by siRNA relieved Dox-induced cardiotoxicity. Similar results were observed when PKC-ζ enzyme activity was inhibited by its pseudosubstrate inhibitor, Myristoylated. PKC-ζ interacted with ß-catenin and inhibited Wnt/ß-catenin signaling pathway. Activation of Wnt/ß-catenin signaling by LiCl protected against Dox-induced cardiotoxicity. The Wnt/ß-catenin inhibitor XAV-939 aggravated Dox-caused decline of ß-catenin and cardiomyocyte apoptosis and mitochondrial damage. Moreover, activation of Wnt/ß-catenin suppressed aggravation of Dox-induced cardiotoxicity due to PKC-ζ overexpression. Taken together, our study revealed that inhibition of PKC-ζ activity was a potential cardioprotective approach to preventing Dox-induced cardiac injury.

20.
Front Pharmacol ; 13: 986510, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36238566

RESUMO

Background: Ulcerative colitis (UC) is one type of inflammatory bowel disease, characterized by inflammation with infiltration and activation of macrophages in colonic tissue. LH011 is a trypsin inhibitor with potential anti-inflammatory effect. Purpose: Here, we aim to assay the effects of LH011 on UC and further investigate the potential mechanisms in vitro and in vivo. Methods: Dextran sulfate sodium (DSS, 3.5%, w/v) was used to induce UC, and lipopolysaccharide (LPS) was used to induce inflammation in RAW 264.7 cells. LH011 was administrated to mice in vivo or to RAW 264.7 cells in vitro at different concentrations. The cytokines (IL-1ß, IL-6, and TNF-α) and the changes of NF-κB and Nrf2 pathways were detected. Results: The results showed that LH011 improved DSS-induced mice colitis, including loss of weight, disease activity index (DAI), and colonic pathological damage. In addition, LH011 inhibited the expressions of IL-1ß, IL-6, and TNF-α and strengthened the anti-oxidative capacity. Mechanically, LH011 downregulated the nuclear localization of NF-κB p65 and upregulated the protein expression of Nrf2. Conclusion: These results demonstrated that LH011 alleviated inflammation and oxidative stress during UC by inhibiting TLR4/NF-κB and activating Nrf2/Keap1/HO-1 signaling pathways.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA