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










Base de datos
Intervalo de año de publicación
1.
iScience ; 26(10): 107888, 2023 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-37766986

RESUMEN

This study found that the level of neuroepithelial cell-transforming gene 1 protein (NET1) was significantly increased in a mouse cardiac fibrosis model. Moreover, the expression level of NET1 was increased in cardiac fibrosis induced by TGF-ß1, suggesting that NET1 was involved in the pathological process of cardiac fibrosis. Overexpression of NET1 promoted ß-catenin expression in the nucleus and significantly increased the proliferation and migration of cardiac fibroblasts. NET1 may form a complex with ß-catenin through GSK3ß. Knockdown of ß-catenin alleviated the effects of NET1 overexpression on collagen production and cell migration. In the heart of NET1 knockout mice, NET1 knockout can reduce the expression of ß-catenin, α-SMA, and collagen content induced by MI. In conclusion, NET1 may regulate the activation of Wnt/ß-catenin and TGF/Smads signaling pathway, promote collagen synthesis in fibroblasts, and participate in cardiac fibrosis. Thus, NET1 may be a potential therapeutic target in cardiac fibrosis.

2.
Cell Death Dis ; 14(5): 344, 2023 05 30.
Artículo en Inglés | MEDLINE | ID: mdl-37253771

RESUMEN

Cardiac hypertrophy is a common structural remodeling in many cardiovascular diseases. Recently, long non-coding RNAs (LncRNAs) were found to be involved in the physiological and pathological processes of cardiac hypertrophy. In this study, we found that LncRNA KCND1 (LncKCND1) was downregulated in both transverse aortic constriction (TAC)-induced hypertrophic mouse hearts and Angiotensin II (Ang II)-induced neonatal mouse cardiomyocytes. Further analyses showed that the knockdown of LncKCND1 impaired cardiac mitochondrial function and led to hypertrophic changes in cardiomyocytes. In contrast, overexpression of LncKCND1 inhibited Ang II-induced cardiomyocyte hypertrophic changes. Importantly, enhanced expression of LncKCND1 protected the heart from TAC-induced pathological cardiac hypertrophy and improved heart function in TAC mice. Subsequent analyses involving mass spectrometry and RNA immunoprecipitation assays showed that LncKCND1 directly binds to YBX1. Furthermore, overexpression of LncKCND1 upregulated the expression level of YBX1, while silencing LncKCND1 had the opposite effect. Furthermore, YBX1 was downregulated during cardiac hypertrophy, whereas overexpression of YBX1 inhibited Ang II-induced cardiomyocyte hypertrophy. Moreover, silencing YBX1 reversed the effect of LncKCND1 on cardiomyocyte mitochondrial function and its protective role in cardiac hypertrophy, suggesting that YBX1 is a downstream target of LncKCND1 in regulating cardiac hypertrophy. In conclusion, our study provides mechanistic insights into the functioning of LncKCND1 and supports LncKCND1 as a potential therapeutic target for pathological cardiac hypertrophy.


Asunto(s)
ARN Largo no Codificante , Factores de Transcripción , Animales , Ratones , Angiotensina II/farmacología , Cardiomegalia/metabolismo , Ratones Endogámicos C57BL , Miocitos Cardíacos/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Canales de Potasio Shal/metabolismo , Factores de Transcripción/metabolismo
3.
Int J Biol Sci ; 18(16): 6008-6019, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36439874

RESUMEN

Cardiac fibrosis is one of the common pathological processes in many cardiovascular diseases characterized by excessive extracellular matrix deposition. SerpinE2 is a kind of protein that inhibits peptidase in extracellular matrix and up-regulated tremendously in mouse model of cardiac fibrosis induced by pressure-overloaded via transverse aortic constriction (TAC) surgery. However, its effect on cardiac fibroblasts (CFs), collagen secretion and the underlying mechanism remains unclear. In this study, DyLight® 488 green fluorescent dye or His-tagged proteins were used to label the exogenous serpinE2 protein. It was showed that extracellular serpinE2 translocated into CFs by low-density lipoprotein receptor-related protein 1 (LRP1) and urokinase plasminogen activator receptor (uPAR) of cell membrane through endocytosis. Knockdown of LRP1 or uPAR reduced the level of serpinE2 in CFs and down-regulated the collagen expression. Inhibition of the endocytosis of serpinE2 could inhibit ERK1/2 and ß-catenin signaling pathways and subsequently attenuated collagen secretion. Knockdown of serpinE2 attenuates cardiac fibrosis in TAC mouse. We conclude that serpinE2 could be translocated into cardiac fibroblasts due to endocytosis through directly interact with the membrane protein LRP1 and uPAR, and this process activated the ERK1/2, ß-catenin signaling pathways, consequently promoting collagen production.


Asunto(s)
beta Catenina , Ratones , Animales , beta Catenina/metabolismo , Serpina E2/metabolismo , Serpina E2/farmacología , Inhibidores de Proteasas/farmacología , Sistema de Señalización de MAP Quinasas/genética , Fibrosis , Transducción de Señal/genética , Endocitosis/genética , Colágeno/metabolismo
4.
Eur J Pharmacol ; 923: 174892, 2022 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-35358494

RESUMEN

Montelukast, cysteinyl leukotriene receptor 1 (CysLT1R) antagonist, is used clinically for patients with asthma, chronic obstructive pulmonary diseases (COPD), and allergic rhinitis. It has been reported that CysLT1R antagonists could reduce the risks of cardiovascular diseases in animal studies. Cardiac fibrosis is one of the major causes of heart failure. But little is known about the role of Montelukast in cardiac fibrosis and its underlying mechanism. In transverse aortic constriction (TAC) mice, Montelukast improved cardiac pumping function and inhibited cardiac fibrosis by down-regulation of the proteins related to the fibrosis, such as connective tissue growth factor (CTGF), Transforming Growth Factor ß (TGF-ß), and Alpha-smooth muscle actin (α-SMA). Montelukast reduced cell proliferation and collagen production in neonatal cardiac fibroblasts (CFs) with the pretreatment of 20% serum, while down-regulating the expression of TGF-ß, CTGF and α-SMA. Molecules docking methods estimated a high affinity of Montelukast to Apelin receptor (APJ) and an effective chemical structure for Montelukast binding APJ. In Chinese hamster ovary (CHO) cells with stable overexpressing APJ, Montelukast inhibited forskolin (1 µM)-mediated cyclic adenosine monophosphate (cAMP) production and extracellular signal-regulated kinase1/2 (ERK1/2) phosphorylation, while these effects were reversed by pertussis toxin (PTX) pretreatment. APJ silence disrupted the effects of Montelukast in CFs pretreatment by serum 20%. So we concluded that Montelukast inhibited cardiac fibrosis due presumably to the coupling to the APJ-mediated Gi signaling pathway, which may be a promising therapeutic target for cardiac fibrosis.


Asunto(s)
Acetatos , Animales , Células CHO , Cricetinae , Cricetulus , Ciclopropanos , Fibrosis , Humanos , Ratones , Quinolinas , Receptores de Leucotrienos , Sulfuros , Factor de Crecimiento Transformador beta
5.
J Cell Physiol ; 236(2): 1094-1104, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-32657438

RESUMEN

Sox4 participates in the progression of embryo development and regulation of apoptosis in tumors. However, the effect and mechanism of Sox4 in myocardial infarction (MI) remains unclear. Therefore, we aimed at examining the role and molecular mechanism of Sox4 in the process of cardiomyocytes apoptosis during MI. The expression of Sox4 were obviously increased both in MI mice and in neonatal mouse cardiomyocytes treated with H2 O2 . Overexpression of Sox4 promoted cardiomyocyte apoptosis with or without H2 O2 , whereas knocking down of Sox4 alleviated H2 O2 -induced apoptosis in cardiomyocytes. Furthermore, silencing Sox4 by AAV-9 carried short hairpin RNA targeting Sox4 (AAV-9-sh-Sox4) markedly decreased cardiac infarct area, imprfoved cardiac dysfunction, and reversed apoptosis in MI mice. Mechanistically, there is a potential Sox4-binding site in the promoter region of Bim, and forced expression of Sox4 significantly promoted Bim expression in cultured cardiomyocytes with or without H2 O2 , whereas knocking down of Sox4 inhibited the expression of Bim. Further studies showed that silencing Bim attenuated Sox4-induced apoptosis in cardiomyocytes, indicating that Sox4 promoted cardiomyocytes apoptosis through regulation of Bim expression, which can be used as a potential therapeutic target for MI.


Asunto(s)
Proteína 11 Similar a Bcl2/genética , Infarto del Miocardio/genética , Isquemia Miocárdica/genética , Factores de Transcripción SOXC/genética , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Regulación de la Expresión Génica/genética , Humanos , Peróxido de Hidrógeno/farmacología , Ratones , Infarto del Miocardio/patología , Isquemia Miocárdica/patología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Regiones Promotoras Genéticas/genética
6.
Mol Ther Nucleic Acids ; 20: 841-850, 2020 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-32464547

RESUMEN

Acute myocardial infarction (AMI) is the leading cause of death worldwide. Identifying the pathways that block cardiac cell death is a therapeutic strategy for ischemic heart disease. We found that long noncoding RNA (lncRNA) myocardial infarction-regulatory factor (MIRF) promoted ischemic myocardial injury by regulating autophagy through targeting miR-26a. However, the role of MIRF-miR-26a in apoptosis during AMI has not been delineated. In this study, we found the downregulation of miR-26a both in the heart of myocardial infarction (MI) mice and in H2O2-treated cardiomyocytes. miR-26a silencing resulted in apoptosis, whereas overexpression of miR-26a attenuated H2O2-induced apoptosis through promoting mitochondrial ATP content and increasing mitochondrial membrane potential (MMP). Moreover, forced expression of miR-26a protected against MI-induced cardiac injury and attenuated cardiac apoptosis. Further studies showed that miR-26a inhibited apoptosis through regulation of Bak1. Furthermore, MIRF decreased ATP content and MMP through regulating miR-26a, which then promoted the cardiomyocyte apoptosis. In contrast, deficiency of MIRF promoted mitochondrial ATP content and increased MMP, and then inhibited MI or H2O2-induced cardiac apoptosis, which was abolished by miR-26a inhibitor. Taken together, these results suggested that MIRF contributed to cardiomyocyte apoptosis through modulating Bak1 by regulation of miR-26a, which can be a potential therapeutic target for the treatment of ischemic heart disease.

7.
Eur J Pharmacol ; 881: 173131, 2020 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-32450177

RESUMEN

Cardiac conduction delay may occur as a common complication of several cardiac diseases. A few therapies and drugs have a good effect on cardiac conduction delay. Metformin (Met) has a protective effect on the heart. This study's aim was to investigate whether Met could ameliorate cardiac conduction delay and its potential mechanism. Cardiac-specific microRNA-1 (miR-1) transgenic (TG) and myocardial infarction (MI) mouse models were used. Mice were administered with Met in an intragastric manner. We found that the expression of miR-1 was significantly up-regulated in H2O2 treated cardiomyocytes as well as in TG and MI mice. The protein levels of inwardly rectifying potassium channel 2.1 (Kir2.1) and Connexin43 (CX43) were down-regulated both in cardiomyocytes treated with H2O2 as well as cardiac tissues of TG and MI mice, as compared to their controls. Furthermore, the PR and QT intervals were prolonged, action potential duration (APD) was delayed, and conduction velocity (CV) was reduced, with upregulation of miR-1 in the hearts. In the meanwhile, intercalated disc injuries were found in the hearts of MI mice. Interestingly, Met can noticeably inhibit miR-1 upregulation and attenuate the changes mentioned above. Taken together, this suggested that Met could play an important role in improving cardiac conduction delay through inhibition of miR-1 expression. Our study proposes that Met is a potential candidate for the treatment of cardiac conduction delay and provides a new idea of treating arrhythmia with a drug.


Asunto(s)
Antiarrítmicos/farmacología , Trastorno del Sistema de Conducción Cardíaco/prevención & control , Sistema de Conducción Cardíaco/efectos de los fármacos , Frecuencia Cardíaca/efectos de los fármacos , Metformina/farmacología , MicroARNs/metabolismo , Infarto del Miocardio/tratamiento farmacológico , Miocitos Cardíacos/efectos de los fármacos , Potenciales de Acción/efectos de los fármacos , Animales , Trastorno del Sistema de Conducción Cardíaco/genética , Trastorno del Sistema de Conducción Cardíaco/metabolismo , Trastorno del Sistema de Conducción Cardíaco/fisiopatología , Conexina 43/metabolismo , Modelos Animales de Enfermedad , Regulación hacia Abajo , Sistema de Conducción Cardíaco/metabolismo , Sistema de Conducción Cardíaco/fisiopatología , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , MicroARNs/genética , Infarto del Miocardio/genética , Infarto del Miocardio/metabolismo , Infarto del Miocardio/fisiopatología , Miocitos Cardíacos/metabolismo , Canales de Potasio de Rectificación Interna/metabolismo , Transducción de Señal
8.
Autophagy ; 16(6): 1077-1091, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-31512556

RESUMEN

More evidence is emerging of the roles long non-coding RNAs (lncRNAs) play as regulatory factors in a variety of biological processes, but the mechanisms underlying the function of lncRNAs in acute myocardial infarction (AMI) have not been explicitly delineated. The present study identified the lncRNA 2810403D21Rik/AK007586/Mirf (myocardial infarction-regulatory factor), that inhibited macroautophagy/autophagy by modulating Mir26a (microRNA 26a). Inhibition of Mir26a led to cardiac injury both in vitro and in vivo, whereas overexpression of Mir26a attenuated ischemic stress-induced cell death by activating autophagy through targeting Usp15 (ubiquitin specific peptidase 15). More importantly, 2810403D21Rik/Mirf acted as a competitive endogenous RNA (ceRNA) of Mir26a; forced expression of 2810403D21Rik/Mirf downregulated Mir26a to inhibit autophagy. In contrast, loss of 2810403D21Rik/Mirf resulted in upregulation of Mir26a to promote autophagy and alleviate cardiac injury, which in turn improved cardiac function in MI mice. This study identified a lncRNA 2810403D21Rik/Mirf that functions as an anti-autophagic molecule via ceRNA activity toward Mir26a. Our findings suggest that knockdown of 2810403D21Rik/Mirf might be a novel therapeutic approach for cardiac diseases associated with autophagy. ABBREVIATIONS: 3-MA: 3-methyladenine; AAV-9: adenovirus associated virus-9; agoMir26a: cholesterol-conjugated Mir26a mimic; AMI: acute myocardial infarction; AMO-26a: Mir26a inhibitor; ATG: autophagy related; BECN1: beclin 1; ceRNA: competitive endogenous RNAs; EF: ejection fraction; f-2810403D21Rik/Mirf: fragment encompassing the Mir26a binding site; FS: fraction shortening; GFP-mRFP: a plasmid expressing green fluorescent protein-monomeric red fluorescent protein; lncRNA: long non-coding RNA; MAP1LC3/LC3: microtubule-associated protein 1 light chain 3; Mirf: myocardial infarction-regulatory factor; miRNAs: microRNAs; NC: negative control; NMCMs: neonatal mice cardiomyocytes; shRNA: short hairpin RNA; siRNA: small interfering RNA; SQSTM1/p62: sequestosome 1; TEM: transmission electron microscopy; Usp15: ubiquitin specific peptidase 15.


Asunto(s)
Autofagosomas/metabolismo , Autofagia/genética , MicroARNs/metabolismo , Infarto del Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , ARN Largo no Codificante/metabolismo , Animales , Autofagosomas/efectos de los fármacos , Autofagosomas/genética , Autofagosomas/ultraestructura , Autofagia/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Silenciador del Gen , Células HEK293 , Humanos , Peróxido de Hidrógeno/toxicidad , Ratones , Ratones Endogámicos C57BL , MicroARNs/genética , Microscopía Electrónica de Transmisión , Mutación , Infarto del Miocardio/genética , Infarto del Miocardio/patología , Miocitos Cardíacos/efectos de los fármacos , ARN Largo no Codificante/genética , Proteasas Ubiquitina-Específicas/genética , Proteasas Ubiquitina-Específicas/metabolismo , Regulación hacia Arriba
9.
Cell Death Dis ; 9(5): 464, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29700328

RESUMEN

Yes-associated protein 1 (YAP1) contributes to the development of multiple tumors, but the mechanism underlying YAP1 deregulation in non-small cell lung cancer (NSCLC) remains unclear. By performing immunohistochemistry (IHC) assays, we found that YAP1 was significantly upregulated in NSCLC compared with adjacent tissues; therefore, we sought to elucidate whether the upregulation of YAP1 contributes to NSCLC progression. MTT and transwell assays showed that YAP1 overexpression promoted proliferation, migration, and invasion in the NSCLC cell lines A549 and H460; YAP1 overexpression also promoted the significant differential expression of epithelial-mesenchymal transition (EMT)-related markers. Nevertheless, YAP1 knockdown alleviated TGF-ß1-induced EMT and proliferation, migration, and invasion in NSCLC. Furthermore, western blotting showed that the co-transcription complex YAP1/TEAD was impaired by YAPS94A (a YAP1 mutant without the TEAD binding site), and verteporfin (a small molecular inhibitor of YAP1) inhibited A549 and H460 cell metastasis and EMT-related markers expression, indicating that TEAD mediated the NSCLC aggressiveness induced by YAP1. Moreover, sequence analysis and ChIP and luciferase assays confirmed that YAP1 transcriptionally activated Slug expression by binding to TEAD. Importantly, silencing YAP1 inhibited A549 cell tumorigenesis and EMT and downregulated Slug expression in vivo. Overall, our findings revealed that YAP1 is a driver of NSCLC metastasis because YAP1 promoted the EMT program by inducing Slug transcription.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Movimiento Celular , Neoplasias Pulmonares/metabolismo , Proteínas de Neoplasias/metabolismo , Fosfoproteínas/metabolismo , Factores de Transcripción de la Familia Snail/metabolismo , Transcripción Genética , Células A549 , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/patología , Femenino , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Invasividad Neoplásica , Proteínas de Neoplasias/genética , Fosfoproteínas/genética , Factores de Transcripción de la Familia Snail/genética , Factores de Transcripción , Proteínas Señalizadoras YAP
10.
Mol Ther Nucleic Acids ; 10: 387-397, 2018 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-29499950

RESUMEN

Cardiac hypertrophy accompanied by maladaptive cardiac remodeling is the uppermost risk factor for the development of heart failure. Long non-coding RNAs (lncRNAs) and microRNAs (miRNAs) have various biological functions, and their vital role in the regulation of cardiac hypertrophy still needs to be explored. In this study, we demonstrated that lncRNA Plscr4 was upregulated in hypertrophic mice hearts and in angiotensin II (Ang II)-treated cardiomyocytes. Next, we observed that overexpression of Plscr4 attenuated Ang II-induced cardiomyocyte hypertrophy. Conversely, the inhibition of Plscr4 gave rise to cardiomyocyte hypertrophy. Furthermore, overexpression of Plscr4 attenuated TAC (transverse aortic constriction)-induced cardiac hypertrophy. Finally, we demonstrated that Plscr4 acted as an endogenous sponge of miR-214 and forced expression of Plscr4 downregulated miR-214 expression to promote Mfn2 and attenuate hypertrophy. In contrast, knockdown of Plscr4 upregulated miR-214 to induce cardiomyocyte hypertrophy. Additionally, luciferase assay showed that miR-214 was the direct target of Plscr4, and overexpression of miR-214 counteracted the anti-hypertrophy effect of Plscr4. Collectively, these findings identify Plscr4 as a negative regulator of cardiac hypertrophy in vivo and in vitro due to its regulation of the miR-214-Mfn2 axis, suggesting that Plscr4 might act as a therapeutic target for the treatment of cardiac hypertrophy and heart failure.

11.
Cell Physiol Biochem ; 39(4): 1561-7, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27614433

RESUMEN

BACKGROUND/AIMS: Liver X receptor (LXR), a member of the nuclear receptor superfamily, is known to induce the expression of SREBP-1c and ChREBP, two master regulators of hepatic lipogenesis. Histone deacyetylases (HDACs) have been shown to play critical roles in glucose and lipids metabolism. However, the exact role of HDAC5 in lipogenesis remains elusive. METHODS: mRNA and protein levels of HDAC5 were analyzed by quantitative real-time PCR and Western blots in high-fat-diet-induced and leptin receptor deficiency-induced obese mice. HDAC5 was overexpressed or depleted in HepG2 cells, followed by analysis of cellular triglycerides contents. Quantitative real-time PCR was used to detect the expression levels of lipogenic genes. Luciferase reporter assay was used to determine the regulation of HDAC on the transcriptional activity of LXR. Co-immunoprecipitation experiment was used to determine the interaction between HDAC5 and LXR. RESULTS: We found that mRNA and protein expression levels of hepatic HDAC5 were reduced in high-fat-diet-induced and leptin receptor deficiency-induced obese mice. In vitro studies further demonstrated that knockdown of HDAC5 promoted cellular triglycerides accumulation, accompanied with up-regulation of lipogenic genes. At the molecular level, HDAC5 was shown to interact with LXR, thereby attenuating its transcriptional activity. CONCLUSION: Overall, our data suggest that hepatic HDAC5 is an important regulator of lipogenesis.


Asunto(s)
Histona Desacetilasas/genética , Lipogénesis/genética , Receptores X del Hígado/genética , Hígado/metabolismo , Obesidad/genética , Transcripción Genética , Animales , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice , Dieta Alta en Grasa , Regulación de la Expresión Génica , Genes Reporteros , Glucosa/metabolismo , Células HEK293 , Células Hep G2 , Histona Desacetilasas/metabolismo , Humanos , Hígado/patología , Receptores X del Hígado/metabolismo , Luciferasas/genética , Luciferasas/metabolismo , Ratones , Ratones Endogámicos C57BL , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Obesidad/etiología , Obesidad/metabolismo , Obesidad/patología , Receptores de Leptina/deficiencia , Receptores de Leptina/genética , Transducción de Señal , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Triglicéridos/metabolismo
12.
J Cell Mol Med ; 20(3): 495-505, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26756969

RESUMEN

Cardiac fibrosis is a major cause of heart failure. MicroRNAs (miRs) are important epigenetic regulators of cardiac function and cardiovascular diseases, including cardiac fibrosis. This study aimed to explore the role of miR-503 and its mechanisms in regulating cardiac fibrosis. miR-503 was found up-regulated in the mouse LV tissues subjected to transverse aortic constriction (TAC) and in neonatal cardiac fibroblasts (CFs) cultured with Angiotension II. The role of miR-503 in regulating CF cell proliferation and/or collagen production in mice neonatal CFs were determined using an MTT assay and RT-PCR respectively. Forced expression of miR-503 increased the cellular proliferation and collagen production in mice neonatal CFs. The effects were abrogated by cotransfection with AMO-503 (a specific inhibitor of miR-503). Injection of antagomiR-503 elevated cardiac function and inhibited the expression of connective tissue growth factor (CTGF) and transforming growth factor (TGF)-ß in the TAC mice. Additional analysis revealed that Apelin-13 is a direct target of miR-503, as the overexpression of miR-503 decreased the protein and mRNA expression levels of Apelin-13. In the CFs with pre-treatment of AngII, we transfected AMO-503 into the cells treated with siRNA-APLN. siRNA-APLN abolished the effects of AMO-503 on the production of collagen I and III and the expression of TGF-ß and CTGF. Furthermore, pre-treatment of CFs with Apelin-13 (1-100 nmol/l) inhibited angiotensin II-mediated collagen production and activation of CTGF and TGF-ß. So we conclude that miR-503 promotes cardiac fibrosis via miR-503-Apelin-13-TGF-ß-CTGF-collagen production pathway. Thus, miR-503 is a promising therapeutic target for reducing cardiac fibrosis.


Asunto(s)
Adipoquinas/genética , Cardiopatías/metabolismo , Péptidos y Proteínas de Señalización Intercelular/genética , MicroARNs/fisiología , Regiones no Traducidas 3' , Adipoquinas/metabolismo , Angiotensina II , Animales , Apelina , Secuencia de Bases , Sitios de Unión , Proliferación Celular , Células Cultivadas , Fibroblastos/fisiología , Fibrosis , Expresión Génica , Cardiopatías/inducido químicamente , Cardiopatías/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Masculino , Ratones Endogámicos C57BL , Miocardio/metabolismo , Miocardio/patología , Interferencia de ARN
13.
Front Med ; 9(4): 444-56, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26626190

RESUMEN

Ventricular hypertrophy is a powerful and independent predictor of cardiovascular morbid events. The vascular properties of low-dose acetyl salicylic acid (aspirin) provide cardiovascular benefits through the irreversible inhibition of platelet cyclooxygenase 1; however, the possible anti-hypertrophic properties and potential mechanism of aspirin have not been investigated in detail. In this study, healthy wild-type male mice were randomly divided into three groups and subjected to transverse aortic constriction (TAC) or sham operation. The TAC-operated mice were treated with the human equivalent of low-dose aspirin (10 mg·kg(-1)·d(-1)); the remaining mice received an equal amount of phosphate buffered saline with 0.65% ethanol, which was used as a vehicle. A cardiomyocyte hypertrophy model induced by angiotensin II (10 nmol·L(-1)) was treated with the human equivalent of low (10 or 100 µmol·L(-1)) and high (1000 µmol·L(-1)) aspirin concentrations in plasma. Changes in the cardiac structure and function were assessed through echocardiography and transmission electron microscopy. Gene expression was determined through RT-PCR and western blot analysis. Results indicated that aspirin treatment abrogated the increased thickness of the left ventricular anterior and posterior walls, the swelling of mitochondria, and the increased surface area in in vivo and in vitro hypertrophy models. Aspirin also normalized the upregulated hypertrophic biomarkers, ß-myosin heavy chain (ß-MHC), atrial natriuretic peptide (ANP), and b-type natriuretic peptide (BNP). Aspirin efficiently reversed the upregulation of ß-catenin and P-Akt expression and the TAC- or ANG II-induced downregulation of GSK-3ß. Therefore, low-dose aspirin possesses significant anti-hypertrophic properties at clinically relevant concentrations for anti-thrombotic therapy. The downregulation of ß-catenin and Akt may be the underlying signaling mechanism of the effects of aspirin.


Asunto(s)
Aspirina/farmacología , Hipertrofia Ventricular Izquierda , Mitocondrias Cardíacas , Miocitos Cardíacos , beta Catenina/metabolismo , Animales , Células Cultivadas , Inhibidores de la Ciclooxigenasa/farmacología , Modelos Animales de Enfermedad , Monitoreo de Drogas , Ecocardiografía/métodos , Hipertrofia Ventricular Izquierda/diagnóstico , Hipertrofia Ventricular Izquierda/etiología , Hipertrofia Ventricular Izquierda/metabolismo , Hipertrofia Ventricular Izquierda/prevención & control , Ratones , Microscopía Electrónica de Transmisión/métodos , Mitocondrias Cardíacas/efectos de los fármacos , Mitocondrias Cardíacas/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Proteína Oncogénica v-akt/metabolismo , Ratas , Transducción de Señal/efectos de los fármacos , Proteínas Wnt/metabolismo
14.
Exp Ther Med ; 9(6): 2325-2330, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26136981

RESUMEN

This study aimed to investigate whether single nucleotide polymorphisms (SNPs) located near the gene of the ABO blood group play an important role in the genetic aetiology of menstrual disorders (MDs). Polymerase chain reaction-ligase detection reaction technology was used to detect eight SNPs near the ABO gene location on the chromosomes in 250 cases of MD and 250 cases of normal menstruation. The differences in the distribution of each genotype, as well as the allele frequency in the normal and control groups, were analysed using Pearson's χ2 test to search for disease-associated loci. SHEsis software was used to analyse the linkage disequilibrium and haplotype frequencies and to inspect the correlation between haplotypes and the disease. Compared with the control group, the experimental group exhibited statistically significant differences in the genotype distribution frequencies of the rs657152 locus of the ABO blood group gene and the rs17250673 locus of the tumour necrosis factor cofactor 2 (TRAF2) gene, which is located downstream of the ABO gene. The allele distribution frequencies of rs657152 and rs495828 loci in the ABO blood group gene exhibited significant differences between the groups. Dominant and recessive genetic model analysis of each locus revealed that the experimental group exhibited statistically significant differences from the control group in the genotype distribution frequencies of rs657152 and rs495828 loci, respectively. These results indicate that the ABO blood group gene and TRAF2 gene may be a cause of MDs.

15.
Diabetes Technol Ther ; 17(7): 445-8, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25826625

RESUMEN

OBJECTIVE: The aim of this preliminary study is to investigate contributions of basal glucose (BG) and postprandial glucose (PPG) increments to overall hyperglycemia in newly diagnosed patients with type 2 diabetes mellitus (T2DM). RESEARCH DESIGN AND METHODS: We evaluated the relative contributions of BG and PPG to overall hyperglycemia in 59 newly diagnosed T2DM patients according to BG baseline value of 6.1 mmol/L and 24-h glucose profiles of normal glucose tolerance (NGT) subjects obtained by continuous glucose monitoring as baseline, respectively. RESULTS: When the baseline was 24-h glucose profiles of the NGT subjects, the relative contributions of PPG in the T2DM patients with hemoglobin A1c (HbA1c) levels of ≤ 7.0%, 7.0-9.0%, and >9.0% were 57.58%, 44.69%, and 21.56%, respectively. When the baseline value was equal to 6.1 mmol/L, the relative contributions of PPG in the T2DM patients with HbA1c levels of ≤ 7.0%, 7.0-9.0%, and >9.0% were 77.23%, 53.43%, and 22.78%, respectively. Compared with the 24-h glucose profiles of the NGT subjects as the baseline, the relative contribution of PPG was overestimated by about 10-20% in the T2DM patients with HbA1c levels of ≤ 9.0% when 6.1 mmol/L was chosen as the baseline. CONCLUSIONS: In the newly diagnosed T2DM patients with mild hyperglycemia, PPG is a predominant contributor, whereas the relative contributions of BG gradually increase from mild to severe hyperglycemia and obviously exceed PPG in the T2DM patients with HbA1c levels of >9.0%. This finding implies that the initial pharmacotherapy may target PPG in those patients with mild hyperglycemia and target BG in those patients with severe hyperglycemia.


Asunto(s)
Glucemia/metabolismo , Diabetes Mellitus Tipo 2/sangre , Hemoglobina Glucada/metabolismo , Hiperglucemia/metabolismo , Periodo Posprandial , Adulto , Anciano , Automonitorización de la Glucosa Sanguínea , Femenino , Prueba de Tolerancia a la Glucosa , Hemoglobina Glucada/análisis , Humanos , Hiperglucemia/sangre , Masculino , Persona de Mediana Edad , Adulto Joven
16.
Exp Ther Med ; 7(6): 1752-1756, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24926379

RESUMEN

The aim of this study was to assess the efficacy of a combination therapy of vildagliptin plus an α-glucosidase inhibitor for patients with type II diabetes mellitus. Type II diabetic patients exhibiting poor glycemic control following α-glucosidase inhibitor treatment for at least two months were selected and randomly distributed into vildagliptin and placebo groups. The body weight, fasting blood glucose (FBG), postprandial glucose (PPG), glycated hemoglobin (HBA1c) and blood lipid levels and hepatorenal functions of the patients were determined before and 12 weeks after the trial. Following the trial, the FBG, PPG, HbA1c, cholesterol (CHOL) and triglyceride (TG) levels in the vildagliptin group were significantly decreased compared with the pretreatment levels (P<0.05), whereas only the PPG level in the placebo group decreased (P<0.05). The FBG, PPG and HbA1c levels in the vildagliptin group were markedly lower than those in the placebo group 12 weeks after the trial. A comparison of the body weights and hepatorenal functions before and after the trial or between groups did not show statistically significant differences. The combination therapy of vildagliptin plus an α-glucosidase inhibitor effectively reduced the FBG, PPG and HbA1c levels in patients without inducing weight gain or hepatorenal dysfunction. However, the therapy may have caused a reduction in the blood lipid levels.

17.
Phytomedicine ; 21(7): 931-5, 2014 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-24680622

RESUMEN

Matrine, a alkaloid of the root of Sophora flavescens, has multiple protective effects on the cardiovascular system including cardiac arrhythmias. However, the molecular and ionic mechanisms of matrine have not been well investigated. Our study aimed at to shed a light on the issue to investigate the antiarrhythmic effects of matrine by using ouabain to construct an arrhythmic model of cardiomyocytes. In this experiment, matrine significantly and dose-dependently increased the doses of ouabain required to induce cardiac arrhythmias and decreased the duration of arrhythmias in guinea pigs. In cardiomyocytes of guinea pigs, ouabain 10 µM prolonged action potential duration by 80% (p<0.05) and increased L-type Ca(2+) currents and Ca(2+) transients induced by KCl (p<0.05). Matrine 100 µM shortened the prolongation of APD and prevented the increase of L-type Ca(2+) currents and Ca(2+) transients induced by ouabain. Taken together, these findings provide the first evidence that matrine possessed arrhythmogenic effect of ouabain by inhibiting of L-type Ca(2+) currents and Ca(2+) overload in guinea pigs.


Asunto(s)
Alcaloides/farmacología , Antiarrítmicos/farmacología , Ouabaína/farmacología , Quinolizinas/farmacología , Sophora/química , Potenciales de Acción/efectos de los fármacos , Animales , Arritmias Cardíacas/inducido químicamente , Arritmias Cardíacas/tratamiento farmacológico , Calcio/metabolismo , Canales de Calcio Tipo L/metabolismo , Células Cultivadas , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Cobayas , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Matrinas
18.
Exp Ther Med ; 7(4): 799-803, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24669235

RESUMEN

The aim of the present study was to assess the efficacy and safety of vildagliptin plus metformin combination therapy in patients with type II diabetes mellitus. Type II diabetic patients with poor glycemic control following at least three months of metformin treatment were selected and randomized into two groups. Vildagliptin or placebo was administered with metformin. Body weight, fasting blood glucose (FBG), postprandial glucose (PPG), glycated hemoglobin (HbA1c), blood lipid and hepatorenal function levels were analyzed in the patients prior to and 24-weeks after the trial. FBG, PPG and HbA1c levels of the patients in the vildagliptin group significantly decreased following the trial, whereas no statistically significant differences were observed in the various indicators of the placebo group prior to and following the trial. The FBG, PPG and HbA1c levels in the vildagliptin group were significantly lower compared with the placebo group 24-weeks after the trial. Comparisons of body weight, blood lipid and hepatorenal function between the groups prior to and following the trial exhibited no statistically significant differences. Therefore, vildagliptin plus metformin combination therapy effectively reduced FBG, PPG and HbA1c levels in patients with no risk of weight gain or hepatorenal dysfunction.

19.
Zhong Yao Cai ; 36(9): 1425-7, 2013 Sep.
Artículo en Chino | MEDLINE | ID: mdl-24620684

RESUMEN

OBJECTIVE: To provide the identification basis for Mongolian medicine Potentilla glabra. METHODS: Macroscopic and microscopic identification were applied to observe macroscopic, histological, superficial, and powder characteristics of its stems and leaves. RESULTS: The following characteristics were observed: The cork layer sandwiched with sclerenchyma ring which was main composed of fibers, accompanied by stone cells and large cell layer; Catheter with fiber existed in xylem; Beaded thickened anticlinal wall in leaf epidermal cell; Stomatal infinitive; Small clusters of calcium oxalate crystal and different type of leaf transverse section. CONCLUSION: These characteristics can provide evidences for the identification and quality control of Potentilla glabra.


Asunto(s)
Medicina Tradicional Mongoliana , Tallos de la Planta/anatomía & histología , Plantas Medicinales/anatomía & histología , Potentilla/anatomía & histología , Microscopía , Hojas de la Planta/anatomía & histología , Hojas de la Planta/citología , Hojas de la Planta/ultraestructura , Tallos de la Planta/citología , Tallos de la Planta/ultraestructura , Plantas Medicinales/citología , Plantas Medicinales/ultraestructura , Potentilla/citología , Potentilla/ultraestructura , Polvos , Control de Calidad
20.
Clin Endocrinol (Oxf) ; 76(6): 810-5, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21854404

RESUMEN

OBJECTIVE: Glycemic variability is poorly studied in the nondiabetic individuals and newly diagnosed patients with type 2 diabetes. The aim of the study is to investigate the characteristics of glucose fluctuations in subjects with normal glucose tolerance (NGT), impaired glucose regulation (IGR) and newly diagnosed, drug-naïve type 2 diabetes mellitus (DM-2). DESIGN AND PATIENTS: This is a cross-sectional study of three groups including 53 subjects with IGR, 56 DM-2 patients and 53 NGT individuals. Monitoring by a continuous glucose monitoring system (CGMS(®) System Gold(™)) was performed for three consecutive days. MEASUREMENTS: Mean blood glucose (MBG), standard deviation of MBG (SDBG), largest amplitude of glycemic excursions (LAGE) and mean amplitude of glycemic excursions (MAGE) were calculated to estimate intraday blood glucose variability. Interday variability of glucose was evaluated by absolute means of daily differences (MODD). Postprandial glucose excursion (PPGE) was calculated to assess the influence of meals on glucose fluctuation. RESULTS: Twenty-two percentage of NGT and 33.9% of IGR individuals experienced blood glucose ≥ 11.1 mmol/l; 49.1% of NGT, 50.9% of IGR and 30.8% of DM-2 participants had hypoglycemic episodes (CGM values <3.9 mmol/l). The IGR and DM-2 groups had greater SDBG (P = 0.010 and P < 0.001), LAGE (P = 0.014 and P < 0.001) and MAGE (P = 0.044 and P < 0.001) compared with the NGT group. Significantly greater MODD and PPGEs were found in the DM-2 groups than in the IGR and NGT groups (P < 0.001). The DM-2 patients had higher 72-MBG and glucose levels overnight than the NGT and IGR subjects (P < 0.001). In the patients with diabetes, MAGE was positively associated with MODD (r = 0.558, P < 0.001) and PPGEs (r = 0.738-0.843, P < 0.001). CONCLUSIONS: Glucose variability is present to an increasing degree from NGT to IGR and IGR to DM-2. Compared with the NGT individuals, the IGR and DM-2 subjects show more predominant intraday glucose fluctuations. The DM-2 patients demonstrate increased PPGEs, higher glucose levels overnight and greater interday fluctuations.


Asunto(s)
Glucemia/metabolismo , Diabetes Mellitus Tipo 2/sangre , Intolerancia a la Glucosa/sangre , Adulto , Glucemia/análisis , Estudios Transversales , Femenino , Prueba de Tolerancia a la Glucosa , Humanos , Masculino , Persona de Mediana Edad , Periodo Posprandial/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...