Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Medicine (Baltimore) ; 101(46): e29328, 2022 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-36401368

RESUMEN

Metastasis is the major cause of death and failure of cancer chemotherapy in patients with breast cancer (BC). Activation of TGF-ß/lncRNA-MALAT1/miR-200c has been reported to play an essential role during the metastasis of BC cells. The present study aimed to validate the suppression of BC-cell migration and invasion by baicalin and explore its regulatory effects on the TGF-ß/lncRNA-MALAT1/miR-200c signaling pathway. We found that baicalin treatment inhibited cell viability and migration and invasion. Mechanistically, baicalin treatment significantly downregulated the expression of TGF-ß, ZEB1, and N-cadherin and upregulated E-cadherin on both mRNA and protein levels. Additionally, baicalin treatment significantly downregulated the expression of lncRNA-MALAT1 and upregulated that of miR-200c. Collectively, baicalin significantly suppresses cell viability, migration, and invasion of BC cells possibly by regulating the TGF-ß/lncRNA-MALAT1/miR-200c pathway.


Asunto(s)
Neoplasias de la Mama , MicroARNs , ARN Largo no Codificante , Humanos , Femenino , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Línea Celular Tumoral , Factor de Crecimiento Transformador beta , Transducción de Señal/genética
2.
Aging (Albany NY) ; 14(3): 1307-1320, 2022 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-35113040

RESUMEN

AIM: Uterine leiomyoma is the most common benign tumor of female genitalia, and the incidence is rising gradually. This study explores the mechanism of miR-29 and STAT3 signaling pathways on uterine leiomyoma. METHODS: GSE64763 and GSE5244 datasets were downloaded. Enrichment analyses were performed in GSE64763. PPI network was constructed, and the significant module was identified. Uterine leiomyoma cell lines were divided into NC, miR-29 mimic, anti-NC, and miR-29 inhibitor groups. Plate clone formation and Transwell assays detected the proliferation, invasion, and migration of cells. The expression levels of STAT3, proliferation, EMT, invasion-associated proteins were determined by Western blotting. RESULTS: Differently expressed genes were mainly enriched in positive regulation of cell migration and gene expression, cell proliferation. Through GSEA, JAK-STAT is a significantly correlated enrichment pathway. A Venn diagram was drawn to identify the common miRNA (miR-29-3p). miR-29 inhibitors promoted protein expression of STAT-3, Cyclin D1, and c-Myc compared with the anti-NC control (P < 0.01), and miR-29 inhibitors promoted cell proliferation in uterine leiomyoma cells (P < 0.05). Furthermore, miR-29 inhibitors promoted the protein expression of MMP-2 and MMP-9 (P < 0.01), and EMT promoting proteins N-cadherin, snail, vimentin, and Transwell assay showed that miR-29 inhibitors promoted cell migration in uterine leiomyoma (P < 0.01). CONCLUSIONS: High expression of miR-29 could inhibit cell proliferation, invasion, and metastasis in uterine leiomyoma, which might be related to the inhibition of the STAT3 signaling pathway, and could provide a novel target for the treatment of uterine leiomyoma.


Asunto(s)
Leiomioma , MicroARNs/metabolismo , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Leiomioma/genética , MicroARNs/genética , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Transducción de Señal
3.
BMC Cardiovasc Disord ; 21(1): 266, 2021 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-34059001

RESUMEN

BACKGROUND: Cardiovascular diseases have become the leading cause of death worldwide, and cardiac hypertrophy is the core mechanism underlying cardiac defect and heart failure. However, the underlying mechanisms of cardiac hypertrophy are not fully understood. Here we investigated the roles of Kallikrein 11 (KLK11) in cardiac hypertrophy. METHODS: Human and mouse hypertrophic heart tissues were used to determine the expression of KLK11 with quantitative real-time PCR and western blot. Mouse cardiac hypertrophy was induced by transverse aortic constriction (TAC), and cardiomyocyte hypertrophy was induced by angiotensin II. Cardiac function was analyzed by echocardiography. The signaling pathway was analyzed by western blot. Protein synthesis was monitored by the incorporation of [3H]-leucine. Gene expression was analyzed by quantitative real-time PCR. RESULTS: The mRNA and protein levels of KLK11 were upregulated in human hypertrophic hearts. We also induced cardiac hypertrophy in mice and observed the upregulation of KLK11 in hypertrophic hearts. Our in vitro experiments demonstrated that KLK11 overexpression promoted whereas KLK11 knockdown repressed cardiomyocytes hypertrophy induced by angiotensin II, as evidenced by cardiomyocyte size and the expression of hypertrophy-related fetal genes. Besides, we knocked down KLK11 expression in mouse hearts with adeno-associated virus 9. Knockdown of KLK11 in mouse hearts inhibited TAC-induced decline in fraction shortening and ejection fraction, reduced the increase in heart weight, cardiomyocyte size, and expression of hypertrophic fetal genes. We also observed that KLK11 promoted protein synthesis, the key feature of cardiomyocyte hypertrophy, by regulating the pivotal machines S6K1 and 4EBP1. Mechanism study demonstrated that KLK11 promoted the activation of AKT-mTOR signaling to promote S6K1 and 4EBP1 pathway and protein synthesis. Repression of mTOR with rapamycin blocked the effects of KLK11 on S6K1 and 4EBP1 as well as protein synthesis. Besides, rapamycin treatment blocked the roles of KLK11 in the regulation of cardiomyocyte hypertrophy. CONCLUSIONS: Our findings demonstrated that KLK11 promoted cardiomyocyte hypertrophy by activating AKT-mTOR signaling to promote protein synthesis.


Asunto(s)
Cardiomegalia/enzimología , Miocitos Cardíacos/enzimología , Biosíntesis de Proteínas , Serina Endopeptidasas/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Anciano , Animales , Cardiomegalia/tratamiento farmacológico , Cardiomegalia/genética , Cardiomegalia/patología , Estudios de Casos y Controles , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Humanos , Inhibidores mTOR/farmacología , Masculino , Ratones Endogámicos C57BL , Persona de Mediana Edad , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , Biosíntesis de Proteínas/efectos de los fármacos , Serina Endopeptidasas/genética , Transducción de Señal , Sirolimus/farmacología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Regulación hacia Arriba
4.
Ann Palliat Med ; 10(1): 597-605, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33545788

RESUMEN

BACKGROUND: Coronary heart disease (CHD) is a common clinical cardiovascular disease. This study aimed to analyze the effects of off-pump coronary artery bypass graft on the clinical efficacy, surgical indicators, and cardiac function of patients with CHD. METHODS: We retrospectively analyzed the clinical data of 120 patients with CHD who were treated in our hospital from May 2017 to May 2020. And they were divided into the control group (extracorporeal coronary artery bypass graft) and the observation group (off-pump coronary artery bypass graft). The clinical efficacy, surgical indicators, cardiac function, myocardial injury, the degree of cardiac autonomic nerve imbalance, incidence of complications and quality of life one year after the operation in the 2 groups were compared. RESULTS: The total effective rate of the observation group was significantly higher than that of the control group. Intraoperative blood loss, operation time, intraoperative blood transfusion, and hospital stay in the observation group were significantly better than those in the control group. After treatment, the levels of cardiac index (CI), ejection fraction (EF), stroke volume (SV), and cardiac output (CO) in the observation group and the control group were higher than those before treatment, especially in the observation group. Compared with those before operation, CK-MB and cTnI of the two groups significantly increased at all time points after surgery. After treatment, SDNN, LF, HF, and TP of patients in the two groups increased, which was significant in the observation group. The incidence of complications such as myocardial infarction, ischemic changes, respiratory insufficiency, and intraoperative ventricular fibrillation in the observation group was significantly lower than that in the control group. The score of quality of life in the observation group was significantly higher than the control group. CONCLUSIONS: In the treatment of patients with CHD, off-pump coronary artery bypass graft has good clinical effects, which can significantly improve the heart function, and cardiac autonomic nerve imbalance of patients, reduce myocardial damage, decrease the incidence of complications, and improve the quality of life. Therefore, off-pump coronary artery bypass graft is worthy of clinical application.


Asunto(s)
Puente de Arteria Coronaria Off-Pump , Enfermedad Coronaria , Puente de Arteria Coronaria Off-Pump/efectos adversos , Enfermedad Coronaria/cirugía , Humanos , Complicaciones Posoperatorias , Calidad de Vida , Estudios Retrospectivos , Resultado del Tratamiento
5.
Cell Biol Int ; 45(6): 1211-1219, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33554444

RESUMEN

Hypertrophic growth of the cardiomyocytes is one of the core mechanisms underlying cardiac hypertrophy. However, the mechanism underlying cardiac hypertrophy remains not fully understood. Here we provided evidence that G protein-coupled receptor 39 (GPR39) promotes cardiac hypertrophy via inhibiting AMP-activated protein kinase (AMPK) signaling. GRP39 expression is overexpressed in hypertrophic hearts of humans and transverse aortic constriction (TAC)-induced cardiac hypertrophy in mice. In neonatal cardiomyocytes, adenovirus-mediated overexpression of GPR39 promoted angiotensin II-induced cardiac hypertrophy, while GPR39 knockdown repressed hypertrophic response. Adeno-associated virus 9-mediated knockdown of GPR39 suppressed TAC-induced decline in fraction shortening and ejection fraction, increase in heart weight and cardiomyocyte size, as well as overexpression of hypertrophic fetal genes. A mechanism study demonstrated that GPR39 repressed the activation of AMPK to activate the mammalian target of rapamycin (mTOR) and ribosomal protein S6 kinase ß-1 (S6K1), subsequently promoted de novo protein synthesis. Inhibition of mTOR with rapamycin blocked the effects of GPR39 overexpression on protein synthesis and repressed cardiac hypertrophy. Collectively, our findings demonstrated that GPR39 promoted cardiac hypertrophy via regulating the AMPK-mTOR-S6K1 signaling pathway, and GRP39 can be targeted for the treatment of cardiac hypertrophy.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Cardiomegalia/metabolismo , Receptores Acoplados a Proteínas G/fisiología , Serina-Treonina Quinasas TOR/metabolismo , Animales , Células Cultivadas , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Miocitos Cardíacos , Biosíntesis de Proteínas
6.
Cell Stress Chaperones ; 25(6): 899-908, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32519137

RESUMEN

5-Hydroxytryptamine receptor 2A (HTR2A) is a central regulator of fetal brain development and cognitive function in adults. However, the roles of HTR2A in the cardiovascular system are not fully understood. Here in this study, we explored the function of HTR2A in cardiac hypertrophy. Significantly, the expression levels of HTR2A mRNA and protein levels were upregulated in hypertrophic hearts of human patients. Besides, the expression of HTR2A was also upregulated in isoproterenol (ISO)-induced cardiac hypertrophy in the mouse. Next, the expression of HTR2A was knocked down with shRNA or overexpressed with adenovirus in neonatal rat cardiomyocytes, and ISO was used to induce cardiomyocyte hypertrophy. We showed that HTR2A knockdown repressed ISO-induced cardiomyocyte hypertrophy, which was demonstrated by decreased cardiomyocyte size and repressed expression of hypertrophic fetal genes (e.g., myosin heavy chain beta (ß-Mhc), atrial natriuretic peptide (Anp), and brain natriuretic peptide (Bnp)). By contrast, HTR2A overexpression promoted cardiomyocyte hypertrophy. Of note, we observed that HTR2A promoted the activation (phosphorylation) of AKT-mTOR (mammalian target of rapamycin) signaling in cardiomyocytes, and repression of AKT-mTOR with perifosine or rapamycin blocked the effects of HTR2A on cardiomyocyte hypertrophy. Finally, we showed that HTR2A regulated AKT-mTOR signaling through activating the PI3K-PDK1 pathway, and inhibition of either PI3K or PDK1 blocked the roles of HTR2A in regulating AKT-mTOR signaling and cardiomyocyte hypertrophy. Altogether, these findings demonstrated that HTR2A activated PI3K-PDK1-AKT-mTOR signaling and promoted cardiac hypertrophy.


Asunto(s)
Proteínas Quinasas Dependientes de 3-Fosfoinosítido/metabolismo , Cardiomegalia/metabolismo , Cardiomegalia/patología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor de Serotonina 5-HT2A/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo , Animales , Animales Recién Nacidos , Cardiomegalia/genética , Humanos , Isoproterenol , Masculino , Ratones Endogámicos C57BL , Modelos Biológicos , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Ratas Sprague-Dawley , Receptor de Serotonina 5-HT2A/genética
7.
Exp Cell Res ; 393(1): 112059, 2020 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-32380038

RESUMEN

F-box and WD repeat domain-containing 7 (FBXW7) is an E3-ubiquitin ligase, which serves as one of the components of the SKP1, CUL1, and F-box protein type ubiquitin ligase (SCF) complex. Previous studies reveal that FBXW7 participates in cancer, inflammation and Parkinson's disease. FBXW7 also contributes to angiogenesis of endothelial cells. However, the function of FBXW7 in cardiac homeostasis remains to elucidate. Here we identified the critical role of FBXW7 during cardiac hypertrophy in humans and rodents. Quantitative real-time PCR (qRT-PCR) and Western blot revealed that the mRNA and protein levels of FBXW7 were upregulated significantly in hypertrophic hearts in human and mouse as well as Angiotensin II (Ang II)-induced hypertrophic neonatal rat cardiomyocytes (NRCM). Gain-of-function (adenovirus) and loss-of-function (siRNA) experiments provided evidence that FBXW7 promoted Ang II-induced cardiomyocyte hypertrophy as demonstrated by the increase in the size of cardiomyocytes and overexpression of hypertrophic fetal genes myosin heavy chain 7 (Myh7) natriuretic peptide a (Nppa), brain natriuretic peptide (Nppb). Further mechanism study revealed that FBXW7 promoted the expression of sine oculis homeobox homolog 1 (SIX1) in cardiomyocytes, which relied on regulation of the stability of the histone methyltransferase EZH2 (Enhancer of zeste homolog 2). Previous work revealed the pro-hypertrophic role of the EZH2-SIX1 axis in rodents. Indeed, our genetic and pharmacological evidence showed that the EZH2-SIX1 signaling was critically involved in FBXW7 functions in Ang II-induced cardiomyocyte hypertrophy. Therefore, we identified FBWX7 as an important regulator of cardiac hypertrophy via modulating the EZH2-SIX1 axis.


Asunto(s)
Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Proteína 7 que Contiene Repeticiones F-Box-WD/metabolismo , Animales , Cardiomegalia/patología , Células Endoteliales/metabolismo , Humanos , Ratones Endogámicos C57BL , Miocitos Cardíacos/metabolismo , Transducción de Señal
8.
Biosci Rep ; 40(5)2020 05 29.
Artículo en Inglés | MEDLINE | ID: mdl-32347291

RESUMEN

Carboxypeptidase A4 (CPA4) is a member of the metallocarboxypeptidase family. Current studies have identified the roles of CPA4 in cancer biology and insulin sensitivity. However, the roles of CPA4 in other diseases are not known. In the present study, we investigated the roles of CPA4 in cardiac hypertrophy. The expression of CPA4 was significantly increased in the hypertrophic heart tissues of human patients and isoproterenol (ISO)-induced hypertrophic heart tissues of mice. We next knocked down Cpa4 with shRNA or overexpressed Cpa4 using adenovirus in neonatal rat cardiomyocytes and induced cardiomyocyte hypertrophy with ISO. We observed that Cpa4 overexpression promoted whereas Cpa4 knockdown reduced ISO-induced growth of cardiomyocyte size and overexpression of hypertrophy marker genes, such as myosin heavy chain ß (ß-Mhc), atrial natriuretic peptide (Anp), and brain natriuretic peptide (Bnp). Our further mechanism study revealed that the mammalian target of rapamycin (mTOR) signaling was activated by Cpa4 in cardiomyocytes, which depended on the phosphoinositide 3-kinase (PI3K)-AKT signaling. Besides, we showed that the PI3K-AKT-mTOR signaling was critically involved in the roles of Cpa4 during cardiomyocyte hypertrophy. Collectively, these results demonstrated that CPA4 is a regulator of cardiac hypertrophy by activating the PI3K-AKT-mTOR signaling, and CPA4 may serve as a promising target for the treatment of hypertrophic cardiac diseases.


Asunto(s)
Carboxipeptidasas A/metabolismo , Cardiomegalia/enzimología , Tamaño de la Célula , Miocitos Cardíacos/enzimología , Fosfatidilinositol 3-Quinasa/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Proteínas Quinasas Dependientes de 3-Fosfoinosítido/metabolismo , Animales , Cardiomegalia/inducido químicamente , Cardiomegalia/genética , Cardiomegalia/patología , Estudios de Casos y Controles , Células Cultivadas , Modelos Animales de Enfermedad , Humanos , Isoproterenol , Masculino , Ratones Endogámicos C57BL , Miocitos Cardíacos/patología , Ratas Sprague-Dawley , Transducción de Señal , Regulación hacia Arriba
9.
Zhong Yao Cai ; 32(3): 384-8, 2009 Mar.
Artículo en Chino | MEDLINE | ID: mdl-19565717

RESUMEN

OBJECTIVE: To study the effect of Berberine on proliferation, differentiation and apoptosis of K562 cells for supplying the theoretical evidence on the clinical application of Coptis chinensis to cure chronic myeloid leukemia. METHODS: The proliferation-inhibiting capability of K562 cells was investigated by MTT assay and colony conform test. The apoptosis effect of Berberine on K562 cells were analyzed by Wright's-Giemsa staining; DNA fragmentation was performed by agarose gel electrophoresis. The cell cycle distribution and the cell surface marker-cluster of differentiation were determined by flow cytometry. RESULTS: Berberine effectively inhibited the proliferation of K562 cells in a dose and time-dependent manner. In addition, when combined with different concentrations of Ara-C, the cells' viability percentage was prominently higher than only Berberine used. Treated with Berberine for 48 hours, the cells could be induced differentiation towards erythrocyte, granulocyte and megakaryocyte and with the treated time extending, the percentage of apoptosis cells gradually increased. CONCLUSIONS: Berberine can efficiently inhibit the proliferation of K562 cells, maybe by the way of blocking cells at the stage of G0/G1 and (or) G2, then leading to its apoptosis and differentiation.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Berberina/farmacología , Diferenciación Celular/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Citarabina/farmacología , Relación Dosis-Respuesta a Droga , Citometría de Flujo , Humanos , Células K562
10.
Bing Du Xue Bao ; 23(1): 22-7, 2007 Jan.
Artículo en Chino | MEDLINE | ID: mdl-17886716

RESUMEN

To explore the anti-HSV-1 effect of silencing gD gene expression by RNA interference, five 21-nucleotide duplex small interfering RNAs(siRNAs) targeting the HSV1 gD sequence were designed and the gD-EGFP fusion gene expression vector was constructed, then co-transfected into Vero cell, and screened the effective siRNA through analyzing the intensity of the EGFP fluorescence. Finally, the anti-HSV1 effect was confirmed by plaque reduction assay, real-time PCR and daughter virus titration of HSV1 infected Vero cells transfected with siRNAs. The study demonstrated that siRNAs could effectively and specifically inhibit gD gene expression in HSV1-infected cells, but only had a little effect on HSV1 infection, so taking gD as the target of siRNA against HSV1 needs further study.


Asunto(s)
Herpesvirus Humano 1/genética , Interferencia de ARN , Proteínas del Envoltorio Viral/genética , Animales , Chlorocebus aethiops , Vectores Genéticos/genética , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Reacción en Cadena de la Polimerasa , ARN Interferente Pequeño/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Transfección , Células Vero , Proteínas del Envoltorio Viral/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA