Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Mol Carcinog ; 57(7): 886-895, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29566278

RESUMEN

POTE ankyrin domain family, member G (poteg) belongs to POTE family. The POTE family is composed of many proteins which are very closely related and expressed in prostate, ovary, testis, and placenta. Some POTE paralogs are related with some cancers. Here we showed that down-regulation of POTEG was detected in about 60% primary esophageal squamous cell carcinoma (ESCC) tumor tissues. Clinical association studies determined that POTEG down-regulation was significantly correlated with tumor differentiation, lymph nodes metastasis and TNM staging. Kaplan-Meier analysis determined that POTEG down-regulation was associated with poorer clinical outcomes of ESCC patients (P = 0.026). Functional studies showed that POTEG overexpression could suppress tumor cell growth and metastasis capacity in vitro and in vivo. Molecular analyses revealed that POTEG downregulated CDKs, leading to subsequent inhibition of Rb phosphorylation, and consequently arrested Cell Cycle at G1/S Checkpoint. POTEG overexpression induced apoptosis by activating caspases and PARP, and regulating canonical mitochondrial apoptotic pathways. On the other side, POTEG inhibited epithelial-mesenchymal transition and suppressed tumor cell metastasis. In conclusion, our study reveals a functionally important control mechanism of POTEG in esophageal cancer pathogenesis, suggesting potential use in the ESCC intervention and therapeutic strategies.


Asunto(s)
Regulación hacia Abajo/genética , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/patología , Carcinoma de Células Escamosas de Esófago/genética , Carcinoma de Células Escamosas de Esófago/patología , Proteínas/genética , Animales , Apoptosis/genética , Diferenciación Celular/genética , Movimiento Celular/genética , Proliferación Celular/genética , Transición Epitelial-Mesenquimal/genética , Puntos de Control de la Fase G1 del Ciclo Celular/genética , Humanos , Estimación de Kaplan-Meier , Metástasis Linfática/genética , Metástasis Linfática/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Estadificación de Neoplasias/métodos , Pronóstico
2.
BMC Cancer ; 16: 669, 2016 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-27549330

RESUMEN

BACKGROUND: Nasopharyngeal carcinoma (NPC) is a type of head-neck cancer with a distinguishable geographic and racial distribution worldwide. Increasing evidence supports that the accumulation of additional genetic and epigenetic abnormalities is important in driving the NPC tumorigenic process. In this study, we aim to investigate the association between EIF5A2 (Eukaryotic translation initiation factor 5A2) expression status and NPC clinical outcomes. METHODS: The expression status of EIF5A2 was investigated in the NPC tissue microarray. Tissues were from 166 NPC patients staging II-IV, collected between 1999 and 2005. All patients were administered 2-3 cycles of DDP (cisplatin) + 5-Fu (5-fluorouracil) induction therapy and then treated with a uniform conventional two-dimensional radiotherapy. Cell motility assay, tumor growth assay and cytotoxicity assay were performed on the EIF5A2 overexpressed cells and control cells. siRNA was also used in the in vitro studies. RESULTS: Positive staining of EIF5A2 was observed in 85.4 % (105/123) informative tumor cases. Multivariate analyses demonstrated that EIF5A2 was an independent prognostic marker of poor overall survival (OS) (P = 0.041), failure-free survival (FFS) (P = 0.029), and distant failure-free survival (D-FFS) (P = 0.043) in patients with locoregionally advanced NPC patients treated with cisplatin + 5-Fu chemoradiotherapy. The forced expression of EIF5A2 in NPC cells enhanced the cells' motility and growth ability. Knock-down of EIF5A2 in NPC cells decreased the cell's motility and growth ability. Our results also demonstrated that EIF5A2 overexpression induced chemoresistance of NPC cells to 5-Fu. CONCLUSIONS: Our findings suggested that EIF5A2 expression, as examined by immunohistochemistry, could function as an independent prognostic factor of outcomes in NPC patients with cisplatin + 5-Fu chemoradiotherapy. EIF5A2 might be a novel therapeutic target for the inhibition of NPC progress.


Asunto(s)
Carcinoma/tratamiento farmacológico , Carcinoma/mortalidad , Quimioterapia de Inducción/métodos , Neoplasias Nasofaríngeas/tratamiento farmacológico , Neoplasias Nasofaríngeas/mortalidad , Factores de Iniciación de Péptidos/biosíntesis , Proteínas de Unión al ARN/biosíntesis , Adulto , Anciano , Biomarcadores de Tumor/genética , Carcinoma/genética , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular , Quimioradioterapia/métodos , Cisplatino/uso terapéutico , Femenino , Fluorouracilo/uso terapéutico , Humanos , Masculino , Persona de Mediana Edad , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas/genética , Factores de Iniciación de Péptidos/genética , Interferencia de ARN , ARN Interferente Pequeño/genética , Proteínas de Unión al ARN/genética , Adulto Joven , Factor 5A Eucariótico de Iniciación de Traducción
3.
Gastroenterology ; 146(7): 1701-13.e9, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24561231

RESUMEN

BACKGROUND & AIMS: Solid tumors often become hypoxic, leading to activation of hypoxia-response genes. We investigated the effects of overexpression of the hypoxia response genes eIF5A2 in esophageal squamous cell carcinoma (ESCC). METHODS: We used quantitative real-time polymerase chain reaction and immunohistochemistry analyses to compare expression of eIF5A2 between paired ESCC samples and nontumor esophageal tissues, and fluorescence in situ hybridization to detect gene copy-number alterations. Luciferase reporter and chromatin immunoprecipitation assays were used to study interactions between eIF5A2 and hypoxia-inducible factor-1α (HIF1α). We determined the effects of eIF5A2 overexpression and knockdown in ESCC cell lines and growth of ESCC xenograft tumors in nude mice. RESULTS: Levels of eIF5A2 messenger RNA and protein were increased in >40% of ESCC samples compared with matched nontumor tissues, along with levels of HIF1α and vascular endothelial growth factor. Increased levels of EIF5A2 were significantly associated with ESCC metastasis to lymph nodes (P < .001) and tissue invasion (P = .037), and shorter survival times of patients (P < .001). Amplification of eIF5A2 was detected in 35.14% of ESCC samples that overexpressed eIF5A2. Hypoxia increased expression of eIF5A2 4- to 8-fold in ESCC cell lines; we observed bidirectional regulation between eIF5A2 and HIF1α. Transient transfection of ESCC cell lines with eIF5A2 increased their migratory and invasive abilities and markers of the epithelial to mesenchymal transition, and eIF5A2 knockdown or HIFα inhibition reduced these. In mice, xenograft tumors grown from ESCC cells that expressed eIF5A2 formed tumors more rapidly than cells that expressed only vector (controls); they also expressed higher levels of HIF1α and vascular endothelial growth factor, and formed more microvessels than controls. Knockdown of eIF5A2 in ESCC cells with interfering RNAs reduced their growth as xenograft tumors in mice, particularly when mice were given docetaxel or cisplatin. CONCLUSIONS: eIF5A2 is overexpressed by gene amplification or hypoxia in ESCCs, and associated with up-regulation of HIF1α, metastasis, and shorter survival times of patients. Increased expression of eIF5A2 increases metastasis and angiogenesis in ESCC via the HIF1α-mediated signaling pathway.


Asunto(s)
Carcinoma de Células Escamosas/metabolismo , Movimiento Celular , Neoplasias Esofágicas/metabolismo , Amplificación de Genes , Neovascularización Patológica , Factores de Iniciación de Péptidos/metabolismo , Proteínas de Unión al ARN/metabolismo , Animales , Antineoplásicos/farmacología , Carcinoma de Células Escamosas/irrigación sanguínea , Carcinoma de Células Escamosas/tratamiento farmacológico , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/mortalidad , Carcinoma de Células Escamosas/secundario , Hipoxia de la Célula , Línea Celular Tumoral , Cisplatino/farmacología , Docetaxel , Transición Epitelial-Mesenquimal , Neoplasias Esofágicas/irrigación sanguínea , Neoplasias Esofágicas/tratamiento farmacológico , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/mortalidad , Neoplasias Esofágicas/patología , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Estimación de Kaplan-Meier , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Persona de Mediana Edad , Invasividad Neoplásica , Factores de Iniciación de Péptidos/genética , Modelos de Riesgos Proporcionales , Unión Proteica , Interferencia de ARN , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/genética , Transducción de Señal , Taxoides/farmacología , Factores de Tiempo , Transfección , Regulación hacia Arriba , Factor A de Crecimiento Endotelial Vascular/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto , Factor 5A Eucariótico de Iniciación de Traducción
4.
Theranostics ; 9(3): 796-810, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30809309

RESUMEN

Background and Aims: Cancer cells prefer aerobic glycolysis to maintain growth advantages, but the role of long non-coding RNAs (lncRNAs) in glycometabolism still remains unclear. Here we identified one cytoplasmic lncRNA LINC01554 as a significantly downregulated lncRNA in hepatocellular carcinoma (HCC) and aimed to investigate its role in cellular glucose metabolism in the development and progression of HCC. Methods: Quantitative real-time PCR was used to determine the expression level of LINC01554. Downregulation of LINC01554 by miR-365a at transcriptional level was assessed by luciferase reporter assay. Subcellular fractionation assay and RNA fluorescence in situ hybridization were performed to detect the subcellular localization of LINC01554. RNA pull-down assay, mass spectrometry, and RNA immunoprecipitation assay were used to identify the underlying molecular mechanisms. The tumor-suppressive function of LINC01554 was determined by both in vitro assay and nude mice xenograft model. Results: LINC01554 was frequently downregulated in HCC, which was significantly associated with tumor invasion (P = 0.005), tumor size (P = 0.041), tumor staging (P = 0.023) and shorter survival (P = 0.035) of HCC patients. Luciferase reporter assay unraveled that LINC01554 was negatively regulated by miR-365a. Subcellular fractionation assay and RNA FISH revealed the cytoplasmic predominance of LINC01554 in MIHA cells and HCC clinical samples. Ectopic expression of LINC01554 inhibited HCC cell growth, colony formation in soft agar, foci formation, and tumor formation in nude mice. LINC01554 promoted the ubiquitin-mediated degradation of PKM2 and inhibited Akt/mTOR signaling pathway to abolish aerobic glycolysis in HCC cells. Further study found that LINC01554-knockout could effectively reverse the tumor-suppressive effect of LINC01554. Conclusions: Our results identify LINC01554 as a novel tumor suppressor in HCC and unravel its underlying molecular mechanism in reprogramming cellular glucose metabolism. LINC01554 could possibly serve as a novel prognostic biomarker and provide the rationale for HCC therapy.


Asunto(s)
Carcinoma Hepatocelular/metabolismo , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Glucosa/metabolismo , Neoplasias Hepáticas/metabolismo , Piruvato Quinasa/genética , ARN Largo no Codificante/genética , Animales , Carcinoma Hepatocelular/genética , Regulación hacia Abajo , Femenino , Humanos , Neoplasias Hepáticas/genética , Masculino , Ratones Endogámicos BALB C , MicroARNs/metabolismo , Persona de Mediana Edad , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal , Serina-Treonina Quinasas TOR/antagonistas & inhibidores
5.
Cancer Res ; 78(16): 4471-4481, 2018 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-29967258

RESUMEN

Glucose metabolic reprogramming from oxidative phosphorylation to glycolysis is one of the hallmarks of cancer development. Coenzyme Q10 (CoQ10) is essential for electron transport in the mitochondrial respiratory chain and for antioxidant defense. Here, we investigated the role of a key factor in CoQ10 synthesis, prenyldiphosphate synthase subunit 2 (PDSS2), in hepatocellular carcinoma (HCC) tumorigenesis. PDSS2 was frequently downregulated in HCC tissues and was significantly associated with poorer HCC prognosis (P = 0.027). PDSS2 downregulation was a prognostic factor independent of T status and stage (P = 0.028). Downregulation of CoQ10 was significantly correlated with downregulation of PDSS2 in HCC tumor tissues (R = 0.414; P < 0.001). Of the six different splicing isoforms of PDSS2, the five variants other than full-length PDSS2 showed loss of function in HCC. Reintroduction of full-length PDSS2 into HCC cells increased CoQ10 and mitochondrial electron transport complex I activity and subsequently induced a metabolic shift from aerobic glycolysis to mitochondrial respiration in cells. Reintroduction of PDSS2 also inhibited foci formation, colony formation in soft agar, and tumor formation in nude mice. Knockdown of PDSS2 induced chromosomal instability in the MIHA immortalized human liver cell line. Furthermore, knockdown of PDSS2 in MIHA induced malignant transformation. Overall, our findings indicate that PDSS2 deficiency might be a novel driving factor in HCC development.Significance: Downregulation of PDSS2 is a driving factor in hepatocellular carcinoma tumorigenesis. Cancer Res; 78(16); 4471-81. ©2018 AACR.


Asunto(s)
Transferasas Alquil y Aril/genética , Carcinoma Hepatocelular/genética , Neoplasias Hepáticas/genética , Mitocondrias/genética , Animales , Biomarcadores de Tumor/genética , Carcinogénesis/genética , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Reprogramación Celular/genética , Regulación Neoplásica de la Expresión Génica , Glucosa/metabolismo , Glucólisis/genética , Humanos , Hígado/metabolismo , Hígado/patología , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Ratones , Mitocondrias/metabolismo , Mitocondrias/patología , Óxido Nítrico Sintasa/genética , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Oncotarget ; 6(28): 26079-89, 2015 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-26317793

RESUMEN

Esophageal squamous cell carcinoma (ESCC) is one of the most lethal malignancies of the digestive tract in East Asian countries. Multimodal therapies, including adjuvant chemotherapy and neo-adjuvant chemotherapy, have become more often used for patients with advanced ESCC. However, the chemotherapy effect is often limited by patients' drug resistance. This study demonstrated that EIF5A2 (eukaryotic translation initiation factor 5A2) overexpression induced stemness and chemoresistance in ESCC cells. We showed that EIF5A2 overexpression in ESCC cells resulted in increased chemoresistance to 5-fluorouracil (5-FU), docetaxel and taxol. In contrast, shRNAs suppressing eIF5A2 increased tumor sensitivity to these chemotherapeutic drugs. In addition, EIF5A2 overexpression was correlated with a poorer overall survival in patients with ESCC who underwent taxane-based chemotherapy after esophagectomy (P < 0.05). Based on these results, we suggest that EIF5A2 could be a predictive biomarker for selecting appropriate chemo-treatment for ESCC patients and EIF5A2 inhibitors might be considered as combination therapy to enhance chemosensitivity in patients with ESCC.


Asunto(s)
Carcinoma de Células Escamosas/genética , Resistencia a Antineoplásicos/genética , Neoplasias Esofágicas/genética , Regulación Neoplásica de la Expresión Génica , Células Madre Neoplásicas/metabolismo , Factores de Iniciación de Péptidos/genética , Proteínas de Unión al ARN/genética , Adulto , Anciano , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Apoptosis/genética , Western Blotting , Carcinoma de Células Escamosas/tratamiento farmacológico , Carcinoma de Células Escamosas/cirugía , Línea Celular Tumoral , Docetaxel , Resistencia a Antineoplásicos/efectos de los fármacos , Neoplasias Esofágicas/tratamiento farmacológico , Neoplasias Esofágicas/cirugía , Femenino , Fluorouracilo/farmacología , Humanos , Inmunohistoquímica , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Células Madre Neoplásicas/efectos de los fármacos , Paclitaxel/farmacología , Factores de Iniciación de Péptidos/metabolismo , Proteínas de Unión al ARN/metabolismo , Estudios Retrospectivos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Taxoides/farmacología , Factor 5A Eucariótico de Iniciación de Traducción
7.
Tumori ; 101(6): 676-83, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26108243

RESUMEN

BACKGROUND: Nasopharyngeal carcinoma (NPC) is an epithelial malignancy exhibiting a strong geographic preference and a close association with Epstein-Barr virus (EBV). The aim of this study was to investigate the precise mechanism of nasopharyngeal epithelial-to-NPC tumorigenesis and to identify possible biomarkers and targets for therapy. METHODS: Proteomic analysis was performed on the immortalized nasopharyngeal epithelial cell line NP69 and the NPC cell line C666. RESULTS: A comparative analysis of total lysates from the cell lines using 2-D gel electrophoresis-mass spectrometry resulted in the identification of 87 different protein spots. The different proteins were grouped into 5 main categories: (i) energy production and general metabolism, (ii) adaptation/stress tolerance, (iii) cell proliferation, (iv) cell structure and (v) epithelial-mesenchymal transition. The detection of metabolism-related proteins indicated that the NPC cells relied on aerobic glycolysis, with reduced use of the citric acid cycle. Glucose uptake and lactate secretion increased in the medium of C666 compared with NP69. CONCLUSIONS: The present study revealed that glycolysis was up-regulated in the NPC cell lines compared with nasopharyngeal epithelial cells. A number of molecules involved in metabolism were identified, and further investigations will be needed to validate these potential biomarkers or targets.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Células Epiteliales/metabolismo , Transición Epitelial-Mesenquimal , Glucólisis , Neoplasias Nasofaríngeas/metabolismo , Western Blotting , Carcinoma , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Electroforesis en Gel Bidimensional , Regulación Neoplásica de la Expresión Génica , Glucosa/metabolismo , Herpesvirus Humano 4/aislamiento & purificación , Humanos , Inmunohistoquímica , Ácido Láctico/metabolismo , Espectrometría de Masas , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas/genética , Neoplasias Nasofaríngeas/virología , Proteómica , Regulación hacia Arriba
8.
PLoS One ; 8(7): e68838, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23894350

RESUMEN

By using cDNA microarray analysis, we identified cornulin (CRNN) gene was frequently downregulated in esophageal squamous cell carcinoma (ESCC). In the present study, we investigated the role of CRNN in ESCC development. The results showed that CRNN was frequently downregulated in primary ESCCs in both mRNA level (26/56, 46.4%) and protein level (137/249, 55%), which was significantly associated with lymph node metastases (P=0.027), advanced clinical stage (P=0.039), and overall survival rate (P<0.001). Multivariate analysis indicated that the CRNN downregulation was an independent prognostic factor for ESCC. Functional studies with both in vitro and in vivo assays demonstrated that CRNN had strong tumor suppressive ability in ESCC cells. The tumor-suppressive mechanism of CRNN was associated with its role in cell cycle arrest at G1/S checkpoint by upregulating expressions of P21(WAF1/CIP1) and Rb. Silencing CRNN expression by RNA interference could effectively inhibit its tumor suppressive effect. In conclusion, our findings demonstrate that CRNN is a tumor suppressor gene that plays a critical tumor suppressive role in ESCC.


Asunto(s)
Carcinoma de Células Escamosas/metabolismo , Ciclo Celular/fisiología , Neoplasias Esofágicas/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Animales , Western Blotting , Carcinoma de Células Escamosas/genética , Ciclo Celular/genética , Puntos de Control del Ciclo Celular/genética , Puntos de Control del Ciclo Celular/fisiología , Línea Celular Tumoral , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Neoplasias Esofágicas/genética , Carcinoma de Células Escamosas de Esófago , Humanos , Inmunohistoquímica , Técnicas In Vitro , Proteínas de la Membrana/genética , Ratones , Ratones Desnudos , Proteínas de Neoplasias/genética , Interferencia de ARN , Proteína de Retinoblastoma/genética , Proteína de Retinoblastoma/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de Matrices Tisulares
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA