Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Acta Pharmacol Sin ; 44(5): 999-1013, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-36347996

RESUMO

Non-healing diabetic wounds (DW) are a serious clinical problem that remained poorly understood. We recently found that topical application of growth differentiation factor 11 (GDF11) accelerated skin wound healing in both Type 1 DM (T1DM) and genetically engineered Type 2 diabetic db/db (T2DM) mice. In the present study, we elucidated the cellular and molecular mechanisms underlying the action of GDF11 on healing of small skin wound. Single round-shape full-thickness wound of 5-mm diameter with muscle and bone exposed was made on mouse dorsum using a sterile punch biopsy 7 days following the onset of DM. Recombinant human GDF11 (rGDF11, 50 ng/mL, 10 µL) was topically applied onto the wound area twice a day until epidermal closure (maximum 14 days). Digital images of wound were obtained once a day from D0 to D14 post-wounding. We showed that topical application of GDF11 accelerated the healing of full-thickness skin wounds in both type 1 and type 2 diabetic mice, even after GDF8 (a muscle growth factor) had been silenced. At the cellular level, GDF11 significantly facilitated neovascularization to enhance regeneration of skin tissues by stimulating mobilization, migration and homing of endothelial progenitor cells (EPCs) to the wounded area. At the molecular level, GDF11 greatly increased HIF-1ɑ expression to enhance the activities of VEGF and SDF-1ɑ, thereby neovascularization. We found that endogenous GDF11 level was robustly decreased in skin tissue of diabetic wounds. The specific antibody against GDF11 or silence of GDF11 by siRNA in healthy mice mimicked the non-healing property of diabetic wound. Thus, we demonstrate that GDF11 promotes diabetic wound healing via stimulating endothelial progenitor cells mobilization and neovascularization mediated by HIF-1ɑ-VEGF/SDF-1ɑ pathway. Our results support the potential of GDF11 as a therapeutic agent for non-healing DW.


Assuntos
Diabetes Mellitus Experimental , Células Progenitoras Endoteliais , Fatores de Diferenciação de Crescimento , Cicatrização , Animais , Humanos , Camundongos , Proteínas Morfogenéticas Ósseas/metabolismo , Quimiocina CXCL12/efeitos dos fármacos , Quimiocina CXCL12/metabolismo , Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Experimental/tratamento farmacológico , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 2/tratamento farmacológico , Diabetes Mellitus Tipo 2/metabolismo , Células Progenitoras Endoteliais/metabolismo , Células Progenitoras Endoteliais/patologia , Fatores de Diferenciação de Crescimento/uso terapêutico , Fatores de Diferenciação de Crescimento/metabolismo , Neovascularização Fisiológica , Fator A de Crescimento do Endotélio Vascular/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/metabolismo , Cicatrização/efeitos dos fármacos , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/uso terapêutico , Subunidade alfa do Fator 1 Induzível por Hipóxia/efeitos dos fármacos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo
2.
J Pineal Res ; 64(2)2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-29024030

RESUMO

Atherosclerosis (AS) is an inflammatory disease linked to endothelial dysfunction. Melatonin is reported to possess substantial anti-inflammatory properties, which has proven to be effective in AS. Emerging literature suggests that pyroptosis plays a critical role during AS progression. However, whether pyroptosis contributes to endothelial dysfunction and the underlying molecular mechanisms remained unexploited. This study was designed to investigate the antipyroptotic effects of melatonin in atherosclerotic endothelium and to elucidate the potential mechanisms. In this study, high-fat diet (HFD)-treated ApoE-/- mice were used as an atherosclerotic animal model. We found intragastric administration of melatonin for 12 weeks markedly reduced the atherosclerotic plaque in aorta. Meanwhile, melatonin also attenuated the expression of pyroptosis-related genes, including NLRP3, ASC, cleaved caspase1, NF-κB/GSDMD, GSDMD N-termini, IL-1ß, and IL-18 in aortic endothelium of melatonin-treated animals. Consistent antipyroptotic effects were also observed in ox-LDL-treated human aortic endothelial cells (HAECs). We found that lncRNA MEG3 enhanced pyroptosis in HAECs. Moreover, MEG3 acted as an endogenous sponge by sequence complementarity to suppress the function of miR-223 and to increase NLRP3 expression and enhance endothelial cell pyroptosis. Furthermore, knockdown of miR-223 blocked the antipyroptotic actions of melatonin in ox-LDL-treated HAECs. Together, our results suggest that melatonin prevents endothelial cell pyroptosis via MEG3/miR-223/NLRP3 axis in atherosclerosis, and therefore, melatonin replacement might be considered a new strategy for protecting endothelium against pyroptosis, thereby for the treatment of atherosclerosis associated with pyroptosis.


Assuntos
Anti-Inflamatórios/farmacologia , Células Endoteliais/efeitos dos fármacos , Melatonina/farmacologia , Piroptose/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Animais , Aterosclerose/patologia , Dieta Hiperlipídica/efeitos adversos , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Regulação da Expressão Gênica/efeitos dos fármacos , Camundongos , Camundongos Knockout , MicroRNAs/biossíntese , Proteína 3 que Contém Domínio de Pirina da Família NLR/biossíntese , RNA Longo não Codificante/biossíntese
3.
Cell Death Differ ; 30(3): 605-617, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-34628486

RESUMO

Non-small cell lung cancer (NSCLC) has the highest mortality rate among all malignancies worldwide. The role of long noncoding RNAs (lncRNAs) in the progression of cancers is a contemporary research hotspot. Based on an integrative analysis of The Cancer Genome Atlas database, we identified lncRNA-RNA Component of Mitochondrial RNA Processing Endoribonuclease (RMRP) as one of the most highly upregulated lncRNAs that are associated with poor survival in NSCLC. Furthermore, N(6)-methyladenosine (m6A) was highly enriched within RMRP and enhanced its RNA stability. In vitro and in vivo experiments showed that RMRP promoted NSCLC cell proliferation, invasion, and migration. In terms of mechanism, RMRP recruited YBX1 to the TGFBR1 promotor region, leading to upregulation of the transcription of TGFBR1. The TGFBR1/SMAD2/SMAD3 pathway was also regulated by RMRP. In addition, RMRP promoted the cancer stem cells properties and epithelial mesenchymal transition, which promote the resistance to radiation therapy and cisplatin. Clinical data further confirmed a positive correlation between RMRP and TGFBR1. In short, our work reveals that m6A RNA methylation-mediated RMRP stability renders proliferation and progression of NSCLC through regulating TGFBR1/SMAD2/SMAD3 pathway.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , MicroRNAs , RNA Longo não Codificante , Humanos , Carcinoma Pulmonar de Células não Pequenas/genética , MicroRNAs/metabolismo , Metilação , Neoplasias Pulmonares/patologia , RNA Longo não Codificante/genética , Receptor do Fator de Crescimento Transformador beta Tipo I/metabolismo , Estabilidade de RNA , Proliferação de Células/genética , Regulação Neoplásica da Expressão Gênica , Linhagem Celular Tumoral , Proteína Smad3/genética , Proteína Smad2/metabolismo
4.
Mol Med Rep ; 26(4)2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-36004471

RESUMO

Subsequently to the publication of this paper, the authors have realized that Fig. 2 was published containing some incorrectly assembled data panels. The E­cadherin control data panel in Fig. 3F was re­used in Fig. 2C; furthermore, the HG / Vimentin data panel in Fig. 4E was re­used in Fig. 2D. The authors have re­examined their original data, and were able to identify that Fig. 2 contained the erroneously assembled data panels. The revised version of Fig. 2, showing the correct E­cadherin control data panel for Fig. 2C and the correct HG / Vimentin data panel for Fig. 2D, is shown below. It was also noted that the white rectangles were not explained in the figure legend; these represent an enlargement of the cells in the E­cad/vimentin panels, and the details are now included in the figure legend (shown in bold). Note that these errors did not significantly affect either the results or the conclusions reported in this paper, and all the authors agree to the publication of this corrigendum. Furthermore, the authors thank the Editor of Molecular Medicine Reports for allowing them the opportunity to publish this corrigendum, and apologize to the readership for any inconvenience caused. [Molecular Medicine Reports 19: 3190­3200, 2019; DOI: 10.3892/mmr.2019.9986].

5.
J Pharm Pharmacol ; 72(1): 76-83, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31702064

RESUMO

OBJECTIVES: Metoprolol is regarded as a first-line medicine for the treatment of myocardial infarction (MI). However, the underlying mechanisms remain largely unknown. This study aimed to investigate the involvement of miR-1 in the pharmacological function of metoprolol. METHODS: In vivo MI model was established by left anterior descending coronary artery (LAD) ligation. The effects of metoprolol on infarct size and cardiac dysfunction were determined by triphenyltetrazolium chloride staining and cardiac echocardiography, respectively. In vitro oxidative stress cardiomyocyte model was established by H2 O2 treatment. The effect of metoprolol on the expression of miR-1 and connexin43 (Cx43) was quantified by real-time PCR and western blot, respectively. The intercellular communication was evaluated by lucifer yellow dye diffusion. KEY FINDINGS: Left anterior descending ligation-induced MI injury was markedly attenuated by metoprolol as shown by reduced infarct size and better cardiac function. Metoprolol reversed the up-regulation of miR-1 and down-regulation of Cx43 in MI heart. Moreover, in H2 O2 -stimulated cardiomyocytes, overexpression of miR-1 abolished the effects of metoprolol on Cx43 up-regulation and increased intercellular communication, indicating that miR-1 may be a necessary mediator for the cardiac protective function of metoprolol. CONCLUSIONS: Metoprolol relieves MI injury via suppression miR-1, thus increasing its target protein Cx43 and improving intercellular communication.


Assuntos
Antagonistas de Receptores Adrenérgicos beta 1/farmacologia , Metoprolol/farmacologia , MicroRNAs/metabolismo , Infarto do Miocárdio/prevenção & controle , Miócitos Cardíacos/efeitos dos fármacos , Animais , Comunicação Celular/efeitos dos fármacos , Células Cultivadas , Conexina 43/metabolismo , Modelos Animais de Doenças , Regulação para Baixo , Junções Comunicantes/efeitos dos fármacos , Junções Comunicantes/metabolismo , Junções Comunicantes/patologia , Masculino , MicroRNAs/genética , Infarto do Miocárdio/genética , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/fisiopatologia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Ratos Wistar , Volume Sistólico/efeitos dos fármacos , Função Ventricular Esquerda/efeitos dos fármacos
6.
Cell Death Dis ; 11(10): 917, 2020 10 25.
Artigo em Inglês | MEDLINE | ID: mdl-33100331

RESUMO

NLRP3 (Nucleotide-binding oligomerization domain-like receptor pyrin domain-containing 3) inflammasome-mediated cardiomyocytes pyroptosis plays a crucial part in progression of acute myocardial infarction (MI). GDF11 (Growth Differentiation Factor 11) has been reported to generate cytoprotective effects in phylogenesis and multiple diseases, but the mechanism that GDF11 contributes to cardioprotection of MI and cardiomyocytes pyroptosis remains poorly understood. In our study, we first determined that GDF11 was abnormally downregulated in the heart tissue of MI mice and hypoxic cardiomyocytes. Moreover, AAV9-GDF11 markedly alleviated heart function in MI mice. Meanwhile, GDF11 overexpression also decreased the pyroptosis of hypoxic cardiomyocytes. PROMO and JASPAR prediction software found that transcription factor HOXA3 was predicted as an important regulator of NLRP3, and was confirmed by ChIP assay. Further analysis identifying GDF11 promoted the Smad2/3 pathway resulted in HOXA3 overexpression. Taken together, our study implies that GDF11 prevents cardiomyocytes pyroptosis via HOXA3/NLRP3 signaling pathway in MI mice.


Assuntos
Proteínas Morfogenéticas Ósseas/uso terapêutico , Cardiotônicos/uso terapêutico , Fatores de Diferenciação de Crescimento/uso terapêutico , Proteínas de Homeodomínio/metabolismo , Infarto do Miocárdio/genética , Animais , Proteínas Morfogenéticas Ósseas/farmacologia , Cardiotônicos/farmacologia , Modelos Animais de Doenças , Fatores de Diferenciação de Crescimento/farmacologia , Humanos , Masculino , Camundongos , Piroptose , Transfecção , Regulação para Cima
7.
Theranostics ; 10(12): 5276-5289, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32373212

RESUMO

Rationale: Nicotine exposure via cigarette smoking is strongly associated with atherosclerosis. However, the underlying mechanisms remain poorly understood. The current study aimed to identify whether endothelial to mesenchymal transition (EndMT) contributes to nicotine-induced atherosclerosis. Methods: ApoE-/- mice were administered nicotine in their drinking water for 12 weeks. The effects of nicotine on EndMT were determined by immunostaining on aortic root and RNA analysis in aortic intima. In vitro nicotine-treated cell model was established on human aortic endothelial cells (HAECs). The effects of nicotine on the expression of EndMT-related markers, ERK1/2 and Snail were quantified by real-time PCR, western blot and immunofluorescent staining. Results: Nicotine treatment resulted in larger atherosclerotic plaques in ApoE-/- mice. The vascular endothelial cells from nicotine-treated mice showed mesenchymal phenotype, indicating EndMT. Moreover, nicotine-induced EndMT process was accompanied by cytoskeleton reorganization and impaired barrier function. The α7 nicotine acetylcholine receptor (α7nAChR) was highly expressed in HAECs and its antagonist could effectively relieve nicotine-induced EndMT and atherosclerotic lesions in mice. Further experiments revealed that ERK1/2 signaling was activated by nicotine, which led to the upregulation of Snail. Blocking ERK1/2 with inhibitor or silencing Snail by small interfering RNA efficiently preserved endothelial phenotype upon nicotine stimulation. Conclusion: Our study provides evidence that EndMT contributes to the pro-atherosclerotic property of nicotine. Nicotine induces EndMT through α7nAChR-ERK1/2-Snail signaling in endothelial cells. EndMT may be a therapeutic target for smoking-related endothelial dysfunction and cardiovascular disease.


Assuntos
Aterosclerose/induzido quimicamente , Transição Epitelial-Mesenquimal/fisiologia , Nicotina/toxicidade , Placa Aterosclerótica/induzido quimicamente , Animais , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Aterosclerose/metabolismo , Western Blotting , Transição Epitelial-Mesenquimal/genética , Feminino , Humanos , Camundongos , Placa Aterosclerótica/metabolismo , Reação em Cadeia da Polimerase em Tempo Real
8.
Eur J Pharmacol ; 885: 173495, 2020 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-32861661

RESUMO

GDF11 has been reported to play a critical role in rejuvenating hypertrophy heart, skeletal muscle, and blood vessel regeneration in aged mice. Whether GDF11 can regulate autophagy in cardiomyocytes remains largely unknown. Thus, the purpose of the present study was to investigate the effects of GDF11 on cardiomyocyte autophagy induced by hypoxia, in addition to the underlying mechanisms. By using the MTT assay, Flow cytometry assay, LIVE/DEAD® Viability/Cytotoxicity Kit Stains and TUNEL assay, we found that exogenous GDF11 decreased apoptosis caused by prolonged hypoxia in cardiomyocytes. The expression of GDF11 was decreased obviously both in the cardiac tissue of myocardial infarction mice and the hypoxia treated cardiomyocytes. Protein levels of cleaved caspase-3, p-AMPK, SQSTM1, LC3B-I/II and GDF11 were detected by western blot. Autophagosomes and autolysosomes were identified by confocal laser microscopy after transfecting with the mRFP-eGFP-LC3 plasmids. Antibody against GDF11 (anti-GDF11) was used to inhibit the function of GDF11. At the molecular level, exogenous GDF11 increased AMPK function and enhanced autophagy activity. Anti-GDF11 inhibited autophagy and aggravated hypoxia-induced apoptosis in cardiomyocytes. Thus, GDF11 might be a potential target for myocardial infarction therapy.


Assuntos
Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Proteínas Morfogenéticas Ósseas/genética , Hipóxia Celular/efeitos dos fármacos , Fatores de Diferenciação de Crescimento/genética , Miócitos Cardíacos/efeitos dos fármacos , Animais , Anticorpos Bloqueadores/farmacologia , Proteínas Morfogenéticas Ósseas/antagonistas & inibidores , Proteínas Morfogenéticas Ósseas/efeitos dos fármacos , Eletrocardiografia/efeitos dos fármacos , Fatores de Diferenciação de Crescimento/antagonistas & inibidores , Fatores de Diferenciação de Crescimento/efeitos dos fármacos , Lisossomos/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fagossomos/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley
9.
Mol Med Rep ; 19(4): 3190-3200, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30816482

RESUMO

Hepatic fibrosis is characterized by the aberrant production and deposition of extracellular matrix (ECM) proteins. Growing evidence indicates that the epithelial­mesenchymal transition serves a crucial role in the progression of liver fibrogenesis. Although a subset of microRNAs (miRNAs or miRs) has recently been identified as essential regulators of the EMT gene expression, studies of the EMT in hyperglycemic­induced liver fibrosis are limited. In the current study, it was observed that high glucose­treated AML12 cells occurred EMT process, and miR­32 expression was markedly increased in the liver tissue of streptozotocin­induced diabetic rats and in high glucose­treated AML12 cells. Additionally, the contribution of the EMT to liver fibrosis by targeting metastasis­associated gene 3 (MTA3) under hyperglycemic conditions was suppressed by AMO­32. The results indicated that miR­32 and MTA3 may be considered as novel drug targets in the prevention and treatment of liver fibrosis under hyperglycemic conditions. These finding improves the understanding of the progression of liver fibrogenesis.


Assuntos
Transição Epitelial-Mesenquimal/genética , Glucose/metabolismo , Cirrose Hepática/etiologia , Cirrose Hepática/metabolismo , MicroRNAs/genética , Proteínas de Neoplasias/genética , Animais , Biomarcadores , Células Cultivadas , Diabetes Mellitus Experimental , Modelos Animais de Doenças , Progressão da Doença , Suscetibilidade a Doenças , Regulação da Expressão Gênica , Glucose/efeitos adversos , Hiperglicemia/complicações , Hiperglicemia/metabolismo , Cirrose Hepática/patologia , Masculino , Interferência de RNA , Ratos
10.
Acta Pharm Sin B ; 9(4): 724-733, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-31384533

RESUMO

Excess activation of cardiac fibroblasts inevitably induces cardiac fibrosis. Emodin has been used as a natural medicine against several chronic diseases. The objective of this study is to determine the effects of emodin on cardiac fibrosis and the underlying molecular mechanisms. Intragastric administration of emodin markedly decreased left ventricular wall thickness in a mouse model of pathological cardiac hypertrophy with excess fibrosis induced by transaortic constriction (TAC) and suppressed activation of cardiac fibroblasts induced by angiotensin II (AngII). Emodin upregulated expression of metastasis associated protein 3 (MTA3) and restored the MTA3 expression in the setting of cardiac fibrosis. Moreover, overexpression of MTA3 promoted cardiac fibrosis; in contrast, silence of MTA3 abrogated the inhibitory effect of emodin on fibroblast activation. Our findings unraveled the potential of emodin to alleviate cardiac fibrosis via upregulating MTA3 and highlight the regulatory role of MTA3 in the development of cardiac fibrosis.

11.
Mol Med Rep ; 19(5): 3527-3536, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-30896796

RESUMO

Myocardial infarction (MI) is the most common event in cardiovascular disease. Carvedilol, a ß­blocker with multiple pleiotropic actions, is widely used for the treatment cardiovascular diseases. However, the underlying mechanisms of carvedilol on alleviating MI are not fully understood. The aim of the present study was to investigate whether the beneficial effects of carvedilol were associated with regulation of microRNA­1 (miR­1). It was demonstrated that carvedilol ameliorated impaired cardiac function and decreased infarct size in a rat model of MI induced by coronary artery occlusion. Similarly, carvedilol reversed the H2O2­induced decrease in cardiomyocyte viability in a dose­dependent manner. The in vivo and in vitro models demonstrated the downregulation of miR­1 following treatment with carvedilol. Overexpression of miR­1, a known pro­apoptotic miRNA, decreased cell viability and induced cell apoptosis. Transfection of miR­1 abolished the beneficial effects of carvedilol. The expression of heat shock protein 60 (HSP60), a direct target of miR­1, was identified to be decreased in MI and H2O2­induced apoptosis, which was associated with a decrease in Bcl­2 and an increase in Bax; expression was restored following treatment with carvedilol. It was concluded that carvedilol partially exhibited its beneficial effects by downregulating miR­1 and increasing HSP60 expression. miR­1 has become a member of the group of carvedilol­responsive miRNAs. Future studies are required to fully elucidate the potential overlapping or compensatory effects of known carvedilol­responsive miRNAs and their underlying mechanisms of action in the pathophysiology of cardiovascular diseases.


Assuntos
Apoptose/efeitos dos fármacos , Apoptose/genética , Carvedilol/farmacologia , Chaperonina 60/genética , MicroRNAs/genética , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Interferência de RNA , Animais , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Citoproteção/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Infarto do Miocárdio/etiologia , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/patologia , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Ratos , Proteína X Associada a bcl-2/metabolismo
12.
Int J Cardiol ; 295: 7-13, 2019 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-31399301

RESUMO

BACKGROUND: Vascular aging has profound effects on cardiovascular diseases. Endothelial to mesenchymal transition (EndMT) is defined as the acquisition of mesenchymal characteristics by endothelial cells (ECs) and has been found induced in a model of ECs aging. However, whether EndMT occurs during aging in vivo, the functional significance of EndMT on vascular biology and the underlying mechanisms remain unknown. METHODS AND RESULTS: In this study, we examined the vascular ECs from young (2 months old) and old (18 months old) mice, and demonstrated that aged ECs underwent EndMT. Moreover, the transwell assay showed that EndMT process was accompanied by increased endothelial permeability. It was found that sirtuin 6 (SIRT6), a nicotinamide adenine dinucleotide+ (NAD+)-dependent histone deacetylase, was down-regulated during ECs aging. Knockdown of SIRT6 in young ECs could induce EndMT. Next, we identified five long non-coding RNAs that are enriched in ECs for downstream effector of SIRT6; only metastasis associated lung adenocarcinoma transcript 1 (MALAT1) was significantly up-regulated in aged ECs. Knockdown of SIRT6 could increase MALAT1 levels. Furthermore, the ChIP assay and luciferase reporter gene assay confirmed that SIRT6 bound directly to the promoter region of MALAT1 and suppressed MALAT1 expression. Finally, we demonstrated that MALAT1 mediated aging-induced EndMT through increasing Snail expression. CONCLUSION: Our study provides in vivo evidence that ECs undergo EndMT during vascular aging, which increases endothelial permeability. SIRT6-mediated transcriptional suppression of MALAT1 is a key mechanism for EndMT. Manipulating EndMT may be considered as a new therapeutic strategy for retarding aging-associated vascular diseases.


Assuntos
Envelhecimento/fisiologia , Endotélio Vascular/metabolismo , Transição Epitelial-Mesenquimal/genética , Regulação da Expressão Gênica , RNA Longo não Codificante/genética , Sirtuínas/genética , Doenças Vasculares/genética , Animais , Células Cultivadas , Modelos Animais de Doenças , Endotélio Vascular/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , RNA Longo não Codificante/metabolismo , Transdução de Sinais , Sirtuínas/metabolismo , Doenças Vasculares/metabolismo , Doenças Vasculares/patologia
13.
Phytomedicine ; 59: 152758, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-31004884

RESUMO

BACKGROUND: Emerging evidence has indicated the therapeutic potential of emodin with its multiple pharmacological effects. PURPOSE: To evaluate role of emodin in regulating insulin resistance (IR) and to elucidate the underlying molecular mechanisms. STUDY DESIGN/METHODS: Fasting blood glucose (FBG) and lipid levels were measured before and after intragastric administration of emodin in type 2 diabetes mellitus (T2DM) rats. Glucose consumption was determined in L6 cells to investigate the effect of emodin on glucose metabolism. Expression of miR-20b and SMAD7 was quantified by real-time PCR for mRNAs or western blot analysis for proteins. RESULTS: Emodin ameliorated hyperglycemia and dyslipidemia in T2DM rats, and glucose metabolism in a concentration- and time-dependent manner. MiR-20b was markedly upregulated in the setting of IR and overexpression of miR-20b disrupted glucose metabolism by repressing SMAD7 in L6 cells. Knockdown of this miRNA produced the opposite effects. Emodin abolished the abnormal upregulation of miR-20b and indirectly upregulated SMAD7. CONCLUSION: Emodin improves glucose metabolism to produce anti-IR effects, and downregulation of miR-20b thereby upregulation of SMAD7 is an underlying mechanism for the beneficial effects of emodin.


Assuntos
Diabetes Mellitus Tipo 2/metabolismo , Emodina/farmacologia , Glucose/metabolismo , Resistência à Insulina , Insulina/metabolismo , Músculo Esquelético/efeitos dos fármacos , RNA Mensageiro , Animais , Diabetes Mellitus Tipo 2/tratamento farmacológico , Regulação para Baixo/efeitos dos fármacos , Emodina/uso terapêutico , Transportador de Glucose Tipo 4/metabolismo , Hiperglicemia/metabolismo , Hiperglicemia/prevenção & controle , Masculino , Músculo Esquelético/metabolismo , RNA Mensageiro/metabolismo , Ratos , Proteína Smad7/metabolismo , Regulação para Cima/efeitos dos fármacos
15.
Eur J Pharmacol ; 838: 170-176, 2018 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-30170066

RESUMO

Elimination of glucose transporter 4 (GLUT4) inevitably induces insulin resistance (IR), aggravating inflammation- and oxidative stress-related disorders. However, the underlying molecular mechanisms remain incompletely understood. In this study, we identified miR-17 as an important regulator of IR by targeting GLUT4. MiR-17 expression was found significantly elevated in skeletal tissues of rats with type 2 diabetes mellitus (T2DM), along with marked downregulation of GLUT4 protein level. Luciferase reporter gene assay demonstrated a direct interaction between miR-17 and the 3'untranslated region of GLUT4 mRNA. Correlation analyses (Spearman, Pearson, and Kendall) revealed that miR-17 level was negatively correlated with GLUT4 expression. Additionally, loss- and gain-of-function analyses showed that overexpression of miR-17 impaired glucose metabolism in L6 rat skeletal muscle cell line. In contrast, knockdown of endogenous miR-17 ameliorated glucose metabolism, accompanied by elevation of GLUT4 protein level. These findings unraveled a novel mechanism for IR that involves repression of GLUT4 by miR-17 and suggested miR-17 as a potential molecular target for the development of new therapeutic approaches for the treatment of T2DM.


Assuntos
Diabetes Mellitus Tipo 2/genética , Transportador de Glucose Tipo 4/genética , Glucose/metabolismo , Resistência à Insulina/genética , MicroRNAs/metabolismo , Animais , Glicemia , Metabolismo dos Carboidratos/genética , Linhagem Celular , Diabetes Mellitus Tipo 2/sangue , Diabetes Mellitus Tipo 2/etiologia , Diabetes Mellitus Tipo 2/metabolismo , Dieta Hiperlipídica/efeitos adversos , Modelos Animais de Doenças , Regulação para Baixo , Técnicas de Silenciamento de Genes , Transportador de Glucose Tipo 4/sangue , Transportador de Glucose Tipo 4/metabolismo , Humanos , Masculino , MicroRNAs/genética , Músculo Esquelético/metabolismo , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar
16.
Cell Death Dis ; 9(2): 171, 2018 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-29416034

RESUMO

Cigarette smoking is a major risk factor for atherosclerosis and other cardiovascular diseases. Increasing evidence has demonstrated that nicotine impairs the cardiovascular system by targeting vascular endothelial cells, but the underlying mechanisms remain obscure. It is known that cell death and inflammation are crucial processes leading to atherosclerosis. We proposed that pyroptosis may be implicated in nicotine-induced atherosclerosis and therefore conducted the present study. We found that nicotine resulted in larger atherosclerotic plaques and secretion of inflammatory cytokines in ApoE-/- mice fed with a high-fat diet (HFD). Treatment of human aortic endothelial cells (HAECs) with nicotine resulted in NLRP3-ASC inflammasome activation and pyroptosis, as evidenced by cleavage of caspase-1, production of downstream interleukin (IL)-1ß and IL-18, and elevation of LDH activity and increase of propidium iodide (PI) positive cells, which were all inhibited by caspase-1 inhibitor. Moreover, silencing NLRP3 or ASC by small interfering RNA efficiently suppressed nicotine-induced caspase-1 cleavage, IL-18 and IL-1ß production, and pyroptosis in HAECs. Further experiments revealed that the nicotine-NLRP3-ASC-pyroptosis pathway was activated by reactive oxygen species (ROS), since ROS scavenger (N-acetyl-cysteine, NAC) prevented endothelial cell pyroptosis. We conclude that pyroptosis is likely a cellular mechanism for the pro-atherosclerotic property of nicotine and stimulation of ROS to activate NLRP3 inflammasome is a signaling mechanism for nicotine-induced pyroptosis.


Assuntos
Aterosclerose/patologia , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Nicotina/toxicidade , Piroptose/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Animais , Aorta/efeitos dos fármacos , Aorta/patologia , Apolipoproteínas E/deficiência , Apolipoproteínas E/metabolismo , Aterosclerose/metabolismo , Proteínas Adaptadoras de Sinalização CARD/metabolismo , Caspase 1/metabolismo , Inibidores de Caspase/farmacologia , Células Endoteliais/efeitos dos fármacos , Humanos , Inflamassomos/metabolismo , Interleucina-18/metabolismo , Interleucina-1beta/metabolismo , Camundongos , Modelos Biológicos
17.
Nat Commun ; 9(1): 4176, 2018 10 09.
Artigo em Inglês | MEDLINE | ID: mdl-30301979

RESUMO

Long non-coding RNAs (lncRNAs) have emerged as a new class of gene expression regulators playing key roles in many biological and pathophysiological processes. Here, we identify cardiac conduction regulatory RNA (CCRR) as an antiarrhythmic lncRNA. CCRR is downregulated in a mouse model of heart failure (HF) and in patients with HF, and this downregulation slows cardiac conduction and enhances arrhythmogenicity. Moreover, CCRR silencing induces arrhythmias in healthy mice. CCRR overexpression eliminates these detrimental alterations. HF or CCRR knockdown causes destruction of intercalated discs and gap junctions to slow longitudinal cardiac conduction. CCRR overexpression improves cardiac conduction by blocking endocytic trafficking of connexin43 (Cx43) to prevent its degradation via binding to Cx43-interacting protein CIP85, whereas CCRR silence does the opposite. We identified the functional domain of CCRR, which can reproduce the functional roles and pertinent molecular events of full-length CCRR. Our study suggests CCRR replacement a potential therapeutic approach for pathological arrhythmias.


Assuntos
Acoplamento Excitação-Contração/genética , Espaço Extracelular/metabolismo , Sistema de Condução Cardíaco/metabolismo , RNA Longo não Codificante/metabolismo , Animais , Arritmias Cardíacas/genética , Conexina 43/metabolismo , Junções Comunicantes/metabolismo , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Insuficiência Cardíaca/genética , Humanos , Masculino , Camundongos Endogâmicos C57BL , Modelos Biológicos , Miocárdio/metabolismo , Miocárdio/patologia , Miocárdio/ultraestrutura , RNA Longo não Codificante/genética , Transdução de Sinais , Frações Subcelulares/metabolismo , Proteínas rab de Ligação ao GTP/metabolismo
18.
Sci Rep ; 6: 19346, 2016 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-26783084

RESUMO

Non-small cell lung cancer (NSCLC), one of the most common causes of cancer-related death, is a worldwide public health problem. MicroRNAs (miRNAs) have recently been identified as a novel class of regulators of carcinogenesis and tumor progression, including miRNAs associated with NSCLC. This study aimed to explore the role of miR-522 in NSCLC and the mechanisms underlying this role. We report here that miR-522 expression was significantly increased in both human NSCLC tissues and cell lines. Furthermore, an MTT assay, 5-Ethynyl-2'-deoxyuridine (EdU) assay kit and flow cytometry confirmed that the inhibition of miR-522 suppressed NSCLC cells proliferation and induced apoptosis. Compared with miR-522 overexpression, miR-522 inhibitor markedly reduced cells migration and invasion, as indicated by wound-healing and transwell assays. In addition, a luciferase assay identified DENN/MADD domain containing 2D (DENND2D) as a direct target of miR-522. qRT-PCR and western blot analyses indicated the reciprocal expression of miR-522 and DENND2D in NSCLC tissue samples. DENND2D was involved in miR-522 induced proliferation and metastasis of NSCLC cells by a miRNA-masking antisense oligonucleotides (miR-mask) technology. These data highlight a novel molecular interaction between miR-522 and DENND2D, which indicates that targeting miR-522 may constitute a potential therapy for NSCLC.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/genética , Regulação Neoplásica da Expressão Gênica , Fatores de Troca do Nucleotídeo Guanina/genética , Neoplasias Pulmonares/genética , MicroRNAs/genética , Interferência de RNA , Proteínas Supressoras de Tumor/genética , Apoptose/genética , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Carcinoma Pulmonar de Células não Pequenas/patologia , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Metástase Neoplásica , Proteínas Supressoras de Tumor/metabolismo
19.
Toxicol Lett ; 250-251: 1-9, 2016 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-27025157

RESUMO

Bisphenol A (BPA) is a widely studied typical endocrine-disrupting chemical. The present study aimed to verify whether BPA could induce proliferation of cardiac fibroblasts and collagen production leading to cardiac interstitial fibrosis. After exposure to BPA for 30 consecutive days, decreased cardiac function was observed in rats using echocardiography, and the deposition of collagen was detected by Masson's trichrome staining and electron microscope. BPA remarkably stimulated proliferation and migration of cultured cardiac fibroblasts and collagen production in a concentration-dependent manner, as revealed by MTT, wound healing assay and collagen assay. Meanwhile, BPA treatment also enhanced phosphorylation of extracellular signal-regulated kinase 1/2 (ERK1/2). In contrast, pretreatment with estrogen receptor inhibitor ICI182780 or ERK inhibitor PD98059 prevented the enhanced phosphorylation of ERK1/2, and subsequently inhibited the up-regulation of transforming growth factor-ß1 (TGF-ß1) expression induced by BPA. As a consequence, these inhibitors also decreased proliferation and collagen production, as well as the fibrosis-related genes expression. Taken together, our results indicated that BPA may act as a promoting factor in proliferative process and collagen production of cardiac fibroblasts via activating ERK1/2.


Assuntos
Compostos Benzidrílicos/toxicidade , Cardiomiopatias/induzido quimicamente , Disruptores Endócrinos/toxicidade , Poluentes Ambientais/toxicidade , Fibroblastos/efeitos dos fármacos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Miocárdio/enzimologia , Fenóis/toxicidade , Transdução de Sinais/efeitos dos fármacos , Animais , Cardiomiopatias/enzimologia , Cardiomiopatias/patologia , Cardiomiopatias/fisiopatologia , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Colágeno/metabolismo , Relação Dose-Resposta a Droga , Ativação Enzimática , Antagonistas de Estrogênios/farmacologia , Fibroblastos/enzimologia , Fibroblastos/ultraestrutura , Fibrose , Proteína Quinase 1 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 3 Ativada por Mitógeno/antagonistas & inibidores , Miocárdio/ultraestrutura , Fosforilação , Inibidores de Proteínas Quinases/farmacologia , Ratos Sprague-Dawley , Fatores de Tempo , Fator de Crescimento Transformador beta1/metabolismo
20.
Sci Rep ; 6: 33787, 2016 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-27671604

RESUMO

Emerging evidence has suggested the critical role of endothelial to mesenchymal transition (EndMT) in fibrotic diseases. The present study was designed to examine whether EndMT is involved in arsenic trioxide (As2O3)-induced cardiac fibrosis and to explore the underlying mechanisms. Cardiac dysfunction was observed in rats after exposure to As2O3 for 15 days using echocardiography, and the deposition of collagen was detected by Masson's trichrome staining and electron microscope. EndMT was indicated by the loss of endothelial cell markers (VE-cadherin and CD31) and the acquisition of mesenchymal cell markers (α-SMA and FSP1) determined by RT-PCR at the mRNA level and Western blot and immunofluorescence analysis at the protein level. In the in-vitro experiments, endothelial cells acquired a spindle-shaped morphology accompanying downregulation of the endothelial cell markers and upregulation of the mesenchymal cell markers when exposed to As2O3. As2O3 activated the AKT/GSK-3ß/Snail signaling pathway, and blocking this pathway with PI3K inhibitor (LY294002) abolished EndMT in As2O3-treated endothelial cells. Our results highlight that As2O3 is an EndMT-promoting factor during cardiac fibrosis, suggesting that targeting EndMT is beneficial for preventing As2O3-induced cardiac toxicity.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA