Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Cell Mol Biol (Noisy-le-grand) ; 70(6): 108-113, 2024 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-38836673

RESUMEN

Oridonin belongs to a small molecule from the Chinese herb Rabdosia rubescens with potent anticancer activity. In spite of the lncRNA AFAP1-AS1 has been proven to exert promoting function in lung cancer, its relationship with oridonin in lung cancer is obscure. Therefore, our study planned to explore the potential of oridonin in lung cancer as well as unveil the regulatory mechanism of oridonin on AFAP1-AS1 in lung cancer cells. In the present study, oridonin inhibited lung cancer cell proliferation, migration, as well as invasion, as evidenced by MTT, wound healing, as well as transwell assays. Besides, we observed that oridonin could downregulate AFAP1-AS1 expression, and overexpressed AFAP1-AS1 could reverse the repressive effects of oridonin on lung cancer cell proliferation, migration, as well as invasion. More importantly, we found that AFAP1-AS1 could bind to IGF2BP1 through starBase prediction and RIP assay. The expression level of IGF2BP1 was also reduced by oridonin treatment but reversed after AFAP1-AS1 overexpression. Additionally, we proved that overexpressed IGF2BP1 could reverse the repressive impacts of oridonin on lung cancer cell proliferation, migration, as well as invasion. Further, in vivo experiments validated the repressive role of oridonin on tumor growth of lung cancer. Together, oridonin inhibits lung cancer cell proliferation as well as migration by modulating AFAP1-AS1/IGF2BP1, and AFAP1-AS1/IGF2BP1 possesses the potential to be a promising therapy targeting for lung cancer, especially in oridonin treatment.


Asunto(s)
Movimiento Celular , Proliferación Celular , Diterpenos de Tipo Kaurano , Regulación Neoplásica de la Expresión Génica , Neoplasias Pulmonares , ARN Largo no Codificante , Proteínas de Unión al ARN , Diterpenos de Tipo Kaurano/farmacología , Humanos , Proliferación Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Proteínas de Unión al ARN/metabolismo , Proteínas de Unión al ARN/genética , Animales , Ratones Desnudos , Ratones , Ratones Endogámicos BALB C , Invasividad Neoplásica , Células A549
2.
Bioengineered ; 13(3): 5305-5317, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35156897

RESUMEN

Airway inflammation is one of the typical pathological characteristics of asthma. MicroRNAs (miRNAs) play important roles in regulating inflammation. Nevertheless, miRNA-885-3p (miR-885-3p)'s role in asthmatic inflammation and the underlying mechanism need to be explained. In this work, miR-885-3p expression and toll-like receptor 4 (TLR4) expression in asthma patients' plasma and lipopolysaccharide (LPS)-treated 16HBE cells were detected through quantitative real-time PCR. The interleukin-8 (IL-8), tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6) levels in 16HBE cell supernatant were examined via enzyme-linked immunosorbent assay. Cell counting kit-8 (CCK-8) assay and flow cytometry were employed to examine 16HBE cell viability and apoptosis, respectively. Western blotting was performed to examine the expression of TLR4, cleaved caspase-3, B-cell lymphoma-2 (Bcl-2), nuclear factor-kappa B (NF-κB) p65, Bcl-2-related X protein (Bax), phosphorylated (p)-NF-κB p65 and myeloid differentiation primitive-response protein 88 (MyD88) in 16HBE cells. Furthermore, the targeted relationship between TLR4 and miR-885-3p in 16HBE cells was determined through dual-luciferase reporter gene assay. Compared with healthy volunteers, miR-885-3p expression in acute asthma patients' plasma was significantly downregulated. In 16HBE cells, the stimulation of LPS reduced miR-885-3p expression. MiR-885-3p overexpression reduced LPS-stimulated 16HBE cell injury by enhancing cell viability, and suppressing the levels of inflammatory factors and apoptosis. Furthermore, TLR4 was identified as miR-885-3p's target gene. TLR4 overexpression weakened the impacts of miR-885-3p on LPS-stimulated cell injury and NF-κB-MyD88 signaling. In conclusion, miR-885-3p can reduce LPS-induced 16HBE cell damage, via targeting TLR4 to suppress the NF-κB-MyD88 pathway.


Asunto(s)
Asma , MicroARNs , Apoptosis , Células Epiteliales/metabolismo , Células Epiteliales/patología , Humanos , Inflamación/patología , Interleucina-6/metabolismo , Lipopolisacáridos/farmacología , MicroARNs/genética , MicroARNs/metabolismo , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/metabolismo , FN-kappa B/genética , FN-kappa B/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo
3.
Medicine (Baltimore) ; 100(31): e26439, 2021 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-34397793

RESUMEN

ABSTRACT: This study aimed to investigate the expression and clinical significance of aurora B kinase (AURKB) gene in lung adenocarcinoma (LUAD) by collecting relevant data in Oncomine database.Firstly, mRNA expression level of AURKB in LUAD was systematically analyzed using the ONCOMINE and the cancer genome atlas databases. Then, the association between AURKB expression and clinical parameters was investigated by UALCAN. The Kaplan-Meier Plotter was used to assess the prognostic significance of AURKB.Pooled analysis showed that AURKB was frequently up-regulated expression in LUAD. In addition, immunohistochemistry showed that AURKB was highly expressed in lung adenocarcinoma tissues, while it was weakly expressed in normal tissues. Subsequently, AURKB expression was identified to be negatively associated with Overall survival (P < 1e-16), post-progression survival (P = .017), first progression (P = 9.8e-09).This study confirms that increased expression of AURKB in LUAD is associated with poor prognosis, suggesting that AURKB might be used as a promising prognostic biomarker and novel therapeutic target for LUAD.


Asunto(s)
Adenocarcinoma/genética , Aurora Quinasa B/genética , Neoplasias Pulmonares/genética , Adenocarcinoma/metabolismo , Adenocarcinoma/secundario , Anciano , Aurora Quinasa B/metabolismo , Biología Computacional , Minería de Datos , Bases de Datos Genéticas , Progresión de la Enfermedad , Femenino , Expresión Génica , Humanos , Estimación de Kaplan-Meier , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Pronóstico , ARN Mensajero/metabolismo , Tasa de Supervivencia , Regulación hacia Arriba
4.
J Clin Lab Anal ; 35(7): e23857, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-34097306

RESUMEN

BACKGROUND: Smoking is likely to facilitate airway inflammation and finally contributes to chronic obstructive pulmonary disease (COPD). This investigation was intended to elucidate miRNAs that were involved in smoking-induced COPD. METHODS: Altogether 155 COPD patients and 77 healthy volunteers were recruited, and their serum levels of miR-221-3p and miR-92a-3p were determined. Besides, human bronchial epithelial cells (16HBECs) were purchased, and they were treated by varying concentrations of cigarette smoke extract (CSE). The 16HBECs were, additionally, transfected by miR-221-3p mimic, miR-92a-3p mimic, miR-221-3p inhibitor or miR-92a-3p inhibitor, and cytokines released by them, including TNF-α, IL-8, IL-1ß, and TGF-ß1, were monitored using enzyme linked immunosorbent assay (ELISA) kits. RESULTS: Chronic obstructive pulmonary disease patients possessed higher serum levels of miR-221-3p and miR-92a-3p than healthy volunteers (p < 0.05), and both miR-221-3p and miR-92a-3p were effective biomarkers in diagnosing stable COPD from acute exacerbation COPD. Moreover, viability of 16HBECs was undermined by CSE treatment (p < 0.05), and exposure to CSE facilitated 16HBECs' release of TNF-α, IL-8, IL-1ß, and TGF-ß1 (p < 0.05). Furthermore, miR-221-3p/miR-92a-3p expression in 16HBECs was significantly suppressed after transfection of miR-221-3p/miR-92a-3p inhibitor (p < 0.05), which abated CSE-triggered increase in cytokine production and decline in viability of 16HBECs (p < 0.05). CONCLUSION: MiR-221-3p and miR-92a-3p were involved in CSE-induced hyperinflammation of COPD, suggesting that they were favorable alternatives in diagnosing COPD patients with smoking history.


Asunto(s)
Inflamación/genética , MicroARNs/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/genética , Fumar/genética , Anciano , Remodelación de las Vías Aéreas (Respiratorias)/genética , Apoptosis/genética , Bronquios/patología , Estudios de Casos y Controles , Línea Celular , Proliferación Celular/genética , Supervivencia Celular/genética , Citocinas/metabolismo , Células Epiteliales/metabolismo , Femenino , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Humanos , Masculino , MicroARNs/genética , Persona de Mediana Edad , Enfermedad Pulmonar Obstructiva Crónica/diagnóstico , Enfermedad Pulmonar Obstructiva Crónica/patología , Enfermedad Pulmonar Obstructiva Crónica/fisiopatología
5.
Mol Med Rep ; 22(6): 4675-4684, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33173954

RESUMEN

As a single cardiac malformation, ventricular septal defect (VSD) is the most common form of congenital heart disease. However, the precise molecular mechanisms underlying VSD are not completely understood. Numerous microRNAs (miRs/miRNAs) are associated with ventricular septal defects. miR-29c inhibits the proliferation and promotes the apoptosis and differentiation of P19 embryonal carcinoma cells, possibly via suppressing Wnt4 signaling. However, to the best of our knowledge, no in vivo studies have been published to determine whether overexpression of miR-29c leads to developmental abnormalities. The present study was designed to observe the effect of miRNA-29c on cardiac development and its possible mechanism in vivo. Zebrafish embryos were microinjected with different doses (1, 1.6 and 2 µmol) miR-29c mimics or negative controls, and hatchability, mortality and cardiac malformation were subsequently observed. The results showed that in zebrafish embryos, miR-29c overexpression attenuated heart development in a dose-dependent manner, manifested by heart rate slowdown, pericardial edema and heart looping disorder. Further experiments showed that overexpression of miR-29c was associated with the Wnt4/β-catenin signaling pathway to regulate zebrafish embryonic heart development. In conclusion, the present results demonstrated that miR-29c regulated the lateral development and cardiac circulation of zebrafish embryo by targeting Wnt4.


Asunto(s)
Defectos del Tabique Interventricular/metabolismo , MicroARNs/metabolismo , Proteína Wnt4/metabolismo , Animales , Apoptosis/genética , Diferenciación Celular/genética , Línea Celular Tumoral , Proliferación Celular/genética , Células Madre de Carcinoma Embrionario/metabolismo , Corazón/embriología , Defectos del Tabique Interventricular/genética , MicroARNs/genética , Transducción de Señal/genética , Proteína Wnt4/genética , Pez Cebra/embriología , Pez Cebra/genética , Proteínas de Pez Cebra/genética
6.
Biochem Biophys Res Commun ; 519(4): 682-688, 2019 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-31543343

RESUMEN

OBJECTIVE: -microRNAs (miRNAs) have emerged as novel regulators for cardiac hypertrophy. MiR-122 is well recognized as a promising therapeutic target in liver disease, whereas recently plays important roles in cardiovascular diseases. The current study aimed to explore the effect of miR-122 on the pathogenesis of cardiomyocyte hypertrophy. METHODS AND RESULTS: -The cardiomyocytes isolated from the neonatal rat ventricular cardiomyocytes (NRVMs) were collected and performed to Angiotensin II (Ang II) administration. We observed a dramatically increased miR-122 expression in hypertrophic cardiomyocytes. The NRVMs transfected with miR-122 mimic or negative control were utilized for the functional analysis. Overexpression of miR-122 increased the morphology size of cardiomyocytes and promoted the pro-hypertrophic genes expression, whereas downregulated the anti-hypertrophic genes upon Ang II stimulation. The bioinformatics analysis and luciferase reporter assays exhibited that miR-122 directly targeted FoxO3 and attenuated its gene level in hypertrophic cardiomyocytes. Moreover, miR-122 negatively regulated FoxO3 but promoted calcineurin signaling pathway activation. Importantly, FoxO3 overexpression significantly reversed the effect of miR-122 on cardiomyocyte hypertrophy. CONCLUSION: -Collected, our finding demonstrated that miR-122 accelerated the development of cardiomyocytes hypertrophy partially via directly regulation of FoxO3-calcineurin pathway.


Asunto(s)
Cardiomegalia/genética , Proteína Forkhead Box O3/genética , Regulación de la Expresión Génica/genética , MicroARNs/genética , Miocitos Cardíacos/metabolismo , Angiotensina II/farmacología , Animales , Animales Recién Nacidos , Calcineurina/metabolismo , Cardiomegalia/metabolismo , Cardiomegalia/patología , Tamaño de la Célula/efectos de los fármacos , Células Cultivadas , Proteína Forkhead Box O3/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Miocitos Cardíacos/citología , Miocitos Cardíacos/efectos de los fármacos , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Vasoconstrictores/farmacología
7.
Curr Stem Cell Res Ther ; 14(3): 230-238, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30047336

RESUMEN

BACKGROUND: Characterization of the fate changes of stem cells is essential to understand the roles of certain stem cells both during development and in diseases, such as cancer. In the past two decades, more and more importance has been paid to the studies of in vivo lineage tracing, because they could authentically reveal the differentiation, migration and even proliferation of stem cells. However, specific genetic tools have only been developed until recently. OBJECTIVE: To summarize the progresses of genetic tools for specific lineage tracing with emphasis on their applications in investigating the stem cell niche signals. RESULTS: Three major genetic strategies have been reviewed according to the development of technique, particularly the advantages and disadvantages of individual methods. CONCLUSION: In vivo specific lineage tracing of stem cells could be achieved by comprehensive application of multiple genetic tools.


Asunto(s)
Linaje de la Célula/genética , Rastreo Celular/métodos , Genes Reporteros , Nicho de Células Madre/genética , Células Madre/citología , Transgenes , Animales , Cromosomas Artificiales Bacterianos , ADN Nucleotidiltransferasas/genética , ADN Nucleotidiltransferasas/metabolismo , Embrión de Mamíferos , Técnicas de Sustitución del Gen , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Integrasas/genética , Integrasas/metabolismo , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Ratones Transgénicos , Células Madre/metabolismo , Proteína Fluorescente Roja
8.
Sci Rep ; 7(1): 14108, 2017 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-29074979

RESUMEN

A major issue in thermochronology and U-Th-Pb dating is the effect of radiation damage, created by α-recoils from α-decay events, on the diffusion of radiogenic elements (e.g., He and Pb) in host mineral. Up until now, thermal events have been considered as the only source of energy for the recovery of radiation-damage. However, irradiation, such as from the α-particle of the α-decay event, can itself induce damage recovery. Quantification of radiation-induced recovery caused by α-particles during α-decay events has not been possible, as the recovery process at the atomic-scale has been difficult to observe. Here we present details of the dynamics of the amorphous-to-crystalline transition process during α-particle irradiations using in situ transmission electron microscopy (TEM) and consecutive ion-irradiations: 1 MeV Kr2+ (simulating α-recoil damage), followed by 400 keV He+ (simulating α-particle annealing). Upon the He+ irradiation, partial recrystallization of the original, fully-amorphous Durango apatite was clearly evident and quantified based on the gradual appearance of new crystalline domains in TEM images and new diffraction maxima in selected area electron diffraction patterns. Thus, α-particle induced annealing occurs and must be considered in models of α-decay event damage and its effect on the diffusion of radiogenic elements in geochronology and thermochronology.

9.
Mol Med Rep ; 13(3): 2527-35, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26848028

RESUMEN

In our previous study, the upregulation of microRNA (miR)-29c was identified in the mother of a fetus with a congenital heart defect. However, the functional and regulatory mechanisms of miR­29c in the development of the heart remain to be elucidated. In the present study, the role and mechanism of miR­29c inhibition in heart development were investigated in an embryonic carcinoma cell model. Inhibition of miR­29c promoted proliferation, and suppressed the apoptosis and differentiation of P19 cells. It was also demonstrated that Wingless­related MMTV integration site 4 (Wnt4) was a target of miR­29c, determined using bioinformatic analysis combined with luciferase assays. The inhibition of miR­29c stimulated the WNT4/ß­catenin pathway, promoting proliferation of the P19 cells, but suppressing their differentiation into cardiomyocytes. Furthermore, the inhibition of miR­29c promoted the expression of B cell lymphoma­2 and inhibited cell apoptosis. These results demonstrate the significance of miR­29c in the process of cardiac development and suggest that miR-29c dysregulation may be associated with the occurrence of CHD. Thus, miR-29c may have therapeutic potential in the future.


Asunto(s)
Apoptosis , MicroARNs/genética , Animales , Diferenciación Celular , Línea Celular Tumoral , Proliferación Celular , Ratones , MicroARNs/metabolismo , Interferencia de ARN , Proteína Wnt4/genética , Proteína Wnt4/metabolismo
10.
Gene ; 576(1 Pt 2): 304-11, 2016 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-26484393

RESUMEN

Compared to healthy controls, microRNA-29c (miR-29c) is highly expressed in the heart during progression towards ventricular septal defect. However, studies on miR-29c function in heart development are scarce. We investigated the role of miR-29c in P19 cell proliferation, apoptosis, and differentiation and the underlying mechanisms. We evaluated proliferation and cell cycle progression, detected morphological changes; apoptosis rate; BAX, BCL2, GATA binding protein 4 (GATA4), cardiac troponin T (cTnT), and myocyte enhancer factor 2C (MEF2C) expression; and caspase-3, -8, and -9 activity in miR-29c-overexpressing P19 cells, and investigated whether WNT4 was a miR-29c target. MiR-29c-overexpressing cells had decreased proliferation, increased G1 cells, and significantly higher apoptotic rate than the controls. Expression of the apoptosis-related BAX and BCL2 genes and caspase-3, -8, and -9 activity were significantly increased in miR-29c-overexpressing cells. Expression of the cardiac-specific markers GATA4, cTnT, and MEF2C revealed promoted differentiation in miR-29c-overexpressing cells compared to the controls. Luciferase assay confirmed that WNT4 is a miR-29c target. Wnt4 and ß-catenin expression was decreased in miR-29c-overexpressing cells. MiR-29c inhibits P19 cell proliferation and promotes apoptosis and differentiation, possibly by suppressing Wnt4 signaling, whose deregulation contributes to congenital heart disease development.


Asunto(s)
Células Madre de Carcinoma Embrionario/patología , MicroARNs/genética , Proteína Wnt4/metabolismo , Regiones no Traducidas 3' , Animales , Apoptosis/genética , Diferenciación Celular/genética , Proliferación Celular/genética , Factor de Transcripción GATA4/genética , Regulación del Desarrollo de la Expresión Génica , Corazón/embriología , Factores de Transcripción MEF2/genética , Ratones , Transducción de Señal/genética , Sincalida/genética , Sincalida/metabolismo , Proteína Wnt4/genética
11.
Cell Physiol Biochem ; 35(5): 1975-85, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25871431

RESUMEN

BACKGROUND/AIMS: PID1 was originally described as an insulin sensitivity relevance protein, which is also highly expressed in heart tissue. However, its function in the heart is still to be elucidated. Thus this study aimed to investigate the role of PID1 in the heart in response to hypertrophic stimuli. METHODS: Samples of human failing hearts from the left ventricles of dilated cardiomyopathy (DCM) patients undergoing heart transplants were collected. Transgenic mice with cardiomyocyte-specific overexpression of PID1 were generated, and cardiac hypertrophy was induced by transverse aortic constriction (TAC). The extent of cardiac hypertrophy was evaluated by echocardiography as well as pathological and molecular analyses of heart samples. RESULTS: A significant increase in PID1 expression was observed in failing human hearts and TAC-treated wild-type mouse hearts. When compared with TAC-treated wild-type mouse hearts, PID1-TG mouse showed a significant exacerbation of cardiac hypertrophy, fibrosis, and dysfunction. Further analysis of the signaling pathway in vivo suggested that these adverse effects of PID1 were associated with the inhibition of AKT, and activation of MAPK pathway. CONCLUSION: Under pathological conditions, over-expression of PID1 promotes cardiac hypertrophy by regulating the Akt and MAPK pathway.


Asunto(s)
Cardiomegalia/patología , Proteínas Portadoras/metabolismo , Animales , Cardiomegalia/diagnóstico por imagen , Cardiomegalia/etiología , Proteínas Portadoras/genética , Modelos Animales de Enfermedad , Insuficiencia Cardíaca/diagnóstico por imagen , Insuficiencia Cardíaca/patología , Ventrículos Cardíacos/metabolismo , Ventrículos Cardíacos/patología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Miocardio/metabolismo , Miocardio/patología , Cadenas Pesadas de Miosina/genética , Presión , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal , Ultrasonografía , Regulación hacia Arriba
12.
Cell Biochem Biophys ; 71(3): 1463-73, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25388842

RESUMEN

Induced pluripotent stem cells (iPSc) hold significant promise for the development of cardiac regenerative therapy for myocardial infarction (MI). However, preclinical optimization and validation of large-animal models will be required before iPSc used clinically. Therefore, we aim to investigate the therapeutic potential of iPSc transplantation for MI and relative mechanisms in a post-infarcted swine model. Left anterior descending coronary artery was balloon-occluded after percutaneous transluminal angiography to generate MI (60-min no-flow ischemia). Animals were then divided into Sham, PBS control, and iPS experimental groups. The cardiac function and LV structural were assessed by dual-source computed tomography. Terminal deoxynucleotidyl nick end labeling, histology, and immunofluorescence were used to examine the effect of transplanted iPS cells on apoptosis, fibrosis, and hypertrophy. At 6 weeks, LV structural abnormality and cardiac dysfunction were less pronounced in iPSc group than in PBS group, and these improvements were accompanied by reduction of scar size. iPSc transplantation was associated with significant increase of vascular density and reduced myocardial apoptosis in the border zone of infarction, which was accompanied by the reduction in fibrosis degree. Moreover, proangiogenic and antiapoptotic factors were increased significantly in iPS group compared with PBS group. Cardiomyocyte hypertrophy was significantly attenuated by iPSc transplantation. In conclusion, these results suggested that transplantation of iPSc may result in functional recovery by promoting angiogenesis, inhibiting apoptosis, and ameliorating cardiac remodeling. This proof of concept study may provide a basis for an autologous iPSc-based therapy of MI.


Asunto(s)
Corazón/fisiopatología , Células Madre Pluripotentes Inducidas/trasplante , Infarto del Miocardio/fisiopatología , Infarto del Miocardio/cirugía , Neovascularización Fisiológica , Trasplante de Células Madre , Remodelación Ventricular , Animales , Apoptosis , Línea Celular , Supervivencia Celular , Citoprotección , Modelos Animales de Enfermedad , Fibrosis , Regulación de la Expresión Génica , Hipertrofia , Células Madre Pluripotentes Inducidas/citología , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Recuperación de la Función , Porcinos , Factor A de Crecimiento Endotelial Vascular/metabolismo
13.
J Cell Biochem ; 115(3): 531-9, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24122925

RESUMEN

Neural remodeling after myocardial infarction (MI) may cause malignant ventricular arrhythmia, which is the main cause of sudden cardiac death following MI. Herein, we aimed to examine whether induced pluripotent stem cells (iPSc) transplantation can ameliorate neural remodeling and reduce ventricular arrhythmias (VA) in a post-infarcted swine model. Left anterior descending coronary arteries were balloon-occluded to generate MI. Animals were then divided into Sham, PBS control, and iPS groups. Dynamic electrocardiography programmed electric stimulation were performed to evaluate VA. The spatial distribution of vascularization, Cx43 and autonomic nerve regeneration were evaluated by immunofluorescence staining. Associated protein expression was detected by Western blotting. Likewise, we measured the enzymatic activities of superoxide dismutase and content of malondialdehyde. Six weeks later, the number of blood vessels increased significantly in the iPSc group. The expression of vascular endothelial growth factor and connexin 43 in the iPS group was significantly higher than the PBS group; however, the levels of nerve growth factor and tyrosine hydroxylase were lower. The oxidative stress was ameliorated by iPSc transplantation. Moreover, the number of sympathetic nerves in the iPSc group was reduced, while the parasympathetic nerve fibers had no obvious change. The transplantation of iPSc also significantly decreased the low-/high-frequency ratio and arrhythmia score of programmed electric stimulation-induced VA. In conclusion, iPSc intramyocardial transplantation reduces vulnerability to VAs, and the mechanism was related to the remodeling amelioration of autonomic nerves and gap junctions. Moreover, possible mechanisms of iPSc transplantation in improving neural remodeling may be related to attenuated oxidative stress and inflammatory response.


Asunto(s)
Arritmias Cardíacas/terapia , Células Madre Pluripotentes Inducidas , Infarto del Miocardio/terapia , Trasplante de Células Madre , Animales , Arritmias Cardíacas/complicaciones , Arritmias Cardíacas/patología , Vasos Coronarios/patología , Modelos Animales de Enfermedad , Electrocardiografía , Humanos , Infarto del Miocardio/complicaciones , Infarto del Miocardio/patología , Regeneración Nerviosa , Estrés Oxidativo , Porcinos , Factor A de Crecimiento Endotelial Vascular
14.
Gene ; 533(2): 481-7, 2014 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-24140453

RESUMEN

Recent findings indicate that microRNAs (miRNAs) are involved in the regulatory network of adipogenesis and obesity. Thus far, only a few human miRNAs are known to function as adipogenic regulators, fanning interest in studies on the functional role of miRNAs during adipogenesis in humans. In a previous study, we used a microarray to assess miRNA expression during human preadipocyte differentiation. We found that expression of the miR-26b was increased in mature adipocytes. MiR-26b is an intronic miRNA located in the intron of CTDSP1 (carboxy terminal domain, RNA polymerase II, polypeptide A, small phosphatase 1). Target prediction and Renilla luciferase analyses revealed the phosphatase and tensin homolog gene (PTEN) as a putative target gene. In this study, we found that miR-26b was gradually upregulated during adipocyte differentiation. To understand the roles of miR-26b in adipogenesis, we adopted a loss-of-function approach to silence miR-26b stably in human preadipocytes. We found that miR-26b inhibition effectively suppressed adipocyte differentiation, as evidenced by decreased lipid droplets and the ability of miR-26b to decrease mRNA levels of adipocyte-specific molecular markers and triglyceride accumulation. Furthermore, the cell growth assay revealed that miR-26b inhibition promoted proliferation. Nevertheless, it had no effect on apoptosis. Taken together, these data indicate that miR-26b may be involved in adipogenesis and could be targeted for therapeutic intervention in obesity.


Asunto(s)
Adipocitos/citología , Adipogénesis/genética , Proliferación Celular , MicroARNs/fisiología , Apoptosis/genética , Células Cultivadas , Perfilación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Células HEK293 , Humanos , MicroARNs/antagonistas & inhibidores , Análisis por Micromatrices , Obesidad/genética , Obesidad/terapia
15.
Mol Med Rep ; 8(3): 806-12, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23846528

RESUMEN

Fatty acid binding protein 3 (FABP3, also termed heart-type fatty acid binding protein) is a member of the intracellular lipid-binding protein family that may be essential in fatty acid transport, cell growth, cellular signaling and gene transcription. Previously, we demonstrated that FABP3 was involved in apoptosis-associated congenital cardiac malformations; however, its mechanism of regulation remains unclear. Apoptosis has increasingly been considered to be important in cardiac development. In the present study, a zebrafish model was used to investigate the involvement of FABP3­morpholino (MO)-induced apoptosis and mitochondrial dysfunction in cardiac development. During the early stages of cardiac development, injection of FABP3­MO into zebrafish resulted in significant impairment in cardiac development and promoted the rate of apoptosis which was correlated with significant dysfunction of the mitochondria. For example, the ATP content was markedly decreased at 24 and 48 h post-fertilization (pf), reactive oxygen species production was significantly enhanced at 24 and 48 h pf and the mitochondrial DNA copy number was reduced at 24, 48 and 72 h pf. Additionally, Nkx2.5 expression was upregulated in FABP3-MO zebrafish, and Wnt signaling molecules (Wnt1, Wnt5 and Wnt11) were also highly expressed in FABP3-MO zebrafish at 24, 48 and 72 h pf. In conclusion, the results indicated that FABP3 knockdown exhibited significant toxic effects on cardiac development and mitochondrial function, which may be responsible for the knockdown of FABP3-induced apoptosis. Apoptosis was one of the mechanisms underlying this effect, and was correlated with the activation of Wnt signaling. These studies identified FABP3 as a candidate gene underlying the etiology of congenital heart defects.


Asunto(s)
Apoptosis/efectos de los fármacos , Proteínas de Unión a Ácidos Grasos/antagonistas & inhibidores , Mitocondrias/metabolismo , Morfolinos/toxicidad , Proteínas de Pez Cebra/antagonistas & inhibidores , Adenosina Trifosfato/metabolismo , Animales , Variaciones en el Número de Copia de ADN/efectos de los fármacos , ADN Mitocondrial/metabolismo , Embrión no Mamífero/metabolismo , Proteína 3 de Unión a Ácidos Grasos , Proteínas de Unión a Ácidos Grasos/metabolismo , Corazón/efectos de los fármacos , Corazón/crecimiento & desarrollo , Proteína Homeótica Nkx-2.5 , Larva/metabolismo , Mitocondrias/efectos de los fármacos , Mitocondrias/genética , Morfolinos/química , Especies Reactivas de Oxígeno/metabolismo , Factores de Transcripción/metabolismo , Proteínas Wnt/metabolismo , Vía de Señalización Wnt , Pez Cebra/crecimiento & desarrollo , Pez Cebra/metabolismo , Proteínas de Pez Cebra/metabolismo
16.
Cell Biochem Biophys ; 66(3): 489-97, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23274913

RESUMEN

NYGGF4, also known as phosphotyrosine interaction domain containing 1(PID1), is a recently discovered gene which is involved in obesity-related insulin resistance (IR) and mitochondrial dysfunction. We aimed to further elucidate the effects and mechanisms underlying NYGGF4-induced IR by investigating the effect of overexpressing mitochondrial transcription factor A (TFAM), which is essential for mitochondrial DNA transcription and replication, on NYGGF4-induced IR and mitochondrial abnormalities in 3T3-L1 adipocytes. Overexpression of TFAM increased the mitochondrial copy number and ATP content in both control 3T3-L1 adipocytes and NYGGF4-overexpressing adipocytes. Reactive oxygen species (ROS) production was enhanced in NYGGF4-overexpressing adipocytes and reduced in TFAM-overexpressing adipocytes; co-overexpression of TFAM significantly attenuated ROS production in NYGGF4-overexpressing adipocytes. However, overexpression of TFAM did not affect the mitochondrial transmembrane potential (ΔΨm) in control 3T3-L1 adipocytes or NYGGF4-overexpressing adipocytes. In addition, co-overexpression of TFAM-enhanced insulin-stimulated glucose uptake by increasing Glucose transporter type 4 (GLUT4) translocation to the PM in NYGGF4-overexpressing adipocytes. Overexpression of NYGGF4 significantly inhibited tyrosine phosphorylation of Insulin receptor substrate 1 (IRS-1) and serine phosphorylation of Akt, whereas overexpression of TFAM strongly induced phosphorylation of IRS-1 and Akt in NYGGF4-overexpressing adipocytes. This study demonstrates that NYGGF4 plays a role in IR by impairing mitochondrial function, and that overexpression of TFAM can restore mitochondrial function to normal levels in NYGGF4-overexpressing adipocytes via activation of the IRS-1/PI3K/Akt signaling pathway.


Asunto(s)
Adipocitos/metabolismo , Adipocitos/patología , Proteínas Portadoras/genética , Proteínas de Unión al ADN/genética , Resistencia a la Insulina , Mitocondrias/patología , Proteínas Mitocondriales/genética , Factores de Transcripción/genética , Células 3T3-L1 , Adenosina Trifosfato/metabolismo , Adipocitos/efectos de los fármacos , Animales , Proteínas Portadoras/metabolismo , Proteínas de Unión al ADN/metabolismo , Expresión Génica , Glucosa/metabolismo , Transportador de Glucosa de Tipo 4/metabolismo , Insulina/metabolismo , Insulina/farmacología , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Tamaño Mitocondrial/efectos de los fármacos , Fosforilación/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Factores de Transcripción/metabolismo
17.
Cell Biochem Biophys ; 66(1): 139-46, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23097025

RESUMEN

Fatty acid-binding protein 3 (FABP3) facilitates the movement of fatty acids in cardiac muscle. Previously, we reported that FABP3 is highly upregulated in the myocardium of ventricular septal defect patients and overexpression of FABP3 inhibited proliferation and promoted apoptosis in embryonic carcinoma cells (P19 cells). In this study, we aimed to investigate the effect of FABP3 gene silencing on P19 cell differentiation, proliferation and apoptosis. We used RNA interference and a lentiviral-based vector system to create a stable FABP3-silenced P19 cell line; knockdown of FABP3 was confirmed by quantitative real-time PCR. Expression analysis of specific differentiation marker genes using quantitative real-time PCR and observation of morphological changes using an inverted microscope revealed that knockdown of FABP3 did not significantly affect the differentiation of P19 cells into cardiomyocytes. CCK-8 proliferation assays and cell cycle analysis demonstrated that FABP3 gene silencing significantly inhibited P19 cell proliferation. Furthermore, Annexin V-FITC/propidium iodide staining and the caspase-3 activity assay revealed that FABP3 gene silencing significantly promoted serum starvation-induced apoptosis in P19 cells. In agreement with our previous research, these results demonstrate that FABP3 may play an important role during embryonic heart development, and that either overexpression or silencing of FABP3 will lead to an imbalance between proliferation and apoptosis, which may result in embryonic cardiac malformations.


Asunto(s)
Apoptosis , Proliferación Celular , Células Madre de Carcinoma Embrionario/patología , Proteínas de Unión a Ácidos Grasos/metabolismo , Regulación del Desarrollo de la Expresión Génica , Silenciador del Gen , Animales , Caspasa 3/genética , Caspasa 3/metabolismo , Diferenciación Celular , Forma de la Célula , Supervivencia Celular/efectos de los fármacos , Medio de Cultivo Libre de Suero , Embrión de Mamíferos/metabolismo , Células Madre de Carcinoma Embrionario/metabolismo , Proteína 3 de Unión a Ácidos Grasos , Proteínas de Unión a Ácidos Grasos/genética , Corazón/embriología , Ratones , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología
18.
J Bioenerg Biomembr ; 44(3): 317-23, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22528395

RESUMEN

Fatty acid binding protein 3 (FABP3) (also known as H-FABP) is a member of the intracellular lipid-binding protein family, and is mainly expressed in cardiac muscle tissue. The in vivo function of FABP3 is proposed to be in fatty acid metabolism, trafficking, and cell signaling. Our previous study found that FABP3 is highly regulated in patients with ventricular septal defect (VSD), and may play a significant role in the development of human VSD. In the present study, we aimed to investigate the impact of FABP3 knockdown by RNA interference (RNAi) on apoptosis and mitochondrial function of embryonic carcinoma (P19) cells. The results revealed that downregulated FABP3 expression promoted apoptosis, and resulted in mitochondrial deformation, increased mitochondrial membrane potential (MMP), and decreased intracellular ATP synthesis. In addition, the knockdown of FABP3 also led to excess intracellular ROS production. However, there was no obvious influence on the amount of mitochondrial DNA. Collectively, our results indicated that FABP3 knockdown promoted apoptosis and caused mitochondrial dysfunction in P19 cells, which might be responsible for the development of human VSD.


Asunto(s)
Apoptosis/fisiología , Células Madre de Carcinoma Embrionario/metabolismo , Células Madre de Carcinoma Embrionario/patología , Proteínas de Unión a Ácidos Grasos/deficiencia , Mitocondrias/metabolismo , Adenosina Trifosfato/biosíntesis , Animales , Diferenciación Celular/fisiología , ADN Mitocondrial/genética , Proteína 3 de Unión a Ácidos Grasos , Proteínas de Unión a Ácidos Grasos/genética , Proteínas de Unión a Ácidos Grasos/metabolismo , Dosificación de Gen , Técnicas de Silenciamiento del Gen , Ratones , Microscopía Electrónica , Mitocondrias/genética , Interferencia de ARN , ARN Mensajero/genética , ARN Mensajero/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA