Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
1.
Br J Cancer ; 117(2): 233-244, 2017 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-28588321

RESUMEN

BACKGROUND: Accumulating evidence indicates that N-cadherin is a cell adhesion molecule that has critical roles in tumour progression. However, the role of N-cadherin in hepatocellular carcinoma (HCC) remains controversial. METHODS: This study aims to investigate the expression status of N-cadherin and its molecular mechanisms in HCC. RESULTS: The expression of N-cadherin was markedly overexpressed in HCC tissues and cell lines. We identified that miR-199b-5p binds to the 3'-UTR of N-cadherin mRNA, thus decreasing N-cadherin expression in HCC cells. We also found the downregulation of miR-199b-5p in HCC specimens, which was inversely correlated with N-cadherin upregulation, predicted poor clinical outcomes in HCC patients. Next, we determined that miR-199b-5p overexpression promoted cell aggregation, suppressed cell migration and invasion in HCC cells, and inhibited xenografts tumour metastasis in nude mice. Moreover, we demonstrated that miR-199b-5p attenuated TGF-ß1 induced epithelial-mesenchymal transition (EMT) -associated traits, while its effects could be partially reversed by N-cadherin restoration. Finally, we examined that N-cadherin downregulation or miR-199b-5p overexpression suppressed TGF-ß1-induced Akt phosphorylation, and inhibition of PI3K/Akt pathway blocked TGF-ß1-induced N-cadherin overexpression in HCC cells. CONCLUSIONS: Our data demonstrate that N-Cadherin was markedly overexpressed and miR-199b-5p was significantly downregulated in HCC. MiR-199b-5p exerts inhibitory effects on EMT, and directly targets N-cadherin in HCC, supporting the potential utility of miR-199b-5p as a promising strategy to treat HCC. Also, a positive regulatory loop exists between N-cadherin and Akt signalling represents a novel mechanism of TGF-ß1-mediated EMT in HCC cells.


Asunto(s)
Cadherinas/biosíntesis , Carcinoma Hepatocelular/genética , Neoplasias Hepáticas/genética , MicroARNs/genética , Factor de Crecimiento Transformador beta1/genética , Animales , Cadherinas/genética , Carcinoma Hepatocelular/patología , Movimiento Celular/genética , Transición Epitelial-Mesenquimal/genética , Regulación Neoplásica de la Expresión Génica , Células Hep G2 , Humanos , Neoplasias Hepáticas/patología , Ratones , Transducción de Señal , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Cell Death Differ ; 24(9): 1577-1587, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28574502

RESUMEN

Familiar clustering of hepatitis B virus (HBV)-related hepatocellular carcinoma (HCC) has been frequently reported. However, limited information is available about the underlying molecular mechanisms in HBV-related HCC patients with family history of HCC. In our previous study, Agilent miRNA Base 16.0 microarray showed miRNA profiles of the plasma of HBV-related HCC patients who had a family history of HCC. This study aims to explore the expression, function, and mechanisms of miR-3188 in HCC that might provide novel insights into the role of family history on the risk of HCC. The expression levels of miR-3188 were markedly overexpressed in HCC tissues, HBV transgenic mice, and HepG2.215 cells. We knocked out miR-3188 in HCC cell lines using the CRISPR/Cas9 system, and demonstrated that miR-3188 knockout (KO) suppressed cell growth, migration, and invasion, and inhibited xenografts tumor growth in nude mice. Next, we determined that miR-3188 KO exerts antitumor functions by directly repressing ZHX2. It has been reported that HBV X protein (HBx) plays a critical role in HBV-related HCC, promoting CREB-mediated activation of miR-3188 and activation of Notch signaling through repressing ZHX2. Finally, we verified that ZHX2 functions as a transcriptional repressor to Notch1 via interaction with NF-YA. Our data demonstrate that the HBx-miR-3188-ZHX2-Notch1 signaling pathway plays an important role in the pathogenesis and progression of HBV-related HCC with family history of HCC. These findings have important implications for identifying new therapeutic targets in HBV-related HCC.


Asunto(s)
Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/metabolismo , MicroARNs/metabolismo , Receptores Notch/metabolismo , Transactivadores/metabolismo , Animales , Carcinoma Hepatocelular/genética , Inmunoprecipitación de Cromatina , Regulación Neoplásica de la Expresión Génica/genética , Regulación Neoplásica de la Expresión Génica/fisiología , Células Hep G2 , Humanos , Técnicas In Vitro , Neoplasias Hepáticas/genética , Ratones , Ratones Noqueados , Ratones Desnudos , Ratones Transgénicos , MicroARNs/genética , Receptores Notch/genética , Transducción de Señal/genética , Transducción de Señal/fisiología , Transactivadores/genética , Proteínas Reguladoras y Accesorias Virales
4.
Mol Med Rep ; 13(2): 1345-52, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26648552

RESUMEN

Transforming growth factor (TGF)-ß induces cell growth arrest in well-differentiated hepatocellular carcinoma (HCC) while hepatitis B virus X protein (HBx) minimizes the tumor suppression of TGF-ß signaling in early chronic hepatitis B. However, how to reverse the oncogenic effect of HBx and sustain the tumor-suppressive action of TGF-ß has yet to be investigated. The present study examined the effect of TGF-ß and a c-Jun N-terminal kinase (JNK) inhibitor on cell growth in HCC cells with forced expression of HBx. It was found that HBx promoted cell growth via activation of the JNK/pSMAD3L pathway and inhibition of the transforming growth factor-beta type I receptor (TßRI)/pSMAD3C pathway. pSMAD3L/SMAD4 and pSMAD3C/SMAD4 complexes antagonized each other to regulate c-Myc expression. In the absence of HBx, TGF-ß induced cell growth arrest through activation of the TßRI/pSMAD3C pathway in well-differentiated HCC cells. In the presence of HBx, TGF-ß had no effect on cell growth. JNK inhibitor SP600125 significantly reversed the oncogenic action of HBx and favored TGF-ß to regain the ability to inhibit the cell growth in HBx-expressing well-differentiated HCC cells. In conclusion, targeting JNK signaling favors TGF-ß to block HBx-induced cell growth promotion in well-differentiated HCC cells. As an adjunct to anti-viral therapy, the combination of TGF-ß and inhibition of JNK signaling is a potential therapy for HBV-infected HCC.


Asunto(s)
Carcinoma Hepatocelular/genética , Proteínas Quinasas JNK Activadas por Mitógenos/genética , Neoplasias Hepáticas/genética , Transactivadores/metabolismo , Factor de Crecimiento Transformador beta/biosíntesis , Carcinoma Hepatocelular/patología , Carcinoma Hepatocelular/virología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Virus de la Hepatitis B/efectos de los fármacos , Virus de la Hepatitis B/genética , Hepatitis B Crónica/genética , Hepatitis B Crónica/patología , Hepatitis B Crónica/virología , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/biosíntesis , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/virología , Inhibidores de Proteínas Quinasas/administración & dosificación , Proteínas Proto-Oncogénicas c-myc/biosíntesis , Proteínas Proto-Oncogénicas c-myc/genética , Transducción de Señal/efectos de los fármacos , Proteína smad3/genética , Proteína Smad4/biosíntesis , Proteína Smad4/genética , Transactivadores/genética , Factor de Crecimiento Transformador beta/genética , Proteínas Reguladoras y Accesorias Virales
5.
Oncol Rep ; 32(2): 835-44, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24919588

RESUMEN

Minocycline, a semisynthetic tetracycline, is a highly lipophilic molecule capable of infiltrating tissues and blood. Previous studies have revealed the functions and mechanisms of minocycline in anti-inflammation, protection of the nervous system and certain tumors. The role of minocycline has never been investigated in hepatocellular carcinoma (HCC). The functions of minocycline on HCC cells were investigated using immunohistochemical staining and western blotting. Minocycline was applied to L02, HepG2 and Huh7 cells, and the growth characteristics were studied. Cisplatin was administered in combination with minocycline in this study. Cell cycle and apoptosis analyses were employed to investigate the mechanisms underlying the growth regulation associated with minocycline and(or) cisplatin. Minocycline caused S phase cell cycle arrest and an increase in the apoptotic rate associated with upregulation of p27, cleaved-caspase8, cleaved-caspase3 and cleaved-PRAP-1. Low dose of cisplatin promoted cell cycle arrest and apoptosis, whereas minocycline was mainly associated with upregulation of cleaved-PARP-1. The combination of cisplatin and minocycline increased the rate and extent of cell cycle arrest and increased the apoptosis rate caused by minocycline. A novel mechanism was revealed. Minocycline functions as an antitumor drug in HCC by regulating p27, caspase-3 and PARP-1. Cisplatin enhanced minocycline's effect on PARP-1.


Asunto(s)
Antineoplásicos/farmacología , Carcinoma Hepatocelular/tratamiento farmacológico , Cisplatino/farmacología , Neoplasias Hepáticas/tratamiento farmacológico , Minociclina/farmacología , Animales , Protocolos de Quimioterapia Combinada Antineoplásica , Apoptosis/efectos de los fármacos , Carcinoma Hepatocelular/patología , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Sinergismo Farmacológico , Células Hep G2 , Humanos , Neoplasias Hepáticas/patología , Ratones , Ratones Endogámicos BALB C , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Med Oncol ; 30(1): 425, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23325444

RESUMEN

Although the role of Krüppel-like factor 17 (KLF17) in regulating epithelial-mesenchymal transition (EMT) has been explored in breast cancer, its influence on primary hepatocellular carcinoma (HCC) remains unclear. This study aims to investigate the expression status of KLF17 in hepatocellular carcinoma (HCC) and the correlation between KLF17 expression and metastatic potential of HCC. KLF17 expression in HCC and adjacent liver tissues was studied by real-time PCR and Western blot, and the relationship between KLF17 expression and the clinicopathological features of HCC was evaluated in 60 patients. By using RNA interference technique, the correlation of KLF17 expression and metastatic potential was investigated by down-regulating KLF17 expression in HepG2 cells, and the effects of KLF17 down-regulation on cell migration, and invasion were then analyzed. Furthermore, the correlation between KLF17 expression and the surgical outcomes of a cohort of HCC patients was analyzed. Reduced expression of KLF17 is associated with a short survival time in clinical patients (P = 0.034). Low KLF17 expression is related to tumor T stage (P = 0.045), tumor size (P = 0.027), lymph node stage (P = 0.030), M stage (P = 0.048), and portal vein tumor thrombosis significantly in HCC. Reduced expression of KLF17 promoted motility and invasion ability of HepG2 cells and changed the expression of E-cadherin, ZO-1, Snai1, and vimentin (genes are associated with EMT). Overall, these findings suggest a repressing role of KLF17 in tumor invasion and a new prognostic indicator in directing therapy. It deserves further exploration.


Asunto(s)
Biomarcadores de Tumor/análisis , Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/metabolismo , Invasividad Neoplásica , Factores de Transcripción/biosíntesis , Western Blotting , Carcinoma Hepatocelular/mortalidad , Carcinoma Hepatocelular/patología , Regulación hacia Abajo , Femenino , Humanos , Estimación de Kaplan-Meier , Neoplasias Hepáticas/mortalidad , Neoplasias Hepáticas/patología , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Pronóstico , Reacción en Cadena en Tiempo Real de la Polimerasa , Factores de Transcripción/análisis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA