Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Biochem Biophys Res Commun ; 505(1): 7-12, 2018 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-30224058

RESUMEN

The role of long non-coding RNA in Renal cell carcinoma (RCC) tumorigenesis and progression remains largely unknown. Here, we found that LINC01510 functions as a tumor suppressor in RCC tumorigenesis. We screened TCGA database and then found that LINC01510 is significantly down-regulated in malignant RCC tissues, and the lower expression of LINC01510 predicts poor prognosis. Moreover, the down-regulated LINC01510 was further confirmed in our fresh tissues and cell lines. Biological functions assays shown that Ectopic expression of LINC01510 not only inhibits RCC cell proliferation both in vitro and in vivo, but also impairs cell invasion ability. Moreover, we found overexpression of LINC01510 inhibits the expression of CCND1 and CCNE1, as well as MMPs (MMP2, MMP7 and MMP9), and thus affecting RCC cell cycle and invasion. Meanwhile, Western blot assays revealed that the expression of ß-catenin is regulated by LINC01510; overexpression of ß-catenin could partly rescue the cell viability and invasion ability caused by ectopic expression of LINC01510. Taken together, we found that LINC01510 regulates cell proliferation and invasion by modulating Wnt/ß-catenin signaling in RCC.


Asunto(s)
Carcinoma de Células Renales/genética , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica , Neoplasias Renales/genética , ARN Largo no Codificante/genética , Vía de Señalización Wnt/genética , Animales , Carcinoma de Células Renales/metabolismo , Carcinoma de Células Renales/patología , Línea Celular Tumoral , Movimiento Celular/genética , Ciclina D1/genética , Ciclina D1/metabolismo , Regulación hacia Abajo , Genes Supresores de Tumor , Humanos , Estimación de Kaplan-Meier , Neoplasias Renales/metabolismo , Neoplasias Renales/patología , Metaloproteinasas de la Matriz/genética , Metaloproteinasas de la Matriz/metabolismo , Ratones Endogámicos BALB C , Ratones Desnudos , Invasividad Neoplásica , Pronóstico , Trasplante Heterólogo
2.
Acta Biochim Biophys Sin (Shanghai) ; 46(4): 283-90, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24492534

RESUMEN

Carcinoembryonic antigen-related cell adhesion molecule 6 (CEACAM6) shows increased expression in a wide variety of human cancers, and its over-expression is associated with enhanced migration, invasion, and in vivo metastasis. Here, we reported that CEACAM6 was up-regulated in gastric cancer (GC) cell lines and tumor tissues. Over-expression of CEACAM6 in MKN-45 and SGC-7901 GC cells promoted migration and invasion in vitro and metastasis in athymic mice, whereas migration and invasion of MKN-28 and SNU-16 GC cells were suppressed by knockdown of CEACAM6. We also observed that steroid receptor coactivator (C-SRC) phosphorylation was increased when CEACAM6 was over-expressed in SGC-7901 cells. Taken together, these results suggested that CEACAM6 functions as an oncoprotein in GC and may be an important metastatic biomarker and therapeutic target.


Asunto(s)
Antígenos CD/fisiología , Moléculas de Adhesión Celular/fisiología , Invasividad Neoplásica/fisiopatología , Metástasis de la Neoplasia/fisiopatología , Neoplasias Gástricas/patología , Adulto , Anciano , Anciano de 80 o más Años , Animales , Antígenos CD/genética , Secuencia de Bases , Moléculas de Adhesión Celular/genética , Femenino , Proteínas Ligadas a GPI/genética , Proteínas Ligadas a GPI/fisiología , Técnicas de Silenciamiento del Gen , Humanos , Masculino , Ratones , Persona de Mediana Edad , Fosforilación , ARN Interferente Pequeño , Regulación hacia Arriba
3.
J Surg Res ; 185(1): 84-91, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23746766

RESUMEN

BACKGROUND: To evaluate the expression of chromodomain helicase/adenosine triphosphatase DNA binding protein 1-like gene (CHD1L) in colorectal carcinoma (CRC) and its clinical significance. Its oncogenic ability was also investigated. MATERIALS AND METHODS: CHD1L amplification and overexpression were detected by fluorescence in situ hybridization, real-time reverse transcriptase-polymerase chain reaction, and immunohistochemistry in 86 patients with CRC. The correlation between the clinical characteristics and prognosis was also determined. To evaluate the tumorigenic ability of CHD1L, it was cloned into expression vector pcDNA3.1(+) and transfected into CRC cell line SW1116. Next, the changes in the biologic behavior of the CRC cells, including cell proliferation, adhesion, migration, and invasion, were examined. Apoptosis and the cell cycle of the CRC cells were detected using flow cytometry. RESULTS: We have demonstrated that CHD1L is frequently amplified and overexpressed in CRC. Overexpression of CHD1L correlated with a large tumor size, deep tumor invasion, and a high histologic grade. It also conferred worse disease-free survival. CHD1L-transfected cells possessed a strong oncogenic ability, increasing the tumorigenicity in nude mice, which could be effectively suppressed by small interfering RNA against CHD1L. Functional studies showed that overexpression of CHD1L could promote G1/S-phase cells and inhibit apoptosis. CONCLUSIONS: Our results suggest that CHD1L is the target oncogene within the 1q21 amplicon and plays a pivotal role in CRC pathogenesis.


Asunto(s)
Apoptosis/genética , Carcinogénesis/genética , Neoplasias Colorrectales , ADN Helicasas/genética , ADN Helicasas/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Anciano , Animales , Movimiento Celular/genética , Proliferación Celular , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/mortalidad , Neoplasias Colorrectales/patología , Femenino , Humanos , Estimación de Kaplan-Meier , Masculino , Ratones , Persona de Mediana Edad , Células 3T3 NIH , Invasividad Neoplásica/genética , Invasividad Neoplásica/patología , Valor Predictivo de las Pruebas , Pronóstico , ARN Interferente Pequeño/genética , Tasa de Supervivencia
4.
Carcinogenesis ; 33(3): 555-62, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22223849

RESUMEN

A major hallmark of pancreatic ductal adenocarcinoma (PDAC) is extensive local tumor invasion and early systemic dissemination. DJ-1 has been shown to prevent cell death via the Akt pathway, thereby playing an important role in cancer progression and Parkinson's disease development. Here, we investigated the role of DJ-1 in tumor invasion and metastasis of pancreatic cancer and showed that DJ-1 is upregulated in 68.5% of pancreatic cancer specimens, correlated with tumor stage and predictive of short overall survival. Knockdown of DJ-1 expression in two PDAC cell lines reduced cell migration and invasion potential in vitro and inhibited metastasis in vivo. Knockdown of DJ-1 led to cytoskeleton disruption and diminished urokinase plasminogen activator (uPA) activity and expression, without affecting plasminogen activator inhibitor-1 and uPA receptor (uPAR) expression. All these effects were reversed by restoration of DJ-1 expression. In determining the pathway through which DJ-1 regulated cell migration and invasion, DJ-1 was found not to regulate Akt phosphorylation. Rather, it promoted extracellular signal-regulated kinase (ERK) and SRC phosphorylation. Inhibition of the ERK pathway in PDAC mimicked the effects of DJ-1 on cell migration, invasion, actin cytoskeleton and uPA/uPAR system and abolished the effects on promoting PDAC cell invasion and migration. These data represent the first identification of an important function of DJ-1, which is to regulate the invasion and metastasis properties of PDAC through the ERK/uPA cascade.


Asunto(s)
Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Oncogénicas/metabolismo , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo , Familia-src Quinasas/metabolismo , Citoesqueleto de Actina , Carcinoma Ductal Pancreático/genética , Línea Celular Tumoral , Movimiento Celular , Quinasas MAP Reguladas por Señal Extracelular/biosíntesis , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Invasividad Neoplásica , Metástasis de la Neoplasia , Estadificación de Neoplasias , Proteínas Oncogénicas/biosíntesis , Proteínas Oncogénicas/genética , Neoplasias Pancreáticas/genética , Fosforilación , Inhibidor 1 de Activador Plasminogénico/metabolismo , Proteína Desglicasa DJ-1 , Proteínas Proto-Oncogénicas c-akt/metabolismo , Interferencia de ARN , ARN Interferente Pequeño , Receptores del Activador de Plasminógeno Tipo Uroquinasa/biosíntesis , Receptores del Activador de Plasminógeno Tipo Uroquinasa/metabolismo , Activador de Plasminógeno de Tipo Uroquinasa/biosíntesis , Familia-src Quinasas/biosíntesis
5.
Int J Mol Sci ; 13(10): 13308-21, 2012 Oct 17.
Artículo en Inglés | MEDLINE | ID: mdl-23202954

RESUMEN

This study focuses on the association between the ABO blood group system and the risk of gastric cancer or Helicobacter pylori infection. The data for the ABO blood group was collected from 1045 cases of gastric cancer, whereby the patient underwent a gastrectomy in Ruijin Hospital, Shanghai. The information on the ABO blood group from 53,026 healthy blood donors was enrolled as control. We searched the Pubmed database on the relationship between ABO blood groups and gastric cancer risk for meta-analysis. In our case-control study, the risk of gastric cancer in blood group A was significantly higher than that in non-A groups (O, B and AB) (odd ratio, OR1.34; 95% confidential interval, CI 1.25-1.44). Compared with non-O groups (A, B and AB), individuals with blood group O demonstrated a reduced risk of gastric cancer (OR = 0.80; 95% CI 0.72-0.88). The proportion of H. pylori infection in blood group A individuals was significantly higher than that in non-A blood groups (OR = 1.42; 95% CI 1.05-1.93). We further combined our data with the published data of others, and crossreferenced the risk of gastric cancer with the blood type, finding consistent evidence that gastric cancer risk in the blood A group was higher than that in the non-A groups (OR = 1.11; 95% CI 1.07-1.15), and that blood type O individuals were consistently shown gastric cancer risk reduction (OR = 0.91; 95% CI 0.89-0.94). Our study concluded that there was a slightly increased risk of gastric cancer in blood group A individuals, and people with blood type A are more prone to be infected by H. pylori than other ABO blood type individuals, whereas, a slightly decreased risk of gastric cancer was identified in blood type O individuals.


Asunto(s)
Sistema del Grupo Sanguíneo ABO , Neoplasias Gástricas/patología , Estudios de Casos y Controles , Estudios de Cohortes , Infecciones por Helicobacter/patología , Helicobacter pylori , Humanos , Oportunidad Relativa , Factores de Riesgo , Neoplasias Gástricas/metabolismo
6.
Cancer Lett ; 545: 215829, 2022 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-35868534

RESUMEN

Acquired docetaxel (doc) resistance, one of the major reasons for unfavorable prognosis in patients with aggressive hormone-independent prostate cancer (HIPC), is a major obstacle for patient treatment. Dysregulation of long non-coding RNAs promotes or suppresses chemoresistance in multiple cancers; however, the specific molecular mechanisms underlying HIPC remain unknown. In this study, we found that the LINC01085, as a tumor-suppressor, which showed significant clinically relevant in HIPC patients with doc-resistance. Mechanistically, in docetaxel-sensitive cells, LINC01085 could specifically bind to both TANK-binding kinase 1 (TBK1) and glycogen synthase kinase 3ß (GSK3ß), and higher LINC01085 RNA levels could inhibit TBK1 dimerization. Whereas, in doc-resistant cells, lower LINC01085 RNA level lost the strong binding with both, meanwhile, the interaction between TBK1 and GSK3ß enhanced which accelerated TBK1 phosphorylation at the Ser-172 site, resulting in decreased expression levels of PD-L1 and NF-κB as well as the secretion of type I/III interferons. Thus, Overexpression of LINC01085 combined with immune checkpoint blockade is an effective strategy for the treatment of HIPC patients.


Asunto(s)
Interferón Tipo I , Neoplasias de la Próstata , Docetaxel/farmacología , Glucógeno Sintasa Quinasa 3 beta , Hormonas , Humanos , Inmunoterapia , Masculino , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/genética , ARN , Transducción de Señal
7.
Zhonghua Bing Li Xue Za Zhi ; 40(7): 475-9, 2011 Jul.
Artículo en Zh | MEDLINE | ID: mdl-22088375

RESUMEN

OBJECTIVE: To establish experimental models for tumor neovascularization and to apply quantitative digital imaging analysis in the study. METHODS: An endothelial tube formation model was established by human umbilical vein endothelial cells (HUVECs). A vasculogenic mimicry model was established by SGC-7901 gastric cancer cell line. Fertilized eggs were used to establish a chorioallantoic membrane angiogenesis model. Using gene transfection experiment, IRX1 tumor suppressor gene was chosen as a therapeutic target. Image Pro Plus (IPP) analysis software was used for digital vascular images analysis with parameters including points, lines, angles and integral absorbance (IA) for the tubular formation or vasculogenic mimicry. RESULTS: Digital image analysis by IPP showed that HUVEC tubular formation was significantly inhibited in IRX1 transfectant, compared with controls. The tubular numbers in three groups were 12.80 +/- 3.83, 29.00 +/- 5.34 and 28.20 +/- 4.32 (P<0.01). The connection points of tubules in three groups were 13.20 +/- 2.59, 25.00 +/- 2.24 and 24.60 +/- 3.21 (P<0.01). The tubular lengths of three groups were (821.5 +/- 12.5), (930.9 +/- 13.5) and (948.4 +/- 18.1) microm (P=0.022). The IA values of PAS stain in three groups were 3606 +/- 363, 14 200 +/- 1251 and 15 043 +/- 1220 (P<0.01). In chick chorioallantoic membrane model, the angular numbers of tubules in three groups were 6.41 +/- 2.60, 10.27 +/- 2.65 and 9.18 +/- 1.99 (P<0.01). CONCLUSIONS: The endothelial tube formation model, vasculogenic mimicry model and chorioallantoic membrane angiogenesis model are useful for gene therapy and drug screening with targeting neoplastic vascularization. Professional image analysis software may greatly facilitate the quantitative analysis of tumor neovascularization.


Asunto(s)
Diagnóstico por Imagen/métodos , Proteínas de Homeodominio/metabolismo , Neovascularización Patológica , Neovascularización Fisiológica , Neoplasias Gástricas/metabolismo , Factores de Transcripción/metabolismo , Animales , Línea Celular Tumoral , Células Cultivadas , Membrana Corioalantoides/irrigación sanguínea , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/fisiología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Programas Informáticos , Neoplasias Gástricas/genética , Neoplasias Gástricas/patología , Factores de Transcripción/genética , Factores de Transcripción/fisiología , Transfección
8.
Biochem Biophys Res Commun ; 398(1): 1-6, 2010 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-20510161

RESUMEN

Deregulation of E2F1 activity is characteristic of gastric tumorigenesis, which involves in complex molecular mechanisms. microRNA is one of the post-transcriptional regulators for gene expression. Here, we report a member of miR-331 family, miR-331-3p, which was decreased in some kinds of malignancies. However, the biological function of miR-331-3p on gastric cancer is largely unknown. In this study, we screened the expressing levels of miR-331-3p and E2F1 in gastric cancer cell lines. We transfected precursor or inhibitor of miR-331-3p into gastric cancer cells. As results, miR-331-3p is down-regulated in all gastric cancer cell lines by real-time PCR. Over-expression of miR-331-3p blocked G1/S transition on SGC-7901 and AGS cell lines. Introduction of miR-331-3p dramatically suppressed the ability of colony formation and cell growth in vitro by interfering E2F1 activity. Our data highlight an important role of miR-331-3p in cell cycle control by targeting 3'-UTR of cell cycle-related molecule E2F1. We concluded that miR-331-3p is a potential tumor suppressor in gastric cancer. Restoring miR-331-3p in gastric cancer cells revealed potential application in gastric cancer therapy.


Asunto(s)
Proliferación Celular , Factor de Transcripción E2F1/genética , Regulación Neoplásica de la Expresión Génica , MicroARNs/metabolismo , Neoplasias Gástricas/patología , Regiones no Traducidas 3'/genética , Secuencia de Bases , Línea Celular Tumoral , Humanos , MicroARNs/antagonistas & inhibidores , MicroARNs/genética , Neoplasias Gástricas/genética
9.
Hepatogastroenterology ; 57(102-103): 1106-12, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-21410040

RESUMEN

BACKGROUND/AIMS: The acquisition of resistance to 5-FU is one of the most prominent obstacles to successful chemotherapy, and the mechanisms underlying the resistance are not fully understood. The aim of this study is to identify novel mediators of 5-FU resistance in colon cancer cells. METHODOLOGY: LoVo colon cancer cells were induced to 5-FU resistance in vitro. The global protein profiles between LoVo and its 5-FU resistant derivative cell line LoVo/5-FU were analyzed by two dimensional gel electrophoresis-based comparative proteomics. The identified proteins expression was confirmed by Western blot analysis. The cytotoxicity of 5-FU was measured in LoVo/5-FU after knockdown of RhoGDI2 (one of the identified protien). RESULTS: Three differentially expressed proteins were identified. RhoGDI2 and CapG were upregulated, whereas proapoptotic protein Maspin was down-regulated in LoVo/5-FU and validated by Western blotting. Furthermore, knockdown of RhoGDI2 expression by transfection with the RhoGDI2-specific siRNA significantly reduced the resistance to 5-FU in LoVo/5-FU (p < 0.05). CONCLUSIONS: These novel data suggest that these differentially expressed proteins may contribute to the development of 5-FU resistance in colon cancer cells.


Asunto(s)
Antimetabolitos Antineoplásicos/farmacología , Neoplasias del Colon/tratamiento farmacológico , Fluorouracilo/farmacología , Inhibidores de Disociación de Guanina Nucleótido/fisiología , Proteínas Supresoras de Tumor/fisiología , Secuencia de Aminoácidos , Línea Celular Tumoral , Resistencia a Antineoplásicos , Inhibidores de Disociación de Guanina Nucleótido/antagonistas & inhibidores , Humanos , Datos de Secuencia Molecular , ARN Interferente Pequeño/genética , Proteínas Supresoras de Tumor/antagonistas & inhibidores , Inhibidor beta de Disociación del Nucleótido Guanina rho , Inhibidores de la Disociación del Nucleótido Guanina rho-Específico
10.
Zhonghua Bing Li Xue Za Zhi ; 39(7): 436-41, 2010 Jul.
Artículo en Zh | MEDLINE | ID: mdl-21055170

RESUMEN

OBJECTIVE: To analyze microarray datasets deposited in the public database and to identify TNM associated genes in gastric cancers. METHODS: Microarray datasets of gastric cancer were selected from GEO database. Differentially expressed genes related to TNM staging were evaluated by significant analysis of the microarray using MultiExperiment Viewer (MEV) platform. Candidate gene expressions in gastric cancer tissues and cell lines were verified by reverse transcriptase polymerase chain reaction (RT-PCR), quantitative RT-PCR, Western blot and immunohistochemistry. RESULTS: GES4007 dataset was re-analyzed leading to the identification of 14 genes associated with TNM staging. Over-expression of matrix gla protein (MGP) was confirmed in gastric cancer cell lines and tumor tissues by quantitative RT-PCR, Western blot and immunohistochemistry. Increased MGP expression was found in 22 of 54 cases of (40.7%) gastric cancer specimens compared to the normal gastric tissues. The up-regulation of MGP mRNA expression closely correlated with TNM stage (P = 0.001) and prognosis (P = 0.006). CONCLUSIONS: Public databases of microarray studies are the valuable resources for data mining. MGP has been identified and confirmed as a novel biomarker for the TNM stage and prognosis of gastric cancer.


Asunto(s)
Proteínas de Unión al Calcio/biosíntesis , Proteínas de la Matriz Extracelular/biosíntesis , Perfilación de la Expresión Génica , Estadificación de Neoplasias , Análisis de Secuencia por Matrices de Oligonucleótidos , Neoplasias Gástricas/metabolismo , Adulto , Anciano , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Proteínas de Unión al Calcio/genética , Línea Celular Tumoral , Proteínas de la Matriz Extracelular/genética , Femenino , Estudios de Seguimiento , Regulación Neoplásica de la Expresión Génica , Humanos , Metástasis Linfática , Masculino , Persona de Mediana Edad , ARN Mensajero/metabolismo , Neoplasias Gástricas/genética , Neoplasias Gástricas/patología , Tasa de Supervivencia , Regulación hacia Arriba , Adulto Joven , Proteína Gla de la Matriz
12.
Free Radic Biol Med ; 93: 165-76, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26855420

RESUMEN

Mitochondrial aldehyde dehydrogenase 2 (ALDH2) is a member of the aldehyde dehydrogenase superfamily and is involved with the metabolic processing of aldehydes. ALDH2 plays a cytoprotective role by removing aldehydes produced during normal metabolism. We examined the cytoprotective role of ALDH2 specifically in gastric mucosa cells. Overexpression of ALDH2 increased the viability of gastric mucosa cells treated with H2O2, while knockdown of ALDH2 had an opposite effect. Moreover, overexpression of ALDH2 protected gastric mucosa cells against oxidative stress-induced apoptosis as determined by flow cytometry, Hoechst 33342, and TUNEL assays. Consistently, ALDH2 knockdown had an opposite effect. Additionally, DNA damage was ameliorated in ALDH2-overexpressing gastric mucosa cells treated with H2O2. We further identified that this cytoprotective role of ALDH2 was mediated by metabolism of 4-hydroxynonenal (4-HNE). Consistently, 4-HNE mimicked the oxidative stress induced by H2O2 in gastric mucosa cells. Treatment with 4-HNE increased levels of DNA damage in ALDH2-knockdown GES-1 cells, while overexpression of ALDH2 decreased 4-HNE-induced DNA damage. These findings suggest that ALDH2 can protect gastric mucosa cells against DNA damage caused by oxidative stress by reducing levels of 4-HNE.


Asunto(s)
Aldehído Deshidrogenasa Mitocondrial/genética , Daño del ADN/efectos de los fármacos , Mucosa Gástrica/enzimología , Estrés Oxidativo/genética , Aldehído Deshidrogenasa Mitocondrial/metabolismo , Aldehídos/metabolismo , Apoptosis/efectos de los fármacos , Línea Celular , Supervivencia Celular/efectos de los fármacos , Humanos , Peróxido de Hidrógeno/farmacología , Mitocondrias/efectos de los fármacos , Mitocondrias/enzimología
13.
Oncotarget ; 7(52): 87496-87510, 2016 Dec 27.
Artículo en Inglés | MEDLINE | ID: mdl-27589838

RESUMEN

Early diagnosis of gastric cancer is crucial to improve patient' outcome. A good biomarker will function in early diagnosis for gastric cancer. In order to find practical and cost-effective biomarkers, we used gas chromatography combined mass spectrometer (GC-MS) to profile urinary metabolites on 293 urine samples. Ninety-four samples are taken as training set, others for validating study. Orthogonal partial least squares discriminant analysis (OPLS-DA), significance analysis of microarray (SAM) and Mann-Whitney U test are used for data analysis. The diagnostic value of urinary metabolites was evaluated by ROC curve. As results, Seventeen metabolites are significantly different between patients and healthy controls in training set. Among them, 14 metabolites show diagnostic value better than classic blood biomarkers by quantitative assay on validation set. Ten of them are amino acids and four are organic metabolites. Importantly, proline, p-cresol and 4-hydroxybenzoic acid disclose outcome-prediction value by means of survival analysis. Therefore, the examination of urinary metabolites is a promising noninvasive strategy for gastric cancer screening.


Asunto(s)
Cromatografía de Gases y Espectrometría de Masas/métodos , Ensayos Analíticos de Alto Rendimiento/métodos , Neoplasias Gástricas/diagnóstico , Adulto , Anciano , Anciano de 80 o más Años , Aminoácidos/orina , Biomarcadores de Tumor/orina , Detección Precoz del Cáncer , Femenino , Humanos , Masculino , Redes y Vías Metabólicas , Persona de Mediana Edad , Curva ROC , Neoplasias Gástricas/mortalidad , Neoplasias Gástricas/orina
14.
Sci Rep ; 5: 9604, 2015 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-25853231

RESUMEN

Inactivation of p53 and/or Rb pathways restrains osteoblasts from cell-cycle exit and terminal differentiation, which underpins osteosarcoma formation coupled with dedifferentiation. Recently, the level of p-S6K was shown to independently predict the prognosis for osteosarcomas, while the reason behind this is not understood. Here we show that in certain high-grade osteosarcomas, immature SSEA-4(+) tumor cells represent a subset of tumor-initiating cells (TICs) whose pool size is maintained by mTORC1 activity. mTORC1 supports not only SSEA-4(+) cell self-renewal through S6K but also the regeneration of SSEA-4(+) TICs by SSEA-4(-) osteosarcoma cell dedifferentiation. Mechanistically, active mTORC1 is required to prevent a likely upregulation of the cell-cycle inhibitor p27 independently of p53 or Rb activation, which otherwise effectively drives the terminal differentiation of SSEA-4(-) osteosarcoma cells at the expense of dedifferentiation. Thus, mTORC1 is shown to critically regulate the retention of tumorigenicity versus differentiation in discrete differentiation phases in SSEA-4(+) TICs and their progeny.


Asunto(s)
Neoplasias Óseas/metabolismo , Neoplasias Óseas/patología , Transformación Celular Neoplásica/metabolismo , Complejos Multiproteicos/metabolismo , Osteosarcoma/metabolismo , Osteosarcoma/patología , Antígenos Embrionarios Específico de Estadio/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Animales , Neoplasias Óseas/tratamiento farmacológico , Neoplasias Óseas/genética , Neoplasias Óseas/mortalidad , Ciclo Celular/genética , Desdiferenciación Celular , Transformación Celular Neoplásica/genética , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Transición Epitelial-Mesenquimal/genética , Femenino , Xenoinjertos , Humanos , Diana Mecanicista del Complejo 1 de la Rapamicina , Complejos Multiproteicos/genética , Clasificación del Tumor , Osteosarcoma/tratamiento farmacológico , Osteosarcoma/genética , Osteosarcoma/mortalidad , Pronóstico , Proteína de Retinoblastoma/genética , Proteína de Retinoblastoma/metabolismo , Antígenos Embrionarios Específico de Estadio/genética , Serina-Treonina Quinasas TOR/genética , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
15.
Oncotarget ; 6(3): 1652-65, 2015 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-25544763

RESUMEN

Claudin-1 (CLDN1) is overexpressed in gastric cancer and correlated with tumor invasion, metastasis and poor outcome. Here, we both down and up regulated CLDN1 expression in gastric cancer cells to elucidate its role in gastric carcinogenesis and tumor progression. We found that deficiency of CLDN1 inhibited cells migration, invasion, and colony formation in vitro and tumorigenicity, metastasis in vivo. Also, CLDN1 promoted cell aggregation and increased anoikis resistance. Down or up regulation of CLDN1 was accompanied with changes of membrane ß-catenin expression as well as Akt and Src activities. When ß-catenin was up-regulated in CLDN1-KD cells, cell aggregation and anoikis resistance were restored, and Akt and Src signal pathways were re-activated. Taken together, these findings suggest that CLDN1 is oncogenic in gastric cancer and its malignant potential may be attributed in part to regulation of anoikis, by mediating membrane ß-catenin-regulated cell-cell adhesion and cell survival.


Asunto(s)
Anoicis/fisiología , Claudina-1/biosíntesis , Neoplasias Gástricas/metabolismo , Animales , Línea Celular Tumoral , Movimiento Celular/fisiología , Proliferación Celular/fisiología , Claudina-1/genética , Regulación hacia Abajo , Técnicas de Silenciamiento del Gen , Xenoinjertos , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Metástasis de la Neoplasia , Transducción de Señal , Neoplasias Gástricas/genética , Neoplasias Gástricas/patología , Regulación hacia Arriba , beta Catenina/biosíntesis
16.
Int J Oncol ; 44(4): 1293-301, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24535143

RESUMEN

Claudin 1 is one of the tight junction proteins, which are critical in the maintenance of epithelial integrity. Aberrant regulation of CLDN1 and its correlation with ß-catenin have been discovered in malignant tumors. The present study aimed to investigate the expression profile and clinical relevance of CLDN1 and ß-catenin. The protein levels of CLDN1 and ß-catenin were examined using immunohistochemical staining. The characteristics of expression profile and prognostic value were analyzed using Pearson's χ² test and Kaplan-Meier analysis, respectively. ß-catenin overexpression and knockdown were used to investigate its role in regulating CLDN1 expression. We showed that CLDN1 was overexpressed in intestinal-type, presence of lymph node metastasis, higher TNM stage in gastric cancer patients and correlated with decreased overall survival. The characteristics of CLDN1 expression were associated with that of ß-catenin. CLDN1 and ß-catenin showed similar prognostic value in intestinal-type gastric cancers. ß-catenin knockdown and overexpression in cell models revealed a positive relation between CLDN1 and ß-catenin. Our study demonstrated that CLDN1 is a biomarker for intestinal-type gastric cancer with shorter survival. The expression of CLDN1 was strongly associated with ß-catenin in gastric cancer patients and a gastric cancer cell model.


Asunto(s)
Claudina-1/biosíntesis , Neoplasias Gástricas/genética , beta Catenina/biosíntesis , Anciano , Biomarcadores de Tumor , Línea Celular Tumoral , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Metástasis Linfática , Masculino , Interferencia de ARN , ARN Interferente Pequeño , Neoplasias Gástricas/mortalidad , Resultado del Tratamiento , beta Catenina/genética
17.
Int J Oncol ; 44(5): 1512-20, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24604109

RESUMEN

SOX11 is involved in gastrulation and in malignant diseases. The aim of this study was to investigate the role of SOX11 in gastric cancer and its expression pattern and clinical significance. SOX11 overexpression cell model was used to examine in vitro and in vivo the role of SOX11 in cell growth and metastasis. Cell cycle analysis and Annexin V/PI double staining were used to investigate the effect of SOX11 on cell cycle progression and apoptosis. The expression of SOX11 in human gastric cancer was examined by immunohistochemistry. The correlation of SOX11 expression with clinicopathological characteristics and survival of patients was analyzed by Pearson's χ(2) and Kaplan-Meier analyses, respectively. Cox's proportional hazard model was employed in multivariate analysis. SOX11 overexpression did not inhibit cell growth but strongly suppressed cell migration/invasion in vitro and in vivo. We found a significant correlation between high SOX11 protein levels and Lauren's classification (intestinal type), differentiation status (high and medium), and early TNM stage. SOX11 is an independent prognostic factor for improved survival in gastric cancer patients. SOX11 was a potential tumor-suppressor and an independent positive prognostic factor in gastric cancer patients with less advanced clinicopathological features.


Asunto(s)
Metástasis de la Neoplasia/patología , Factores de Transcripción SOXC/genética , Factores de Transcripción SOXC/metabolismo , Neoplasias Gástricas/genética , Adulto , Anciano , Anciano de 80 o más Años , Animales , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Ciclo Celular , Diferenciación Celular , Línea Celular Tumoral , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Ratones , Ratones Desnudos , Persona de Mediana Edad , Invasividad Neoplásica/genética , Invasividad Neoplásica/patología , Metástasis de la Neoplasia/genética , Estadificación de Neoplasias , Neoplasias Experimentales , Neoplasias Gástricas/patología
18.
Gene ; 527(1): 102-8, 2013 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-23769968

RESUMEN

HIC1 is a tumor suppressor gene that is down-expressed in different malignancies, in part, because of promoter hypermethylation. However, the biological function of HIC1 in gastric cancer remains unclear. It is known that small double-stranded RNAs can induce gene expression by targeting promoter sequences. In the present study, we examined the expression levels of HIC1 in gastric cancer tissue. Several pieces of small double-stranded RNAs were used for the activation of HIC1. Tissue microarray analysis of gastric cancer indicated that down-regulation of HIC1 in gastric cancer tissue was dramatic compared with the adjacent gastric mucosa. Gastric cancer cell lines also showed down-regulated HIC1 expression compared with a human immortalized gastric mucosa cell line. One out of four dsRNAs produced activation of HIC1 as assessed by real-time PCR and Western blotting. Use of a cell counting kit 8 and clonogenicity assays indicated that dsRNA-mediated re-expression of HIC1 inhibited cell proliferation and clonogenicity in gastric cancer. Reactivation of HIC1 suppressed cell migration and induced cell cycle arrest in the G0/G1 phase, as well as induced apoptosis. These results suggest that HIC1 is a potential target of gene therapy against gastric cancer, and that dsRNAs could function as a therapeutic option for up-regulating tumor suppressor genes in gastric cancer and other malignancies.


Asunto(s)
Mucosa Gástrica/metabolismo , Factores de Transcripción de Tipo Kruppel/genética , ARN Pequeño no Traducido/genética , Neoplasias Gástricas/metabolismo , Apoptosis , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Regulación hacia Abajo , Femenino , Puntos de Control de la Fase G1 del Ciclo Celular , Regulación Neoplásica de la Expresión Génica , Humanos , Factores de Transcripción de Tipo Kruppel/metabolismo , Masculino , Persona de Mediana Edad , Neoplasias Gástricas/genética , Análisis de Matrices Tisulares , Activación Transcripcional
19.
Anticancer Agents Med Chem ; 13(2): 270-5, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22934698

RESUMEN

MicroRNAs (miRNAs) and promoter hypermethylation are vital epigenetic mechanisms for transcriptional inactivation of tumor suppressor. IRX1 is a newly identified tumor suppressor gene and hypermethylation involves the decreased expression in gastric cancer. However, the microRNA regulatory mechanism on IRX1 expression is still unclear. In this study, we report an IRX1-targeting miRNA-544, which directly targets 3'-UTR of IRX1 gene by luciferase reporter assay. miR-544 suppresses the protein expression of IRX1 gene by Western blot and immunocytochemistry. Ectopic expression of miR-544 promotes cell proliferation and cell cycle progression significantly in vitro on gastric cancer cells. The study suggests that miR-544 is an oncogenic microRNA in gastric cancer. Over expression of miR-544 contributes to the inactivation and low-expression of IRX1 in gastric cancer. These findings are helpful for clarifying the molecular mechanisms involved in gastric carcinogenesis and indicate that miR-544 is a key regulator in switching cell cycle on or off. miR-544 may be a potential molecular target in miRNA-based strategy on gastric cancer.


Asunto(s)
MicroARNs/genética , Terapia Molecular Dirigida , Neoplasias Gástricas/genética , Ciclo Celular , Línea Celular , Proliferación Celular , Regulación Neoplásica de la Expresión Génica/genética , Humanos , ARN Neoplásico/genética , Neoplasias Gástricas/patología
20.
Oncol Lett ; 5(1): 255-260, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23255931

RESUMEN

Resistance to 5-fluorouracil (5-FU) in patients with gastric cancer is a serious therapeutic problem and major efforts are underway to understand the underlying mechanisms. We have previously identified RhoGDI2 as a contributor to 5-FU resistance in colon cancer cells using 2D electrophoresis and mass spectrometry and the current study aimed to further investigate this role. The expression of RhoGDI2 in seven gastric cancer cell lines was positively correlated with resistance to 5-FU. Lower 5-FU sensitivity of isolated tumor cells from patients with gastric cancer was also associated with higher RhoGDI2 expression. Ectopic expression of RhoGDI2 in gastric cancer cells increased the resistance to 5-FU and reverted low dose 5-FU-induced G2/M phase arrest without affecting the population of sub-G1 cells. Overall, these findings suggest that RhoGDI2 is associated with 5-FU resistance and is a potential therapeutic target for enhancing chemotherapy efficacy in gastric cancer.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA