Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 69
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Eur Heart J ; 45(9): 688-703, 2024 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-38152853

RESUMEN

BACKGROUND AND AIMS: Anti-hypertensive agents are one of the most frequently used drugs worldwide. However, no blood pressure-lowering strategy is superior to placebo with respect to survival in diabetic hypertensive patients. Previous findings show that Wnt co-receptors LDL receptor-related proteins 5 and 6 (LRP5/6) can directly bind to several G protein-coupled receptors (GPCRs). Because angiotensin II type 1 receptor (AT1R) is the most important GPCR in regulating hypertension, this study examines the possible mechanistic association between LRP5/6 and their binding protein Dickkopf-1 (DKK1) and activation of the AT1R and further hypothesizes that the LRP5/6-GPCR interaction may affect hypertension and potentiate cardiac impairment in the setting of diabetes. METHODS: The roles of serum DKK1 and DKK1-LRP5/6 signalling in diabetic injuries were investigated in human and diabetic mice. RESULTS: Blood pressure up-regulation positively correlated with serum DKK1 elevations in humans. Notably, LRP5/6 physically and functionally interacted with AT1R. The loss of membrane LRP5/6 caused by injection of a recombinant DKK1 protein or conditional LRP5/6 deletions resulted in AT1R activation and hypertension, as well as ß-arrestin1 activation and cardiac impairment, possibly because of multiple GPCR alterations. Importantly, unlike commonly used anti-hypertensive agents, administration of the anti-DKK1 neutralizing antibody effectively prevented diabetic cardiac impairment in mice. CONCLUSIONS: These findings establish a novel DKK1-LRP5/6-GPCR pathway in inducing diabetic injuries and may resolve the long-standing conundrum as to why elevated blood DKK1 has deleterious effects. Thus, monitoring and therapeutic elimination of blood DKK1 may be a promising strategy to attenuate diabetic injuries.


Asunto(s)
Diabetes Mellitus Experimental , Cardiomiopatías Diabéticas , Hipertensión , Receptores de LDL , Animales , Humanos , Ratones , Antihipertensivos , Cardiomiopatías Diabéticas/prevención & control , Hipertensión/prevención & control , Receptores de LDL/antagonistas & inhibidores
2.
Stem Cells ; 39(4): 443-457, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33426760

RESUMEN

ERG1, a potassium ion channel, is essential for cardiac action potential repolarization phase. However, the role of ERG1 for normal development of the heart is poorly understood. Using the rat embryonic stem cells (rESCs) model, we show that ERG1 is crucial in cardiomyocyte lineage commitment via interactions with Integrin ß1. In the mesoderm phase of rESCs, the interaction of ERG1 with Integrin ß1 can activate the AKT pathway by recruiting and phosphorylating PI3K p85 and focal adhesion kinase (FAK) to further phosphorylate AKT. Activation of AKT pathway promotes cardiomyocyte differentiation through two different mechanisms, (a) through phosphorylation of GSK3ß to upregulate the expression levels of ß-catenin and Gata4; (b) through promotion of nuclear translocation of nuclear factor-κB by phosphorylating IKKß to inhibit cell apoptosis, which occurs due to increased Bcl2 expression. Our study provides solid evidence for a novel role of ERG1 on differentiation of rESCs into cardiomyocytes.


Asunto(s)
Canal de Potasio ERG1/genética , Células Madre Embrionarias/metabolismo , Regulación del Desarrollo de la Expresión Génica , Integrina beta1/genética , Miocitos Cardíacos/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Animales , Apoptosis/genética , Diferenciación Celular , Fosfatidilinositol 3-Quinasa Clase Ia/genética , Fosfatidilinositol 3-Quinasa Clase Ia/metabolismo , Canal de Potasio ERG1/metabolismo , Embrión de Mamíferos , Células Madre Embrionarias/citología , Factor de Transcripción GATA4/genética , Factor de Transcripción GATA4/metabolismo , Glucógeno Sintasa Quinasa 3 beta/genética , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Quinasa I-kappa B/genética , Quinasa I-kappa B/metabolismo , Integrina beta1/metabolismo , Miocitos Cardíacos/citología , FN-kappa B/genética , FN-kappa B/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Ratas , Transducción de Señal , beta Catenina/genética , beta Catenina/metabolismo
3.
Biochem Biophys Res Commun ; 571: 8-13, 2021 09 24.
Artículo en Inglés | MEDLINE | ID: mdl-34298338

RESUMEN

Rats have long been an ideal model for disease research in the field of biomedicine, but the bottleneck of in vitro culture of rat embryonic stem (ES) cells hindered the wide application as genetic disease models. Here, we optimized a special medium which we named 5N-medium for rat embryonic stem cells, which improved the in vitro cells with better morphology and higher pluripotency. We then established a drug selection schedule harboring a prior selection of 12 h that achieved a higher positive selection ratio. These treatments induced at least 50% increase of homologous recombination efficiency compared with conventional 2i culture condition. Moreover, the ratio of euploid ES clones also increased by 50% with a higher germline transmission rate. Finally, we successfully knocked in a 175 kb human Bacterial Artificial Chromosome (BAC) fragment to rat ES genome through recombinase mediated cassette exchange (RMCE). Hence, we provide a promising system for generating sophisticated rat models which could be benefit for biomedical researches.


Asunto(s)
Células Madre Embrionarias/citología , Animales , Proliferación Celular , Células Cultivadas , Modelos Animales , Ratas , Ratas Endogámicas F344 , Ratas Sprague-Dawley
4.
Thromb J ; 19(1): 8, 2021 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-33568153

RESUMEN

BACKGROUND: The progression of coagulation in COVID-19 patients with confirmed discharge status and the combination of autopsy with complete hemostasis parameters have not been well studied. OBJECTIVE: To clarify the thrombotic phenomena and hemostasis state in COVID-19 patients based on epidemiological statistics combining autopsy and statistical analysis. METHODS: Using autopsy results from 9 patients with COVID-19 pneumonia and the medical records of 407 patients, including 39 deceased patients whose discharge status was certain, time-sequential changes in 11 relevant indices within mild, severe and critical infection throughout hospitalization according to the Chinese National Health Commission (NHC) guidelines were evaluated. Statistical tools were applied to calculate the importance of 11 indices and the correlation between those indices and the severity of COVID-19. RESULTS: At the beginning of hospitalization, platelet (PLT) counts were significantly reduced in critically ill patients compared with severely or mildly ill patients. Blood glucose (GLU), prothrombin time (PT), activated partial thromboplastin time (APTT), and D-dimer levels in critical patients were increased compared with mild and severe patients during the entire admission period. The International Society on Thrombosis and Haemostasis (ISTH) disseminated intravascular coagulation (DIC) score was also high in critical patients. In the relatively late stage of nonsurvivors, the temporal changes in PLT count, PT, and D-dimer levels were significantly different from those in survivors. A random forest model indicated that the most important feature was PT followed by D-dimer, indicating their positive associations with disease severity. Autopsy of deceased patients fulfilling diagnostic criteria for DIC revealed microthromboses in multiple organs. CONCLUSIONS: Combining autopsy data, time-sequential changes and statistical methods to explore hemostasis-relevant indices among the different severities of the disease helps guide therapy and detect prognosis in COVID-19 infection.

5.
J Cell Mol Med ; 23(5): 3629-3640, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30907509

RESUMEN

The generation of germline competent rat embryonic stem cells (rESCs) allows the study of their lineage commitment. Here, we developed a highly efficient system for rESC-derived cardiomyocytes, and even the formation of three-dimensional (3D)-like cell clusters with cTNT and α-Actinin. We have validated that laminin can interact with membrane integrin to promote the phosphorylation of both phosphatidylinositol 3-kinase (PI3K) p85 and the focal adhesion kinase (FAK). In parallel, GATA4 was up-regulated. Upon inhibiting the integrin, laminin loses the effect on cardiomyocyte differentiation, accompanied with a down-regulation of phosphorylation level of PI3K p85 and FAK. Meanwhile, the expression of Gata4 was inhibited as well. Taken together, laminin is a crucial component in the differentiation of rESCs into cardiomyocytes through increasing their proliferation via interacting with integrin pathway. These results provide new insights into the pathways mediated by extracellular laminin involved in the fate of rESC-derived cardiomyocytes.


Asunto(s)
Diferenciación Celular/fisiología , Fosfatidilinositol 3-Quinasa Clase Ia/metabolismo , Células Madre Embrionarias/metabolismo , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Integrinas/metabolismo , Laminina/metabolismo , Miocitos Cardíacos/metabolismo , Animales , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Células Cultivadas , Células Madre Embrionarias/citología , Células Madre Embrionarias/efectos de los fármacos , Factor de Transcripción GATA4/genética , Factor de Transcripción GATA4/metabolismo , Expresión Génica/efectos de los fármacos , Laminina/farmacología , Miocitos Cardíacos/citología , Miocitos Cardíacos/efectos de los fármacos , Fosforilación/efectos de los fármacos , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Transducción de Señal/fisiología
6.
J Cell Mol Med ; 23(11): 7796-7809, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31503410

RESUMEN

Sepsis-induced cardiac dysfunction represents a main cause of death in intensive care units. Previous studies have indicated that GSK-3ß is involved in the modulation of sepsis. However, the signalling details of GSK-3ß regulation in endotoxin lipopolysaccharide (LPS)-induced septic myocardial dysfunction are still unclear. Here, based on the rat septic myocardial injury model, we found that LPS could induce GSK-3ß phosphorylation at its active site (Y216) and up-regulate FOXO3A level in primary cardiomyocytes. The FOXO3A expression was significantly reduced by GSK-3ß inhibitors and further reversed through ß-catenin knock-down. This pharmacological inhibition of GSK-3ß attenuated the LPS-induced cell injury via mediating ß-catenin signalling, which could be abolished by FOXO3A activation. In vivo, GSK-3ß suppression consistently improved cardiac function and relieved heart injury induced by LPS. In addition, the increase in inflammatory cytokines in LPS-induced model was also blocked by inhibition of GSK-3ß, which curbed both ERK and NF-κB pathways, and suppressed cardiomyocyte apoptosis via activating the AMP-activated protein kinase (AMPK). Our results demonstrate that GSK-3ß inhibition attenuates myocardial injury induced by endotoxin that mediates the activation of FOXO3A, which suggests a potential target for the therapy of septic cardiac dysfunction.


Asunto(s)
Cardiotónicos/farmacología , Proteína Forkhead Box O3/metabolismo , Glucógeno Sintasa Quinasa 3 beta/antagonistas & inhibidores , Inflamación/patología , Miocardio/patología , Inhibidores de Proteínas Quinasas/farmacología , Adenilato Quinasa/metabolismo , Animales , Apoptosis/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Pruebas de Función Cardíaca , Lipopolisacáridos , Masculino , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , FN-kappa B/metabolismo , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos , beta Catenina/metabolismo
7.
J Cell Biochem ; 118(12): 4460-4467, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28464250

RESUMEN

The cardiomyocyte differentiation from mouse embryonic stem cells (mESCs) is a dynamic and complex process that involved in the precision regulation of histone acetylation. The formation of action potential (AP) in mature cardiomyocytes is based on the expression pattern of Na+ , Ca2+ , and K+ ion channels, in which the slow delayed rectifier potassium current (IKs ), the rapid delayed rectifier potassium current (IKr ) and the inwardly rectifying Kir current (IK1 ) mainly contribute to repolarization for AP in different species. However, the expression status of potassium channels conducted IKs , IKr , and IK1 in cardiomyocyte differentiation are not fully defined. Here, we investigated the expression pattern of the slow delayed rectifier potassium channel and the rapid delayed rectifier potassium channel using a model of mouse cardiomyocyte differentiation under different conditions of histone acetylation. We found that expression levels of both the delayed rectifier potassium channel and the inwardly rectifying potassium channel were more sensitive to histone hyperacetylation during differentiation from mESCs into cardiomyocytes. Especially, histone H4 hyperacetylation induced by Class I HDACs inhibitors promoted the expression profiles of potassium channels (Kcnj2, Kcnj3, Kcnj5, Kcnj11, and Kcnh2) in the process. Our results provide a clue for expression status of potassium channels which may be essential to forming functional cardiomyocyte in the cardiac lineage commitment of mESC. J. Cell. Biochem. 118: 4460-4467, 2017. © 2017 Wiley Periodicals, Inc.


Asunto(s)
Regulación de la Expresión Génica/efectos de los fármacos , Inhibidores de Histona Desacetilasas/farmacología , Histonas/metabolismo , Células Madre Embrionarias de Ratones/metabolismo , Miocitos Cardíacos/metabolismo , Acetilación/efectos de los fármacos , Animales , Perfilación de la Expresión Génica , Ratones
8.
Int J Mol Sci ; 17(10)2016 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-27735862

RESUMEN

As a physiological small molecular product from the microbial fermentation of dietary fibers, butyrate plays an important role in maintaining intestinal health. Our previous works have proved that the effect of sodium butyrate (NaB) on the intestinal barrier function is mediated by activation of AMP-activated protein kinase (AMPK). However, the detailed pathway involved remains unknown. Using the calcium switch assay in the Caco-2 cell monolayer model, we found here that NaB activated AMPK mainly by increasing the calcium level, but not the ATP concentration, via promoting store-operated calcium entry (SOCE). Upon the activation of AMPK, NaB promoted the reassembly of tight junctions (TJs) based on reducing the phosphorylation of myosin II regulatory light chain (MLC2) at Ser19 and increasing phosphorylation of protein kinase C ß2 (PKCß2) at Ser660. Inhibiting (protein kinase C ß) PKCß blocked the reassembly of TJs induced by NaB in the barrier monolayer model. These results indicated that NaB could activate the calcium/calmodulin-dependent protein kinase kinase ß (CaMKKß) pathway to mediate AMPK phosphorylating, which then inhibited the phosphorylation of MLC2 and promoted the phosphorylation of PKCß2, respectively, so that the downstream molecules of AMPK coordinately contributed to the reassembly of TJs in the Caco-2 barrier model. These results suggested a potential mechanism of butyrate for intestine homeostasis and protection.


Asunto(s)
Ácido Butírico/farmacología , Miosinas Cardíacas/metabolismo , Cadenas Ligeras de Miosina/metabolismo , Quinasa de Cadena Ligera de Miosina/metabolismo , Proteína Quinasa C beta/metabolismo , Uniones Estrechas/efectos de los fármacos , Proteínas Quinasas Activadas por AMP/metabolismo , Western Blotting , Células CACO-2 , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/metabolismo , Miosinas Cardíacas/antagonistas & inhibidores , Humanos , Inmunoprecipitación , Cadenas Ligeras de Miosina/antagonistas & inhibidores , Quinasa de Cadena Ligera de Miosina/antagonistas & inhibidores , Fosforilación/efectos de los fármacos , Uniones Estrechas/metabolismo
9.
Int J Mol Sci ; 17(6)2016 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-27258257

RESUMEN

MicroRNAs (miRNAs) have been identified as key players in cardiomyocyte hypertrophy, which is associated with significant risks of heart failure. However, many microRNAs are still not recognized for their functions in pathophysiological processes. In this study, we evaluated effects of miR-218 in cardiomyocyte hypertrophy using both in vitro and in vivo models. We found that miR-218 was evidently downregulated in a transverse aortic constriction (TAC) mouse model. Overexpression of miR-218 is sufficient to reduce hypertrophy, whereas the suppression of miR-218 aggravates hypertrophy in primary cardiomyocytes induced by isoprenaline (ISO). In addition, we identified RE1-silencing transcription factor (REST) as a novel target of miR-218; it negatively regulated the expression of REST in hypertrophic cardiomyocytes and the TAC model. These results showed that miR-218 plays a crucial role in cardiomyocyte hypertrophy, likely via targeting REST, suggesting a potential candidate target for interfering hypertrophy.


Asunto(s)
Constricción Patológica/genética , Isoproterenol/efectos adversos , MicroARNs/genética , Miocitos Cardíacos/patología , Proteínas Represoras/genética , Animales , Células Cultivadas , Constricción Patológica/inducido químicamente , Modelos Animales de Enfermedad , Regulación hacia Abajo , Regulación de la Expresión Génica/efectos de los fármacos , Ratones , Miocitos Cardíacos/citología , Miocitos Cardíacos/efectos de los fármacos , Ratas , Transducción de Señal
10.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 33(6): 875-877, 2016 Dec 10.
Artículo en Zh | MEDLINE | ID: mdl-27984626

RESUMEN

Heart is the first organ to function during mammalian embryogenesis. The differentiation of embryonic stem cells (ESCs) into cardiomyocyte is complex and dynamic, which involves 4 differentiation stages including ESCs, mesoderm, cardiac precursor, and terminal cardiomyocytes. Abnormal expression of certain genes can lead to congenital heart diseases during cardiomyocyte differentiation. Epigenetic regulation plays a crucial role on the switch of gene activation and deactivation during cardiomyocyte differentiation. Non-coding RNA, particularly microRNA and long non-coding RNA, may significantly influence gene expression. Exploring the regulatory roles of non-coding RNA in cardiomyocyte differentiation may contribute to the understanding of the functions of myocardial cells and mechanism of congenital heart diseases.


Asunto(s)
Diferenciación Celular/genética , Miocitos Cardíacos/fisiología , ARN Largo no Codificante/genética , Animales , Embrión de Mamíferos/fisiología , Células Madre Embrionarias/fisiología , Epigénesis Genética/genética , Humanos
11.
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
12.
Mol Cell Biochem ; 391(1-2): 1-9, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24573887

RESUMEN

Importance of actin organization in control of chondrocyte phenotype is well established, but little is known about the role of transforming growth factor-ß1 (TFGß1) in regulating of ROCK I signal pathway. Here, we investigated the role of the TGFß1, a well-studied member of the TGF-ß superfamily, in chondrogenesis. Newborn Rats were randomly assigned to developmental dysplasia of the hip (DDH) group and control group. The isolated hips were performed with HE staining and immunohistochemistry. The chondrocytes was isolated and stained by immunofluorescence. The relative quantification of TGFß1 on mRNA level was determined using real-time RT-PCR, and its secretion in culture supernatant in each well was detected by means of ELISA. The expression of ROCK I and ROCK II was detected by means of Western Blot. The relative amounts of actin in detergent-soluble and insoluble fractions were determined. Furthermore, TGFß1 were employed to stimulate normal primary culture chondrocytes in vitro. We found TFGß1 significantly changed in acetabulum chondrocytes after mechanical overloading. Over expression of TFGß1 was observed by means of RT-PCR and ELISA assay. The expression of ROCK I was significantly increased in DDH acetabulum chondrocytes compared with normal cells. The detergent-soluble actin was confirmed reorganization in DDH chondrocytes. Furthermore, TFGß1 can stimulate the ROCK I signaling to modulate actin location in vitro. In conclusion, our data suggested that TFGß1 expression suppresses chondrogenesis through the control of ROCK signaling and actin organization.


Asunto(s)
Luxación de la Cadera/enzimología , Luxación de la Cadera/patología , Transducción de Señal , Factor de Crecimiento Transformador beta1/metabolismo , Quinasas Asociadas a rho/metabolismo , Acetábulo/patología , Actinas/metabolismo , Animales , Diferenciación Celular/efectos de los fármacos , Forma de la Célula/efectos de los fármacos , Condrocitos/efectos de los fármacos , Condrocitos/metabolismo , Condrocitos/patología , Modelos Animales de Enfermedad , Femenino , Modelos Biológicos , Fenotipo , Ratas Wistar , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal/efectos de los fármacos , Factor de Crecimiento Transformador beta1/farmacología
13.
BMC Biol ; 11: 107, 2013 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-24131868

RESUMEN

BACKGROUND: Neural crest defects lead to congenital heart disease involving outflow tract malformation. Integrin-linked-kinase (ILK) plays important roles in multiple cellular processes and embryogenesis. ILK is expressed in the neural crest, but its role in neural crest and outflow tract morphogenesis remains unknown. RESULTS: We ablated ILK specifically in the neural crest using the Wnt1-Cre transgene. ILK ablation resulted in abnormal migration and overpopulation of neural crest cells in the pharyngeal arches and outflow tract and a significant reduction in the expression of neural cell adhesion molecule (NCAM) and extracellular matrix components. ILK mutant embryos exhibited an enlarged common arterial trunk and ventricular septal defect. Reduced smooth muscle differentiation, but increased ossification and neurogenesis/innervation were observed in ILK mutant outflow tract that may partly be due to reduced transforming growth factor ß2 (TGFß2) but increased bone morphogenetic protein (BMP) signaling. Consistent with these observations, microarray analysis of fluorescence-activated cell sorting (FACS)-sorted neural crest cells revealed reduced expression of genes associated with muscle differentiation, but increased expression of genes of neurogenesis and osteogenesis. CONCLUSIONS: Our results demonstrate that ILK plays essential roles in neural crest and outflow tract development by mediating complex crosstalk between cell matrix and multiple signaling pathways. Changes in these pathways may collectively result in the unique neural crest and outflow tract phenotypes observed in ILK mutants.


Asunto(s)
Diferenciación Celular , Movimiento Celular , Cresta Neural/citología , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Proteínas Morfogenéticas Óseas/metabolismo , Adhesión Celular , Embrión de Mamíferos , Femenino , Eliminación de Gen , Regulación del Desarrollo de la Expresión Génica , Ratones , Ratones Noqueados , Músculo Liso/citología , Moléculas de Adhesión de Célula Nerviosa/metabolismo , Cresta Neural/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta2/metabolismo , Proteína Wnt1/genética
14.
J Genet Genomics ; 2024 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-38575112

RESUMEN

Pathological myocardial hypertrophy is a common early clinical manifestation of heart failure, with noncoding RNAs exerting regulatory influence. However, the molecular function of circular RNAs (circRNAs) in the progression from cardiac hypertrophy to heart failure remains unclear. To uncover functional circRNAs and identify the core circRNA signaling pathway in heart failure, we construct a global triple network (microRNA, circRNA, and mRNA) based on the competitive endogenous RNA (ceRNA) theory. We observe that cardiac hypertrophy related circRNA (circRNA CHRC), within the ceRNA network, is down-regulated in both transverse aortic constriction (TAC) mice and Ang-II--treated primary mouse cardiomyocytes. Silencing circRNA CHRC increases cross-sectional cell area, atrial natriuretic peptide, and ß-myosin heavy chain levels in primary mouse cardiomyocytes. Further screening reveals that circRNA CHRC targets the miR-431-5p/KLF15 axis implicated in heart failure progression in vivo and in vitro. Immunoprecipitation with anti-Ago2-RNA confirms the interaction between circRNA CHRC and miR-431-5p, while miR-431-5p mimics reverse Klf15 activation caused by circRNA CHRC overexpression. In summary, circRNA CHRC attenuates cardiac hypertrophy via sponging miR-431-5p to maintain the normal level of Klf15 expression.

15.
Int J Mol Med ; 53(2)2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-38063256

RESUMEN

The Kv11.1 potassium channel encoded by the Kcnh2 gene is crucial in conducting the rapid delayed rectifier K+ current in cardiomyocytes. Homozygous mutation in Kcnh2 is embryonically lethal in humans and mice. However, the molecular signaling pathway of intrauterine fetal loss is unclear. The present study generated a Kcnh2 knockout rat based on edited rat embryonic stem cells (rESCs). Kcnh2 knockout was embryonic lethal on day 11.5 of development due to a heart configuration defect. Experiments with human embryonic heart single cells (6.5­7 weeks post­conception) suggested that potassium voltage­gated channel subfamily H member 2 (KCNH2) plays a crucial role in the development of compact cardiomyocytes. By contrast, apoptosis was found to be triggered in the homozygous embryos, which could be attributed to the failure of KCNH2 to form a complex with integrin ß1 that was essential for preventing the process of apoptosis via inhibition of forkhead box O3A. Destruction of the KCNH2/integrin ß1 complex reduced the phosphorylation level of AKT and deactivated the glycogen synthase kinase 3 ß (GSK­3ß)/ß­catenin pathway, which caused early developmental abnormalities in rats. The present work reveals a basic mechanism by which KCNH2 maintains intact embryonic heart development.


Asunto(s)
Canal de Potasio ERG1 , Cardiopatías Congénitas , Animales , Femenino , Humanos , Ratones , Embarazo , Ratas , Desarrollo Embrionario , Canal de Potasio ERG1/genética , Canal de Potasio ERG1/metabolismo , Canales de Potasio Éter-A-Go-Go/genética , Canales de Potasio Éter-A-Go-Go/metabolismo , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Cardiopatías Congénitas/metabolismo , Integrina beta1/genética , Integrina beta1/metabolismo , Miocitos Cardíacos/metabolismo
16.
J Cell Mol Med ; 17(9): 1119-27, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23837875

RESUMEN

Sarcolemmal Na(+) /H(+) exchanger 1 (NHE1) activity is essential for the intracellular pH (pHi ) homeostasis in cardiac myocytes. Emerging evidence indicates that sarcolemmal NHE1 dysfunction was closely related to cardiomyocyte death, but it remains unclear whether defective trafficking of NHE1 plays a role in the vital cellular signalling processes. Dynamin (DNM), a large guanosine triphosphatase (GTPase), is best known for its roles in membrane trafficking events. Herein, using co-immunoprecipitation, cell surface biotinylation and confocal microscopy techniques, we investigated the potential regulation on cardiac NHE1 activity by DNM. We identified that DNM2, a cardiac isoform of DNM, directly binds to NHE1. Overexpression of a wild-type DNM2 or a dominant-negative DNM2 mutant with defective GTPase activity in adult rat ventricular myocytes (ARVMs) facilitated or retarded the internalization of sarcolemmal NHE1, whereby reducing or increasing its activity respectively. Importantly, the increased NHE1 activity associated with DNM2 deficiency led to ARVMs apoptosis, as demonstrated by cell viability, terminal deoxynucleotidyl transferase-mediated dUTP nick-end labelling assay, Bcl-1/Bax expression and caspase-3 activity, which were effectively rescued by pharmacological inhibition of NHE1 with zoniporide. Thus, our results demonstrate that disruption of the DNM2-dependent retrograde trafficking of NHE1 contributes to cardiomyocyte apoptosis.


Asunto(s)
Apoptosis , Dinamina II/metabolismo , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Intercambiadores de Sodio-Hidrógeno/metabolismo , Animales , Supervivencia Celular , Dinamina II/deficiencia , Células HEK293 , Ventrículos Cardíacos/citología , Humanos , Masculino , Transporte de Proteínas , Ratas , Ratas Sprague-Dawley , Sarcolema/metabolismo
17.
Am J Hum Genet ; 86(6): 872-80, 2010 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-20560207

RESUMEN

Congenital long QT syndrome (LQTS) is a hereditary disorder that leads to sudden cardiac death secondary to fatal cardiac arrhythmias. Although many genes for LQTS have been described, the etiology remains unknown in 30%-40% of cases. In the present study, a large Chinese family (four generations, 49 individuals) with autosomal-dominant LQTS was clinically evaluated. Genome-wide linkage analysis was performed by using polymorphic microsatellite markers to map the genetic locus, and positional candidate genes were screened by sequencing for mutations. The expression pattern and functional characteristics of the mutated protein were investigated by western blotting and patch-clamp electrophysiology. The genetic locus of the LQTS-associated gene was mapped to chromosome 11q23.3-24.3. A heterozygous mutation (Kir3.4-Gly387Arg) was identified in the G protein-coupled, inwardly rectifying potassium channel subunit Kir3.4, encoded by the KCNJ5 gene. The Kir3.4-Gly387Arg mutation was present in all nine affected family members and absent in 528 ethnically matched controls. Western blotting of human cardiac tissue demonstrated significant Kir3.4 expression levels in the cardiac ventricles. Heterologous expression studies with Kir3.4-Gly387Arg revealed a loss-of-function electrophysiological phenotype resulting from reduced plasma membrane expression. Our findings suggest a role for Kir3.4 in the etiology of LQTS.


Asunto(s)
Canales de Potasio Rectificados Internamente Asociados a la Proteína G/genética , Adolescente , Adulto , Anciano , Aminofilina , Atropina , Mapeo Cromosómico , Combinación de Medicamentos , Femenino , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/metabolismo , Ligamiento Genético , Humanos , Recién Nacido , Síndrome de QT Prolongado/genética , Masculino , Persona de Mediana Edad , Mutación , Miocardio/metabolismo , Nitroglicerina , Papaverina , Linaje , Fenobarbital
18.
Biochem Biophys Res Commun ; 439(4): 591-6, 2013 Oct 04.
Artículo en Inglés | MEDLINE | ID: mdl-24041700

RESUMEN

The cardiac transcription factor GATA4 is essential for cardiac development, and mutations in this gene have been implicated in a wide variety of congenital heart diseases in both animal models and humans. However, whether mutated GATA4 predisposes to dilated cardiomyopathy (DCM) remains unknown. In this study, the whole coding region and splice junction sites of the GATA4 gene was sequenced in 110 unrelated patients with idiopathic DCM. The available relatives of the index patient harboring an identified mutation and 200 unrelated ethnically matched healthy individuals used as controls were genotyped. The functional effect of the mutant GATA4 was characterized in contrast to its wild-type counterpart using a luciferase reporter assay system. As a result, a novel heterozygous GATA4 mutation, p.C271S, was identified in a family with DCM inherited as an autosomal dominant trait, which co-segregated with DCM in the family with complete penetrance. The missense mutation was absent in 400 control chromosomes and the altered amino acid was completely conserved evolutionarily among species. Functional analysis demonstrated that the GATA4 mutant was associated with significantly decreased transcriptional activity and remarkably reduced synergistic activation between GATA4 and NKX2-5, another transcription factor crucial for cardiogenesis. The findings provide novel insight into the molecular mechanisms involved in the pathogenesis of DCM, suggesting the potential implications in the prenatal diagnosis and gene-specific treatment for this common form of myocardial disorder.


Asunto(s)
Cardiomiopatía Dilatada/genética , Factor de Transcripción GATA4/genética , Mutación , Adulto , Femenino , Factor de Transcripción GATA4/metabolismo , Predisposición Genética a la Enfermedad , Proteína Homeótica Nkx-2.5 , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
19.
ScientificWorldJournal ; 2013: 393570, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23476131

RESUMEN

Microarray data are high dimension with high noise ratio and relatively small sample size, which makes it a challenge to use microarray data to identify candidate disease genes. Here, we have presented a hybrid method that combines estimation of distribution algorithm with support vector machine for selection of key feature genes. We have benchmarked the method using the microarray data of both diffuse B cell lymphoma and colon cancer to demonstrate its performance for identifying key features from the profile data of high-dimension gene expression. The method was compared with a probabilistic model based on genetic algorithm and another hybrid method based on both genetics algorithm and support vector machine. The results showed that the proposed method provides new computational strategy for hunting candidate disease genes from the profile data of disease gene expression. The selected candidate disease genes may help to improve the diagnosis and treatment for diseases.


Asunto(s)
Algoritmos , Biología Computacional/métodos , Genes Relacionados con las Neoplasias , Linfoma de Células B Grandes Difuso/diagnóstico , Máquina de Vectores de Soporte , Neoplasias del Colon/diagnóstico , Neoplasias del Colon/genética , Regulación Neoplásica de la Expresión Génica , Genoma Humano , Humanos , Linfoma de Células B Grandes Difuso/genética , Modelos Estadísticos , Análisis de Secuencia por Matrices de Oligonucleótidos , Sensibilidad y Especificidad , Transcriptoma
20.
Biomed Pharmacother ; 165: 115275, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37541173

RESUMEN

BACKGROUND: Pathological cardiac hypertrophy is a hallmark of various cardiovascular diseases (CVD) including chronic heart failure (HF) and an important target for the treatment of these diseases. Aberrant activation of Angiotensin II (Ang II)/AT1R signaling pathway is one of the main triggers of cardiac hypertrophy, which further gives rise to excessive inflammation that is mediated by the key transcription factor NF-κB. Resveratrol (REV) is a natural polyphenol with multiple anti-inflammatory and anti-oxidative effects, however the ability of REV in preventing Ang II-induced cardiac hypertrophy in combination with NF-κB signaling activation remains unclear. METHODS: Murine models of cardiac hypertrophy was conducted via implantation of Ang II osmotic pumps. Primary neonatal rat cardiomyocyte and heart tissues were examined to determine the effect and underlying mechanism of REV in preventing Ang II-induced cardiac hypertrophy. RESULTS: Administrations of REV significantly prevented Ang II-induced cardiac hypertrophy, as well as robustly attenuated Ang II-induced cardiac fibrosis, and cardiac dysfunction. Furthermore, REV not only directly prevented Ang II/AT1R signal transductions, but also prevented Ang II-induced expressions of pro-inflammatory cytokines and activation of NF-κB signaling pathway. CONCLUSIONS: Our study provides important new mechanistic insight into the cardioprotective effects of REV in preventing Ang II-induced cardiac hypertrophy via inhibiting adverse NF-κB signaling activation. Our findings further suggest the therapeutic potential of REV as a promising drug for the treatment of cardiac hypertrophy and heart failure.


Asunto(s)
Insuficiencia Cardíaca , FN-kappa B , Ratas , Ratones , Animales , FN-kappa B/metabolismo , Resveratrol/efectos adversos , Angiotensina II/farmacología , Transducción de Señal , Cardiomegalia/inducido químicamente , Cardiomegalia/tratamiento farmacológico , Cardiomegalia/metabolismo , Miocitos Cardíacos , Insuficiencia Cardíaca/inducido químicamente , Insuficiencia Cardíaca/tratamiento farmacológico , Insuficiencia Cardíaca/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA