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
1.
Infect Agent Cancer ; 14: 13, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31114628

RESUMEN

AIM: To evaluate the correlation of serum PIVKA-II levels and development of portal vein tumor thrombus (PVTT) in hepatocellular carcinoma (HCC) patients. METHODS: One hundred and twenty-three patients with newly diagnosed HCC were included in this study between March 2016 and October 2018. Thirty-five of these patients were detected with PVTT and all subjects were randomly divided to analysis group (N = 73) and validation (N = 50) group. Serum levels of PIVKA-II, laboratory tests including serum aspartate aminotransferase, total bilirubin, platelet count, albumin levels were demonstrated in all the patients. T-test, chi-squared test and logistic regression was used for analyzing data. Diagnostic efficiency and cut-off value of PIVKA-II in PVTT development of HCC patients were calculated using receiver operator curve (ROC) analysis. RESULTS: Serum level of PIVKA-II in HCC patients with PVTT was significantly higher than that in HCC patients without PVTT (995.8 mAU/ml vs 94.87 mAU/ml; P = 0.003), as well as D-dimer levels (2.12 mg/L vs 0.56 mg/L P = 0.001). Univariate analysis showed that high serum D-dimer level was an independent risk factor for development of PVTT (OR = 1.22, 95%CI 1.02-1.45). ROC curve showed that among analysis group, the area under ROC curve (AUROC) of PIVKA-II was 0.73 (95%CI 0.59-0.86). For the detection of PVTT in HCC, PIVKA-II had a sensitivity of 83.7% and a specificity of 69.2% at a cutoff of 221.26 mAU/ml, which had a sensitivity of 85.71% and a specificity of 55.56% in validation group, respectively. CONCLUSION: Serum PIVKA-II level is a potential marker for diagnosis of PVTT in HCC patients, which may guide therapeutic strategy and assessment of tumor prognosis of HCC.

2.
Int J Clin Exp Med ; 8(6): 9793-8, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26309658

RESUMEN

BACKGROUND: IL-17A G197A and IL-17F A7488G gene polymorphisms are found to be associated with the risk of several diseases, however few studies have focused on their correlation with risk of liver cirrhosis (LC). AIMS: The aim of this study was to assess the impact of IL-17A G197A and IL-17F A7488G gene polymorphisms on development of LC from chronic hepatitis B (CHB) in Chinese patients. METHODS: A total of 163 HBV-related LC patients and 168 CHB patients were enrolled in the present study. IL-17A and IL-17F gene polymorphisms were analyzed using a polymerase chain reaction (PCR) restriction fragment length polymorphism assay (PCR-RFLP). RESULTS: Frequencies of IL-17A G197A genotype AA and allele A were significantly higher in LC patients compared with that in CHB patients (AA 42.33% vs 27.98%, P = 0.032; A 56.34% vs 46.15%, P=0.011, respectively); While no significant difference in frequencies of genotypes and alleles of IL-17F A7488G was found between the two groups. The AA genotype of IL-17A G197A significantly increased LC risk (OR 4.186, 95% CI: 1.479-11.844), while A allele carriers were also associated with an increased LC risk (OR 1.856, 95% CI: 1.161-2.967). CONCLUSION: IL-17A G197A is a candidate gene that confers the genetic susceptibility for LC development from CHB in Chinese population.

3.
Tumour Biol ; 36(4): 2885-92, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25480416

RESUMEN

The objective of the study was to investigate the impact of Numb on cell growth, cell migration, and invasion in human clear cell renal cell carcinoma (ccRCC). Endogenous expression of Numb was evaluated in the ccRCC cell lines (786-O, Caki-1, and Caki-2) and control reference human renal proximal tubular epithelial cells. Numb expression was decreased in the ccRCC cells compared with the control cells (P < 0.01). Then, 786-O and Caki-1 cells described as suitable transfection hosts were used in transfection to carry out biological function studies. The three experimental groups were as follows: Numb-ORF (transfected with Numb-ORF plasmid), blank-vector (transfected with pCMV6-entry), and cell-alone group (no DNA). Numb expression in the Numb-ORF groups was significantly higher than that in the controls (P < 0.01). Cell growth was remarkably reduced (P < 0.01), and the number of migrating or invading cells was reduced (P < 0.01) in the Numb-ORF groups compared with controls. Furthermore, the ratio of G0/G1 phase in the Numb-ORF group of 786-O cells was increased, and the S phase fraction and proliferation index was decreased (P < 0.01). Cyclin D1 and MMP-9 expression was reduced in the Numb-ORF groups compared with controls. Here, we have provided data for attenuated Numb expression in the ccRCC cells. Overexpression of Numb could induce G0/G1 phase arrest and inhibit cell proliferation, migration, and invasion. The suppressive effects might be due to downregulation of cyclin D1 or MMP-9 expression. Taken together, our data suggest that Numb may possibly function as a tumor suppressor involved in the carcinogenesis of ccRCC.


Asunto(s)
Apoptosis/genética , Carcinoma de Células Renales/genética , Proliferación Celular/genética , Proteínas de la Membrana/biosíntesis , Proteínas del Tejido Nervioso/biosíntesis , Carcinoma de Células Renales/patología , Línea Celular Tumoral , Movimiento Celular/genética , Ciclina D1/biosíntesis , Fase G1/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Metaloproteinasa 9 de la Matriz/genética , Proteínas de la Membrana/genética , Invasividad Neoplásica/genética , Proteínas del Tejido Nervioso/genética
4.
Am J Cancer Res ; 4(6): 710-24, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25520862

RESUMEN

Dapper, Dishevelled-associated antagonist of ß-catenin (DACT), is a key regulator of Wnt signaling pathway. The purpose of this study is to explore the epigenetic changes and the function ofDACT2 in human gastric cancer (GC). Eight human gastric cancer cell lines, 167 cases of primary gastric cancer and 8 cases of normal gastric mucosa were involved in this study. In addition, methylation Specific PCR (MSP), semi-quantitative RT-PCR, colony formation assay, flow cytometry assay, siRNA, immunofluorescence techniques and xenograft mice models were employed. The results indicate that DACT2 is frequently methylated in human primary gastric cancer (55.7%), and that methylation of DACT2 is associated with lost or reduction in its expression (X(2) test, P<0.01). We found that DACT2 expression was regulated by promoter region hypermethylation. Methylation of DACT2 is associated with tumor differentiation, invasion and intravascular cancerous emboli (X(2) test, P<0.05, P<0.05 and P<0.05). In gastric cancer patients treated with 5-FU and cisplatin, the five-year survival rates are higher in DACT2 methylated cases. DACT2 inhibits cell proliferation, migration and invasion in gastric cancer cells and suppresses gastric cancer xenografts in mice. Restoration of DACT2 expression inhibits both canonical and noncanonical WNT signaling in SGC7901 cells. Restoration of DACT2 expression sensitized gastric cancer cells to paclitaxel and 5-FU. In conclusion, DACT2 is frequently methylated in human gastric cancer and DACT2 expression is silenced by promoter region hypermethylation. DACT2 suppressed gastric cancer proliferation, invasion and metastasis by inhibiting Wnt signaling both in vitro and in vivo.

5.
Discov Med ; 14(74): 33-40, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22846201

RESUMEN

The purpose of this study was to explore epigenetic changes and functions of SOX17 in human lung cancer. Five lung cancer cell lines and 88 primary lung cancer samples were examined in this study. Methylation-specific polymerase chain reaction (MSP), semi-quantitative reverse-transcription PCR, immunohistochemistry, luciferase reporter assays, colony-formation assays, and western blotting were used to analyze methylation changes and functions of SOX17 in lung cancer. SOX17 methylation was found in 60.2% of primary human lung cancer samples, and promoter region methylation of SOX17 silenced its expression. SOX17 methylation was associated with female patients and lung cancer differentiation. Colony-formation assays revealed that SOX17 suppressed lung cancer cell proliferation. Re-expression of SOX17 inhibited Wnt signaling in H23 lung cancer cell line. SOX17 acts as a Wnt signaling inhibitor.


Asunto(s)
Metilación de ADN/genética , Neoplasias Pulmonares/genética , Factores de Transcripción SOXF/genética , Vía de Señalización Wnt , Anciano , Línea Celular Tumoral , Células Clonales , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Pulmonares/patología , Masculino , Persona de Mediana Edad , Factores de Transcripción SOXF/metabolismo , Activación Transcripcional/genética , Ensayo de Tumor de Célula Madre , beta Catenina/genética , beta Catenina/metabolismo
6.
J Mol Diagn ; 14(6): 577-85, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22921431

RESUMEN

In this study, we explored the possibility of SOX17 promoter region methylation as an esophageal cancer detection marker, the regulation of SOX17 expression, and the function of SOX17 in the WNT signaling pathway in esophageal cancer. Eight esophageal cancer cell lines, 9 normal esophageal mucosa samples, 60 cases of dysplasia, and 169 cancer tissue samples were included. Methylation-specific PCR, semiquantitative reverse transcription-PCR, immunohistochemistry, luciferase reporter assay, colony formation, and Western blot analysis were used to analyze methylation and function of SOX17 in esophageal cancer. MicroRNA-related detection methods were performed to evaluate microRNA regulation of SOX17. SOX17 methylation was found in progression tendency with 0% of normal mucosa, 39% of grade 1 dysplasia, 48% of grades 2 and 3 dysplasia, and 65% of primary cancer. SOX17 methylation is related to esophageal cancer patients' history of alcohol use and may induce ß-catenin expression and redistribution. Loss of SOX17 expression is correlated to promoter region hypermethylation, and re-expression was activated by 5-aza-2'-deoxycytidine treatment in esophageal cancer cell lines. Restoration of SOX17 expression suppresses TCF/ß-catenin-dependent transcription and colony formation. MicroRNA 141 was also found to down-regulate SOX17 expression and activate the WNT signal pathway. SOX17 is frequently methylated in esophageal cancer and in a progression tendency during esophageal carcinogenesis. Loss of SOX17 removes the normal inhibition of WNT signaling and promotes esophageal tumorigenesis.


Asunto(s)
Neoplasias Esofágicas/genética , Neoplasias Esofágicas/patología , Esófago/patología , Regulación Neoplásica de la Expresión Génica , MicroARNs/genética , Factores de Transcripción SOXF/genética , Vía de Señalización Wnt , Línea Celular Tumoral , Regulación hacia Abajo , Neoplasias Esofágicas/metabolismo , Esófago/metabolismo , Femenino , Humanos , Masculino , Metilación , Regiones Promotoras Genéticas , beta Catenina/genética
7.
Epigenomics ; 4(2): 135-46, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22449186

RESUMEN

AIMS: To explore the epigenetic changes and the function of TFPI-2 in esophageal cancer. MATERIALS & METHODS: Nine esophageal cancer cell lines, nine normal esophageal mucosa, 60 esophageal dysplasia and 106 advanced esophageal cancer samples were included in this study. TFPI-2 methylation was examined by methylation-specific PCR. TFPI-2 expression was evaluated by immunohistochemistry in tissue samples. The effect of TFPI-2 on proliferation, apoptosis, invasion and migration was analyzed by colony formation assay, western blot assay, transwell assay and flow cytometric analysis. RESULTS: TFPI-2 expression was regulated by promoter region hypermethylation in human esophageal cancer cell lines, and TFPI-2 expression is inversely correlated with methylation in primary cancer. Methylation was found in 28.2, 33.3 and 33.3% of grade 1, 2 and 3 esophageal dysplasia, and 67% of primary esophageal cancer, but no methylation was found in normal mucosa. Methylation is significantly related to tumor differentiation. Inhibition of invasion, migration, colony formation and proliferation, and induction of apoptosis occurred with the restoration of TFPI-2 expression in the KYSE70 cell line. CONCLUSION: TFPI-2 is frequently methylated in esophageal cancer with a progression tendency. TFPI-2 is a potential tumor suppressor in esophageal cancer.


Asunto(s)
Metilación de ADN , Neoplasias Esofágicas/metabolismo , Glicoproteínas/metabolismo , Adulto , Anciano , Apoptosis , Puntos de Control del Ciclo Celular , Línea Celular Tumoral , Movimiento Celular , Transformación Celular Neoplásica/genética , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/patología , Femenino , Glicoproteínas/genética , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Membrana Mucosa/metabolismo , Estadificación de Neoplasias , Regiones Promotoras Genéticas
8.
Tumour Biol ; 33(2): 307-14, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22095135

RESUMEN

The purpose of this study is to determine the epigenetic changes and function of High in Normal-1 (HIN-1) in non-small cell lung cancer (NSCLC). HIN-1 expression was examined by semiquantitative RT-PCR before and after 5-aza-2'-deoxycytidine (5-aza) treatment in NSCLC cell lines. Promoter methylation status of HIN-1 was tested by methylation-specific PCR (MSP). Effect of forced expression of HIN-1 on different key molecules of AKT signaling pathway was tested by Western Blot analysis in H157 and H23 cell lines. Promoter methylations are inversely correlated with expression of HIN-1 in eight (H23, H157, 95D, H1299, H358, H1752, H460, A549) of ten NSCLC cell lines and re-expression was observed by 5-aza treatment. We then tested promoter methylation of HIN-1 in primary NSCLC tissues. Methylation was detected in 73 out of 152 (48%) NSCLC cases. Forced expression of HIN-1 in NSCLC cell lines inhibited colony formation and induce apoptosis. Furthermore, overexpression of HIN-1 reduces the expression of phosphorated-AKT (p-AKT), c-myc, Bcl-2 and cyclinD1 while Bax was increased. Our data suggest that HIN-1 is a potential tumor suppressor gene in NSCLC, silenced by promoter hypermethylation and negatively regulate AKT signaling pathway.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/genética , Citocinas/genética , Regulación Neoplásica de la Expresión Génica , Neoplasias Pulmonares/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Supresoras de Tumor/genética , Anciano , Azacitidina/farmacología , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Línea Celular Tumoral , Femenino , Silenciador del Gen , Genes Supresores de Tumor , Humanos , Neoplasias Pulmonares/metabolismo , Masculino , Persona de Mediana Edad , Regiones Promotoras Genéticas , Reacción en Cadena en Tiempo Real de la Polimerasa/métodos , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...