Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
In Vitro Cell Dev Biol Anim ; 60(4): 333-342, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38438604

RESUMEN

Hypoxia-induced cardiomyocyte apoptosis is one major pathological change of acute myocardial infarction (AMI), but the underlying mechanism remains unexplored. CDC-like kinase 3 (CLK3) plays crucial roles in cell proliferation, migration and invasion, and nucleotide metabolism, however, the role of CLK3 in AMI, especially hypoxia-induced apoptosis, is largely unknown. The expression of CLK3 was elevated in mouse myocardial infarction (MI) models and neonatal rat ventricular myocytes (NRVMs) under hypoxia. Furthermore, CLK3 knockdown significantly promoted apoptosis and inhibited NRVM survival, while CLK3 overexpression promoted NRVM survival and inhibited apoptosis under hypoxic conditions. Mechanistically, CLK3 regulated the phosphorylation status of AKT, a key player in the regulation of apoptosis. Furthermore, overexpression of AKT rescued hypoxia-induced apoptosis in NRVMs caused by CLK3 deficiency. Taken together, CLK3 deficiency promotes hypoxia-induced cardiomyocyte apoptosis through AKT signaling pathway.


Asunto(s)
Apoptosis , Hipoxia de la Célula , Miocitos Cardíacos , Proteínas Serina-Treonina Quinasas , Proteínas Proto-Oncogénicas c-akt , Transducción de Señal , Animales , Masculino , Ratones , Ratas , Ratones Endogámicos C57BL , Infarto del Miocardio/patología , Infarto del Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Fosforilación , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/deficiencia , Proteínas Tirosina Quinasas/metabolismo , Proteínas Tirosina Quinasas/genética , Proteínas Tirosina Quinasas/deficiencia , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas Sprague-Dawley
2.
BMC Cardiovasc Disord ; 22(1): 417, 2022 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-36123615

RESUMEN

BACKGROUND: As a near-physiological pacing innovation, left bundle branch area pacing (LBBAP) has drawn much attention recently. This study was aimed to investigate the electrophysiological characteristics, unipolar/bipolar pacing parameters and mid- to long-term effects and safety of three different pacing methods and identify possible predictors of adverse left ventricular remodeling. METHODS: Ninety-two patients were divided into the LBBAP group, right ventricular septal pacing (RVSP) group and right ventricular apical pacing (RVAP) group. Baseline information, electrophysiological, pacing and echocardiographic parameters were collected. RESULTS: The three pacing methods were performed with a similar high success rate. The paced QRSd was significantly different among the LBBAP, RVSP and RVAP groups (105.93 ± 15.85 ms vs. 143.63 ± 14.71 ms vs. 155.39 ± 14.17 ms, p < 0.01). The stimulus to left ventricular activation time (Sti-LVAT) was the shortest in the LBBAP group, followed by the RVSP and RVAP groups (72.80 ± 12.07 ms vs. 86.29 ± 8.71 ms vs. 94.14 ± 10.14 ms, p < 0.001). LBBAP had a significantly lower tip impedance during the procedure and 3-month follow up as compared to RVSP and RVAP (p < 0.001). Higher bipolar captured thresholds were observed in LBBAP during the procedure (p < 0.001). Compared to the baseline values, there was a greater reduction in left ventricular end-diastolic dimension (LVEDD) in the LBBAP group (p = 0.046) and a significant enlargement in LVEDD in the RVAP group (p = 0.008). Multiple regression analysis revealed that the Sti-LVAT was a significant predictor of LVEDD at 12 months post-procedure. At the 24-h post-procedure, significant elevations were observed in the cTnI levels in LBBAP (p < 0.001) and RVSP (p < 0.05). More transient RBB injury was observed in LBBAP. But no significant difference was found in cardiac composite endpoints among three groups (p > 0.05). CONCLUSIONS: LBBAP demonstrated a stable captured threshold, a low tip impedance and a high R-wave amplitude during the 12-month follow-up. Left ventricular remodeling was improved at 12 months post-procedure through LBBAP. The Sti-LVAT was a significant predictor of left ventricular remodeling. LBBAP demonstrated its feasibility, effectiveness, safety and some beneficial electrophysiological characteristics during this mid- to long-term follow-up, which should be confirmed by further studies.


Asunto(s)
Fascículo Atrioventricular , Marcapaso Artificial , Estimulación Cardíaca Artificial/efectos adversos , Estimulación Cardíaca Artificial/métodos , Electrocardiografía/métodos , Humanos , Remodelación Ventricular
3.
Front Cardiovasc Med ; 8: 748891, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34988124

RESUMEN

Sorting nexin 17 (SNX17), a member of sorting nexin (SNX) family, acts as a modulator for endocytic recycling of membrane proteins. Results from our previous study demonstrated the embryonic lethality of homozygous defect of SNX17. In this study, we investigated the role of SNX17 in rat fetal development. Specifically, we analyzed patterns of SNX17 messenger RNA (mRNA) expression in multiple rat tissues and found high expression in the cardiac outflow tract (OFT). This expression was gradually elevated during the cardiac OFT morphogenesis. Homozygous deletion of the SNX17 gene in rats resulted in mid-gestational embryonic lethality, which was accompanied by congenital heart defects, including the double-outlet right ventricle and atrioventricular and ventricular septal defects, whereas heterozygotes exhibited normal fetal development. Moreover, we found normal migration distance and the number of cardiac neural crest cells during the OFT morphogenesis. Although cellular proliferation in the cardiac OFT endocardial cushion was not affected, cellular apoptosis was significantly suppressed. Transcriptomic profiles and quantitative real-time PCR data in the cardiac OFT showed that SNX17 deletion resulted in abnormal expression of genes associated with cardiac development. Overall, these findings suggest that SNX17 plays a crucial role in cardiac development.

4.
Oxid Med Cell Longev ; 2020: 9815039, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33014281

RESUMEN

AIMS: We aimed to investigate whether LCZ696 protects against pathological cardiac hypertrophy by regulating the Sirt3/MnSOD pathway. METHODS: In vivo, we established a transverse aortic constriction animal model to establish pressure overload-induced heart failure. Subsequently, the mice were given LCZ696 by oral gavage for 4 weeks. After that, the mice underwent transthoracic echocardiography before they were sacrificed. In vitro, we introduced phenylephrine to prime neonatal rat cardiomyocytes and small-interfering RNA to knock down Sirt3 expression. RESULTS: Pathological hypertrophic stimuli caused cardiac hypertrophy and fibrosis and reduced the expression levels of Sirt3 and MnSOD. LCZ696 alleviated the accumulation of oxidative reactive oxygen species (ROS) and cardiomyocyte apoptosis. Furthermore, Sirt3 deficiency abolished the protective effect of LCZ696 on cardiomyocyte hypertrophy, indicating that LCZ696 induced the upregulation of MnSOD and phosphorylation of AMPK through a Sirt3-dependent pathway. CONCLUSIONS: LCZ696 may mitigate myocardium oxidative stress and apoptosis in pressure overload-induced heart failure by regulating the Sirt3/MnSOD pathway.


Asunto(s)
Aminobutiratos/farmacología , Estrés Oxidativo/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Sirtuina 3/metabolismo , Superóxido Dismutasa/metabolismo , Tetrazoles/farmacología , Remodelación Ventricular/efectos de los fármacos , Proteínas Quinasas Activadas por AMP/metabolismo , Aminobutiratos/uso terapéutico , Animales , Apoptosis/efectos de los fármacos , Compuestos de Bifenilo , Cardiomegalia/metabolismo , Cardiomegalia/patología , Cardiomegalia/prevención & control , Combinación de Medicamentos , Masculino , Ratones , Ratones Endogámicos C57BL , Miocardio/patología , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Sirtuina 3/antagonistas & inhibidores , Sirtuina 3/genética , Tetrazoles/uso terapéutico , Regulación hacia Arriba/efectos de los fármacos , Valsartán
5.
FASEB J ; 33(5): 6197-6208, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-30758987

RESUMEN

TLRs have been proven to be essential mediators for the early innate immune response. Overactivation of TLR-mediated immune signaling promotes deterioration of cardiovascular diseases; however, the role of TLRs in the heart under physiologic conditions remains neglected. Here, we show that Tlr3 deficiency induced the endoplasmic reticulum (ER) retention of Kv4.2/4.3 proteins and consequent degradation via the ubiquitin-proteasome pathway. Knockout of Tlr3 resulted in a prolonged QT interval (the space between the start of the Q wave and the end of the T wave) in mice with no significant signs of inflammation and tissue abnormality in cardiac muscles. Prolongation of action potential duration resulted from the depression of transient outward potassium channel (Ito) currents in Tlr3-deficient ventricular myocytes mirrored the change in QT interval. Mechanistically, we found that Tlr3 was exclusively localized in the ER of cardiomyocytes where it interacted with Kv4.2/4.3 subunits of Ito channel. Thus, our data indicated that TLR3 directly regulates Ito channel protein dynamics to maintain cardiac repolarization, which may implicate a new molecular surveillance system for cardiac electrophysiological homeostasis.-Gao, X., Gao, S., Guan, Y., Huang, L., Huang, J., Lin, L., Liu, Y., Zhao, H., Huang, B., Yuan, T., Liu, Y., Liang, D., Zhang, Y., Ma, X., Li, L., Li, J., Zhou, D., Shi, D., Xu, L., Chen, Y.-H. Toll-like receptor 3 controls QT interval on the electrocardiogram by targeting the degradation of Kv4.2/4.3 channels in the endoplasmic reticulum.


Asunto(s)
Electrocardiografía , Retículo Endoplásmico/metabolismo , Canales de Potasio Shal/metabolismo , Receptor Toll-Like 3/fisiología , Animales , Células HEK293 , Humanos , Ratones , Ratones Endogámicos C57BL , Miocitos Cardíacos/metabolismo , Complejo de la Endopetidasa Proteasomal/fisiología
6.
Arterioscler Thromb Vasc Biol ; 38(11): 2665-2677, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30354240

RESUMEN

Objective- Mitochondria are the important yet most underutilized target for cardio-cerebrovascular function integrity and disorders. The Tom (translocases of outer membrane) complex are the critical determinant of mitochondrial homeostasis for making organs acclimate physiological and pathological insults; however, their roles in the vascular system remain unknown. Approach and Results- A combination of studies in the vascular-specific transgenic zebrafish and genetically engineered mice was conducted. Vascular casting and imaging, endothelial angiogenesis, and mitochondrial protein import were performed to dissect potential mechanisms. A loss-of-function genetic screening in zebrafish identified that selective inactivation of the tomm7 (translocase of outer mitochondrial membrane 7) gene, which encodes a small subunit of the Tom complex, specially impaired cerebrovascular network formation. Ablation of the ortholog Tomm7 in mice recapitulated cerebrovascular abnormalities. Restoration of the cerebrovascular anomaly by an endothelial-specific transgenesis of tomm7 further indicated a defect in endothelial function. Mechanistically, Tomm7 deficit in endothelial cells induced an increased import of Rac1 (Ras-related C3 botulinum toxin substrate 1) protein into mitochondria and facilitated the mitochondrial Rac1-coupled redox signaling, which incurred angiogenic impairment that underlies cerebrovascular network malformation. Conclusions- Tomm7 drives brain angiogenesis and cerebrovascular network formation through modulating mitochondrial Rac1 signaling within the endothelium.


Asunto(s)
Encéfalo/irrigación sanguínea , Proteínas Portadoras/metabolismo , Células Endoteliales/enzimología , Endotelio Vascular/enzimología , Proteínas de la Membrana/metabolismo , Mitocondrias/enzimología , Proteínas Mitocondriales/metabolismo , Neovascularización Fisiológica , Neuropéptidos/metabolismo , Proteínas de Pez Cebra/metabolismo , Pez Cebra/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Animales , Proteínas Portadoras/genética , Células Cultivadas , Trastornos Cerebrovasculares/enzimología , Trastornos Cerebrovasculares/genética , Endotelio Vascular/embriología , Regulación del Desarrollo de la Expresión Génica , Regulación Enzimológica de la Expresión Génica , Humanos , Proteínas de la Membrana/genética , Ratones Noqueados , Proteínas del Complejo de Importación de Proteínas Precursoras Mitocondriales , Proteínas Mitocondriales/genética , Neovascularización Fisiológica/genética , Neuropéptidos/genética , Transducción de Señal , Pez Cebra/embriología , Pez Cebra/genética , Proteínas de Pez Cebra/genética , Proteína de Unión al GTP rac1/genética
7.
J Cell Mol Med ; 22(10): 4653-4663, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30070011

RESUMEN

Low-density lipoprotein receptor-related protein 6 (LRP6) serves as a Wnt coreceptor. Although Wnt/LRP6 signalling is best known for the ß-catenin-dependent regulation of target genes in tissue development and homeostasis, emerging evidence demonstrates the biological aspects of LRP6 beyond a Wnt coreceptor. Whether LRP6 modulates tissue development in a Wnt/ß-catenin signalling-independent manner remains unknown. Using a model of striated muscle development, we observed that LRP6 was almost undetectable in proliferating myoblasts, whereas its expression gradually increased in the nucleus of myodifferentiating cells. During myodifferentiation, LRP6 modulated the muscle-specific splicing of integrin-ß1D and consequent myotube maturation independently of the ß-catenin-dependent Wnt signalling. Furthermore, we identified that the carboxy-terminal serine-rich region in LRP6 bond to the adenine-rich sequence within alternative exon D (AED) of integrin-ß1 pre-mRNA, and therefore, elicited AED inclusion when the spliceosome was recruited to the splice site. The interaction of LRP6 with the adenine-rich sequence was sufficient to overcome AED exclusion by a splicing repressor, polypyrimidine tract binding protein-1. Besides the integrin-ß1, deep RNA sequencing in different types of cells revealed that the LRP6-mediated splicing regulation was widespread. Thus, our findings implicate LRP6 as a potential regulator for alternative pre-mRNA splicing.


Asunto(s)
Empalme Alternativo , Regulación del Desarrollo de la Expresión Génica , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad/genética , Desarrollo de Músculos/genética , Músculo Estriado/metabolismo , Precursores del ARN/genética , Animales , Animales Recién Nacidos , Secuencia de Bases , Diferenciación Celular , Línea Celular , Núcleo Celular/metabolismo , Proliferación Celular , Citosol/metabolismo , Exones , Ribonucleoproteínas Nucleares Heterogéneas/genética , Ribonucleoproteínas Nucleares Heterogéneas/metabolismo , Células Endoteliales de la Vena Umbilical Humana/citología , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Integrina beta1/genética , Integrina beta1/metabolismo , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad/metabolismo , Ratones , Músculo Estriado/citología , Músculo Estriado/crecimiento & desarrollo , Mioblastos/citología , Mioblastos/metabolismo , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Proteína de Unión al Tracto de Polipirimidina/genética , Proteína de Unión al Tracto de Polipirimidina/metabolismo , Cultivo Primario de Células , Precursores del ARN/metabolismo , Ratas , Vía de Señalización Wnt , beta Catenina/genética , beta Catenina/metabolismo
8.
Nat Commun ; 7: 11775, 2016 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-27250245

RESUMEN

Low-density lipoprotein receptor-related protein 6 (LRP6) is a Wnt co-receptor in the canonical Wnt/ß-catenin signalling. Here, we report the scaffold function of LRP6 in gap junction formation of cardiomyocytes. Cardiac LRP6 is spatially restricted to intercalated discs and binds to gap junction protein connexin 43 (Cx43). A deficiency in LRP6 disrupts Cx43 gap junction formation and thereby impairs the cell-to-cell coupling, which is independent of Wnt/ß-catenin signalling. The defect in Cx43 gap junction resulting from LRP6 reduction is attributable to the defective traffic of de novo Cx43 proteins from the endoplasmic reticulum to the Golgi apparatus, leading to the lysosomal degradation of Cx43 proteins. Accordingly, the hearts of conditional cardiac-specific Lrp6-knockout mice consistently exhibit overt reduction of Cx43 gap junction plaques without any abnormality in Wnt signalling and are predisposed to lethal arrhythmias. These findings uncover a distinct role of LRP6 as a platform for intracellular protein trafficking.


Asunto(s)
Arritmias Cardíacas/genética , Uniones Comunicantes/metabolismo , Ventrículos Cardíacos/metabolismo , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad/genética , Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Animales , Arritmias Cardíacas/metabolismo , Arritmias Cardíacas/fisiopatología , Conexina 43/genética , Conexina 43/metabolismo , Ecocardiografía , Retículo Endoplásmico/metabolismo , Retículo Endoplásmico/ultraestructura , Uniones Comunicantes/ultraestructura , Regulación de la Expresión Génica , Aparato de Golgi/metabolismo , Aparato de Golgi/ultraestructura , Ventrículos Cardíacos/patología , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad/deficiencia , Ratones , Ratones Noqueados , Miocardio/patología , Miocitos Cardíacos/patología , Técnicas de Cultivo de Órganos , Cultivo Primario de Células , Transporte de Proteínas , Ratas , Ratas Sprague-Dawley , Transducción de Señal , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
9.
J Mol Cell Biol ; 7(5): 476-85, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26260029

RESUMEN

The mammalian nuclear pore complex is comprised of ∼ 30 different nucleoporins (Nups). It governs the nuclear import of gene expression modulators and the export of mRNAs. In cardiomyocytes, Na(+)-H(+) exchanger-1 (NHE1) is an integral membrane protein that exclusively regulates intracellular pH (pHi) by exchanging one intracellular H(+) for one extracellular Na(+). However, the role of Nups in cardiac NHE1 expression remains unknown. We herein report that Nup35 regulates cardiomyocyte NHE1 expression by controlling the nucleo-cytoplasmic trafficking of nhe1 mRNA. The N-terminal domain of Nup35 determines nhe1 mRNA nuclear export by targeting the 5'-UTR (-412 to -213 nt) of nhe1 mRNA. Nup35 ablation weakens the resistance of cardiomyocytes to an acid challenge by depressing NHE1 expression. Moreover, we identify that Nup35 and NHE1 are simultaneously downregulated in ischemic cardiomyocytes both in vivo and in vitro. Enforced expression of Nup35 effectively counteracts the anoxia-induced intracellular acidification. We conclude that Nup35 selectively regulates cardiomyocyte pHi homeostasis by posttranscriptionally controlling NHE1 expression. This finding reveals a novel regulatory mechanism of cardiomyocyte pHi, and may provide insight into the therapeutic strategy for ischemic cardiac diseases.


Asunto(s)
Proteínas de Transporte de Catión/metabolismo , Miocitos Cardíacos/metabolismo , Intercambiadores de Sodio-Hidrógeno/metabolismo , Proteínas de Transporte de Catión/genética , Línea Celular , Homeostasis/genética , Homeostasis/fisiología , Humanos , Concentración de Iones de Hidrógeno , Hibridación Fluorescente in Situ , Transporte de Proteínas/genética , Transporte de Proteínas/fisiología , Intercambiador 1 de Sodio-Hidrógeno , Intercambiadores de Sodio-Hidrógeno/genética
10.
PLoS One ; 10(3): e0120550, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25806802

RESUMEN

The cellular mechanisms of primary varicose great saphenous veins (GSVs) involve inflammation, apoptosis, and proliferation of local cells and extracellular matrix degradation. Long non-coding RNAs (lncRNAs) play important roles in these cellular processes; however, which and how lncRNAs related to these mechanisms take effect on GSVs remain unclear. By screening lncRNAs that might experience changes in GSV varicosities, we selected the lower expressed lncRNA-GAS5 (growth arrest specific transcript 5) for functional assessments. Silencing of lncRNA-GAS5 promoted cell proliferation and migration, and cell cycle of the human saphenous vein smooth muscle cells (HSVSMCs), whereas overexpressing it inhibited these cellular behaviors and reduced apoptosis of HSVSMCs. RNA pull-down experiment revealed a direct bind of lncRNA-GAS5 to a Ca2+-dependent RNA-binding protein, Annexin A2. Further experiments showed that silencing of Annexin A2 reduced the HSVSMCs proliferation and vice versa. In the context of lncRNA-GAS5 knockdown, silencing of Annexin A2 reduced the proliferation of HSVSMCs while overexpression of Annexin A2 increased the proliferation. Thus, the low expression of lncRNA-GAS5 may facilitate HSVSMCs proliferation and migration through Annexin A2 and thereby the pathogenesis of GSV varicosities.


Asunto(s)
Miocitos del Músculo Liso/metabolismo , ARN Largo no Codificante/metabolismo , Vena Safena/citología , Adulto , Anciano , Anexina A2/química , Anexina A2/metabolismo , Apoptosis , Movimiento Celular , Proliferación Celular , Células Cultivadas , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Femenino , Humanos , Masculino , Persona de Mediana Edad , Miocitos del Músculo Liso/citología , Unión Proteica , Interferencia de ARN , ARN Largo no Codificante/antagonistas & inhibidores , ARN Largo no Codificante/genética , ARN Interferente Pequeño/metabolismo
11.
Nat Commun ; 5: 5177, 2014 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-25295779

RESUMEN

Heart failure (HF) is associated with complicated molecular remodelling within cardiomyocytes; however, the mechanisms underlying this process remain unclear. Here we show that sorting nexin-13 (SNX13), a member of both the sorting nexin and the regulator of G protein signalling (RGS) protein families, is a potent mediator of HF. Decreased levels of SNX13 are observed in failing hearts of humans and of experimental animals. SNX13-deficient zebrafish recapitulate HF with striking cardiomyocyte apoptosis. Mechanistically, a reduction in SNX13 expression facilitates the degradative sorting of apoptosis repressor with caspase recruitment domain (ARC), which is a multifunctional inhibitor of apoptosis. Consequently, the apoptotic pathway is activated, resulting in the loss of cardiac cells and the dampening of cardiac function. The N-terminal PXA structure of SNX13 is responsible for mediating the endosomal trafficking of ARC. Thus, this study reveals that SNX13 profoundly affects cardiac performance through the SNX13-PXA-ARC-caspase signalling pathway.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Apoptosis , Insuficiencia Cardíaca/metabolismo , Proteínas Musculares/metabolismo , Miocitos Cardíacos/metabolismo , Nexinas de Clasificación/metabolismo , Animales , Caspasa 8/metabolismo , Modelos Animales de Enfermedad , Humanos , Ratones , Pez Cebra
12.
J Cell Mol Med ; 18(10): 1992-9, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25092467

RESUMEN

Ischaemic cardiac arrhythmias cause a large proportion of sudden cardiac deaths worldwide. The ischaemic arrhythmogenesis is primarily because of the dysfunction and adverse remodelling of sarcolemma ion channels. However, the potential regulators of sarcolemma ion channel turnover and function in ischaemic cardiac arrhythmias remains unknown. Our previous studies indicate that dynamin-2 (DNM2), a cardiac membrane-remodelling GTPase, modulates ion channels membrane trafficking in the cardiomyocytes. Here, we have found that DNM2 plays an important role in acute ischaemic arrhythmias. In rat ventricular tissues and primary cardiomyocytes subjected to acute ischaemic stress, the DNM2 protein and transcription levels were markedly down-regulated. This DNM2 reduction was coupled with severe ventricular arrhythmias. Moreover, we identified that the down-regulation of DNM2 within cardiomyocytes increases the action potential amplitude and prolongs the re-polarization duration by depressing the retrograde trafficking of Nav1.5 and Kir2.1 channels. These effects are likely to account for the DNM2 defect-induced arrhythmogenic potentials. These results suggest that DNM2, with its multi-ion channel targeting properties, could be a promising target for novel antiarrhythmic therapies.


Asunto(s)
Arritmias Cardíacas/metabolismo , Dinamina II/metabolismo , Isquemia/metabolismo , Miocitos Cardíacos/metabolismo , Canal de Sodio Activado por Voltaje NAV1.5/metabolismo , Canales de Potasio de Rectificación Interna/metabolismo , Animales , Arritmias Cardíacas/genética , Arritmias Cardíacas/patología , Biotinilación , Western Blotting , Células Cultivadas , Dinamina II/genética , Electrocardiografía , Electrofisiología , Técnicas para Inmunoenzimas , Isquemia/genética , Isquemia/patología , Masculino , Miocitos Cardíacos/patología , Canal de Sodio Activado por Voltaje NAV1.5/genética , Canales de Potasio de Rectificación Interna/genética , ARN Mensajero/genética , Ratas , Ratas Sprague-Dawley , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
13.
PLoS One ; 9(7): e103097, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25068583

RESUMEN

microRNAs (miRNAs) play essential roles in cardiogenesis. The altered expression of miRNAs can result in cardiac malformations by inducing abnormalities in the behavior of cardiac cells. However, the role of miR-10a in the regulation of cardiomyocyte progenitor cells (CMPCs) remains undetermined. In the present study, we found that up- or down-regulation of miR-10a inhibited or promoted the proliferation of human CMPCs, respectively, without affecting their differentiation toward cardiomyocytes. miR-10a bound to GATA6 directly and reduced GATA6 expression. Over-expression of GATA6 greatly attenuated the miR-10a-mediated inhibitory effect on the proliferation of human CMPCs. Thus, our results indicate that miR-10a could effectively modulate the proliferation of human CMPCs by targeting GATA6. The finding provides novel insights into the potency of miR-10a during heart development.


Asunto(s)
Factor de Transcripción GATA6/genética , MicroARNs/genética , Mioblastos Cardíacos/metabolismo , Interferencia de ARN , ARN Mensajero/genética , Animales , Diferenciación Celular/genética , Proliferación Celular , Células Cultivadas , Regulación hacia Abajo , Regulación de la Expresión Génica , Humanos , Ratones , MicroARNs/química , Mioblastos Cardíacos/citología , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , ARN Mensajero/química
14.
Cell Res ; 24(8): 977-93, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25022898

RESUMEN

Pathological cardiac hypertrophy is an inevitable forerunner of heart failure. Regardless of the etiology of cardiac hypertrophy, cardiomyocyte mitochondrial alterations are always observed in this context. The translocases of mitochondrial outer membrane (Tom) complex governs the import of mitochondrial precursor proteins to maintain mitochondrial function under pathophysiological conditions; however, its role in the development of pathological cardiac hypertrophy remains unclear. Here, we showed that Tom70 was downregulated in pathological hypertrophic hearts from humans and experimental animals. The reduction in Tom70 expression produced distinct pathological cardiomyocyte hypertrophy both in vivo and in vitro. The defective mitochondrial import of Tom70-targeted optic atrophy-1 triggered intracellular oxidative stress, which led to a pathological cellular response. Importantly, increased Tom70 levels provided cardiomyocytes with full resistance to diverse pro-hypertrophic insults. Together, these results reveal that Tom70 acts as a molecular switch that orchestrates hypertrophic stresses and mitochondrial responses to determine pathological cardiac hypertrophy.


Asunto(s)
Cardiomegalia/patología , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Animales , Cardiomegalia/metabolismo , Modelos Animales de Enfermedad , Regulación hacia Abajo , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/patología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas de Transporte de Membrana Mitocondrial/antagonistas & inhibidores , Proteínas de Transporte de Membrana Mitocondrial/genética , Proteínas del Complejo de Importación de Proteínas Precursoras Mitocondriales , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Estrés Oxidativo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Ratas , Ratas Sprague-Dawley , Pez Cebra/metabolismo
15.
PLoS One ; 9(1): e86156, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24497937

RESUMEN

Long non-coding RNAs (lncRNAs) are key regulatory molecules involved in a variety of biological processes and human diseases. However, the pathological effects of lncRNAs on primary varicose great saphenous veins (GSVs) remain unclear. The purpose of the present study was to identify aberrantly expressed lncRNAs involved in the prevalence of GSV varicosities and predict their potential functions. Using microarray with 33,045 lncRNA and 30,215 mRNA probes, 557 lncRNAs and 980 mRNAs that differed significantly in expression between the varicose great saphenous veins and control veins were identified in six pairs of samples. These lncRNAs were sub-grouped and mRNAs expressed at different levels were clustered into several pathways with six focused on metabolic pathways. Quantitative real-time PCR replication of nine lncRNAs was performed in 32 subjects, validating six lncRNAs (AF119885, AK021444, NR_027830, G36810, NR_027927, uc.345-). A coding-non-coding gene co-expression network revealed that four of these six lncRNAs may be correlated with 11 mRNAs and pathway analysis revealed that they may be correlated with another 8 mRNAs associated with metabolic pathways. In conclusion, aberrantly expressed lncRNAs for GSV varicosities were here systematically screened and validated and their functions were predicted. These findings provide novel insight into the physiology of lncRNAs and the pathogenesis of varicose veins for further investigation. These aberrantly expressed lncRNAs may serve as new therapeutic targets for varicose veins. The Human Ethnics Committee of Shanghai East Hospital, Tongji University School of Medicine approved the study (NO.: 2011-DF-53).


Asunto(s)
Perfilación de la Expresión Génica , ARN Largo no Codificante/genética , Vena Safena/metabolismo , Várices/genética , Adulto , Anciano , Femenino , Ontología de Genes , Redes Reguladoras de Genes , Humanos , Masculino , Persona de Mediana Edad , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Vena Safena/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...