Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
2.
Onco Targets Ther ; 11: 5219-5226, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30214227

RESUMEN

INTRODUCTION: Magnolol (Mag), a biologically active compound isolated from the root and stem bark of Magnolia officinalis, has been reported to induce apoptosis in several cancer cell lines in vitro. In the present study, we aimed to determine the anticancer effects of Mag on hepatocellular carcinoma (HCC) cells. MATERIALS AND METHODS: The HepG2 cells were treated with varying concentrations of Mag (10, 20, and 30 µM) for 48 hours. The effects of Mag on the proliferation, migration, invasion, apoptosis and cell cycle progression of HepG2 cells were respectively detected by MTT assay, transwell assays, and flow cytometric analysis. A HepG2 cell-based tumor-bearing model was established to evaluate the effect of Mag on HCC tumor growth in vivo. The protein expression levels were determined by Western blot analysis. RESULTS: Our results showed that Mag inhibited the proliferation, migration, and invasion of HepG2 cells in vitro in a dose-dependent manner. In addition, Mag reduced the HCC tumor volume and weight in the mouse xenograft model. Subsequent studies showed that Mag induced apoptosis in HepG2 cells, accompanied by a loss in mitochondrial membrane potential, cytochrome c release, and induction of endoplasmic reticulum stress. Furthermore, inhibition of the endoplasmic reticulum stress by CHOP knockdown restored the effects of Mag in HepG2 cells. CONCLUSION: The present study highlighted the possibility of using Mag as a novel therapeutic drug for HCC treatment.

3.
Biomed Pharmacother ; 106: 134-141, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29957463

RESUMEN

Long non-coding RNAs (lncRNAs) have emerged as key regulators in the development of hepatocellular carcinoma (HCC). In the present study, we explored the expression profile and biological role of lncRNA FEZF1-AS1 in HCC. We observed remarkable upregulation of FEZF1-AS1 in HCC tissues and cell lines, and high FEZF1-AS1 expression was correlated with aggressive phenotypes and poor prognosis of HCC patients. Furthermore, we found that FEZF1-AS1 knockdown markedly inhibited the proliferation of HCC cells by inducing cell cycle arrest. In addition, FEZF1-AS1 knockdown suppressed HCC tumor growth in vivo. Moreover, FEZF1-AS1 knockdown inhibited the migration and invasion of HCC cells through suppression of JAK2/STAT3 signaling-mediated epithelial-mesenchymal transition (EMT). In conclusion, the present study for the first time demonstrated that FEZF1-AS1 serves as an oncogenic lncRNA in human HCC and implicated FEZF1-AS1 as a valuable therapeutic target for HCC treatment.


Asunto(s)
Carcinoma Hepatocelular/enzimología , Movimiento Celular , Transición Epitelial-Mesenquimal , Janus Quinasa 2/metabolismo , Neoplasias Hepáticas/enzimología , ARN Largo no Codificante/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Animales , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/mortalidad , Carcinoma Hepatocelular/patología , Puntos de Control del Ciclo Celular , Proliferación Celular , Femenino , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Células Hep G2 , Humanos , Janus Quinasa 2/genética , Estimación de Kaplan-Meier , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/mortalidad , Neoplasias Hepáticas/patología , Masculino , Ratones Endogámicos BALB C , Ratones Desnudos , Persona de Mediana Edad , Invasividad Neoplásica , ARN Largo no Codificante/genética , Factor de Transcripción STAT3/genética , Factores de Tiempo , Carga Tumoral
4.
Exp Ther Med ; 15(5): 4508-4514, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-29725386

RESUMEN

The aim of this study was to investigate the function of microRNA (miR)-193-3p in human intrahepatic cholangiocarcinoma (ICC) tissues and cells. To evaluate whether miR-193-3p was aberrantly upregulated, we used reverse transcription-quantitative polymerase chain reaction to detect the level of miR-193-3p in ICC tissues and ICC-9810 cells. The effects of miR-193-3p downregulation on ICC cell proliferation, migration and invasion were also measured by MTT, wound-healing and Transwell assays. Additionally, transforming growth factor-ß receptor type 3 (TGFBR3) was investigated as a direct target of miR-193-3p by dual-luciferase reporter assays and western blot analyses. The results demonstrated that miR-193-3p was aberrantly upregulated in ICC tissues and cell lines. Furthermore, TGFBR3 was confirmed to be a target of miR-193-3p in ICC and was notably upregulated by miR-193-3p knockdown in the ICC-9810 cells. The knockdown of miR-193-3p also exerted direct inhibitory effects on the proliferation, migration and invasion of the ICC-9810 cells. Therefore, we present evidence that miR-193-3p plays a key role in promoting human ICC by regulating TGFBR3.

5.
Sci Rep ; 8(1): 302, 2018 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-29321592

RESUMEN

Alzheimer's disease (AD) is the most common cause of dementia in the elderly. At the early stages of AD development, the soluble ß-amyloid (Aß) induces synaptic dysfunction, perturbs the excitation/inhibition balance of neural circuitries, and in turn alters the normal neural network activity leading to cognitive decline, but the underlying mechanisms are not well established. Here by using whole-cell recordings in acute mouse brain slices, we found that 50 nM Aß induces hyperexcitability of excitatory pyramidal cells in the cingulate cortex, one of the most vulnerable areas in AD, via depressing inhibitory synaptic transmission. Furthermore, by simultaneously recording multiple cells, we discovered that the inhibitory innervation of pyramidal cells from fast-spiking (FS) interneurons instead of non-FS interneurons is dramatically disrupted by Aß, and perturbation of the presynaptic inhibitory neurotransmitter gamma-aminobutyric acid (GABA) release underlies this inhibitory input disruption. Finally, we identified the increased dopamine action on dopamine D1 receptor of FS interneurons as a key pathological factor that contributes to GABAergic input perturbation and excitation/inhibition imbalance caused by Aß. Thus, we conclude that the dopamine receptor 1-dependent disruption of FS GABAergic inhibitory input plays a critical role in Aß-induced excitation/inhibition imbalance in anterior cingulate cortex.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Neuronas GABAérgicas/metabolismo , Giro del Cíngulo/metabolismo , Receptores de Dopamina D1/metabolismo , Análisis de Varianza , Animales , Potenciales Postsinápticos Excitadores , Interneuronas/metabolismo , Masculino , Ratones , Microscopía Confocal , Células Piramidales/metabolismo , Potenciales Sinápticos , Ácido gamma-Aminobutírico/metabolismo
6.
Asian Pac J Trop Med ; 7(6): 468-72, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25066396

RESUMEN

OBJECTIVE: To explore inhibition effects of veliparib as PARP inhibitor combined doxorubicin for BEL-7404 proliferation of human liver cancer cell line. METHODS: BEL-7404 was taken as the object of study and conventional culture was performed. It was treated by doxorubicin and (or) veliparib after 24 h. Cell proliferation rate was detected by four methyl thiazolyl tetrazolium (MTT) assay, cell apoptosis was measured with annexin V-FITC/PI double staining method by flow cytometry, DNA damage degree evaluation by single cell gel electrophoresis assay, and cytosolic C levels of the mitochondrial and cytosol by polyacrylamide gel electrophoresis (Western blotting). RESULTS: Cell proliferation rate of doxorubicin combined veliparib group was lower than that of the control group and doxorubicin alone treated group significantly (P<0.01), the apoptosis rate was significantly higher than that of the control group and doxorubicin alone treated group (P<0.05). At the same time, DNA damage level of doxorubicin combined with veliparib group was significantly higher than doxorubicin alone treatment group and the control group (P<0.01), and cytochrome C in the cytosol was significantly higher than that of control group and doxorubicin alone treated group (P<0.01). CONCLUSIONS: Veliparib, PARP inhibitor could inhibit PARP activity, block tumor cell DNA repair, and have significant sensitizing effect for hepatocellular carcinoma cell line BEL-7404 treated with doxorubicin. This might provide a new target for clinical treatment of hepatic carcinoma.


Asunto(s)
Antineoplásicos/farmacología , Bencimidazoles/farmacología , Proliferación Celular/efectos de los fármacos , Doxorrubicina/farmacología , Neoplasias Hepáticas/metabolismo , Antineoplásicos/toxicidad , Apoptosis/efectos de los fármacos , Bencimidazoles/toxicidad , Línea Celular Tumoral , Citocromos c/metabolismo , Daño del ADN/efectos de los fármacos , Doxorrubicina/toxicidad , Humanos
7.
Planta Med ; 78(7): 698-702, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22411728

RESUMEN

The intestinal permeability of forskolin was investigated using a single pass intestinal perfusion (SPIP) technique in rats. SPIP was performed in different intestinal segments (duodenum, jejunum, ileum, and colon) with three concentrations of forskolin (11.90, 29.75, and 59.90 µg/mL). The investigations of adsorption and stability were performed to ensure that the disappearance of forskolin from the perfusate was due to intestinal absorption. The results of the SPIP study indicated that forskolin could be absorbed in all segments of the intestine. The effective permeability (P (eff)) of forskolin was in the range of drugs with high intestinal permeability. The P (eff) was highest in the duodenum as compared to other intestinal segments. The decreases of P (eff) in the duodenum and ileum at the highest forskolin concentration suggested a saturable transport process. The addition of verapamil, a P-glycoprotein inhibitor, significantly enhanced the permeability of forskolin across the rat jejunum. The absorbed fraction of dissolved forskolin after oral administration in humans was estimated to be 100 % calculated from rat P (eff). In conclusion, dissolved forskolin can be absorbed readily in the intestine. The low aqueous solubility of forskolin might be a crucial factor for its poor oral bioavailability.


Asunto(s)
Coleus/química , Colforsina/administración & dosificación , Colforsina/farmacocinética , Mucosa Intestinal/metabolismo , Plectranthus/química , Administración Oral , Animales , Disponibilidad Biológica , Colon/metabolismo , Duodeno/metabolismo , Humanos , Íleon/metabolismo , Absorción Intestinal/efectos de los fármacos , Yeyuno/metabolismo , Masculino , Perfusión/métodos , Permeabilidad , Fitoterapia , Extractos Vegetales/administración & dosificación , Extractos Vegetales/farmacocinética , Ratas , Ratas Sprague-Dawley , Verapamilo/farmacología
8.
Zhonghua Yi Xue Za Zhi ; 83(14): 1274-7, 2003 Jul 25.
Artículo en Chino | MEDLINE | ID: mdl-12930647

RESUMEN

OBJECTIVE: To construct the tumor-specific expression vector driven by human telomerase reserve transcriptase gene promotor. METHODS: The fragment of the enhanced green fluorescent protein (EGFP) gene was PCR amplified from the pEGFP-N1 plasmid and cloned into the multiple cloning site of pLNCX vector, and the recombinant was named as pLNCX-EGFP. The fragment of human telomerase reserve transcriptase gene promoter was amplified from the human genome by using the human telomerase reserve transcriptase gene specific primers, and cloned into the pLNCX-EGFP vector, where the cytomegalovirus promoter was previously removed using restriction enzymes, in sense orientation relative to the green fluorescent protein coding sequence. Then the expression vector pLNT-EGFP under the control of the human telomerase reserve transcriptase gene promoter, containing green fluorescent protein reporter gene, was successfully constructed. To detect the transcriptional activity of the human telomerase reserve transcriptase gene promoter, transient transfection of this specific expression vector into HLF cell lines with high telomerase activity and WI38 cell lines without telomerase activity was performed. RESULTS: The expression vector proven by restriction enzymes digestion and sequencing was in correspondence with the design. The results of transient transfection showed that the pLNT-EGFP vector could highly expressed green fluorescent protein reporter gene in telomerase-positive cells, but not in telomerase-negative cells. CONCLUSION: A tumor-specific expression vector driven by human telomerase reserve transcriptase gene promotor has been successfully constructed.


Asunto(s)
Vectores Genéticos , Regiones Promotoras Genéticas , Telomerasa/genética , Citomegalovirus/genética , ADN sin Sentido/análisis , ADN Complementario/análisis , Proteínas de Unión al ADN , Terapia Genética , Humanos , Neoplasias/metabolismo , Neoplasias/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA