Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Proc Natl Acad Sci U S A ; 116(28): 14144-14153, 2019 07 09.
Artículo en Inglés | MEDLINE | ID: mdl-31235597

RESUMEN

Epstein-Barr virus (EBV) induces histone modifications to regulate signaling pathways involved in EBV-driven tumorigenesis. To date, the regulatory mechanisms involved are poorly understood. In this study, we show that EBV infection of epithelial cells is associated with aberrant histone modification; specifically, aberrant histone bivalent switches by reducing the transcriptional activation histone mark (H3K4me3) and enhancing the suppressive mark (H3K27me3) at the promoter regions of a panel of DNA damage repair members in immortalized nasopharyngeal epithelial (NPE) cells. Sixteen DNA damage repair family members in base excision repair (BER), homologous recombination, nonhomologous end-joining, and mismatch repair (MMR) pathways showed aberrant histone bivalent switches. Among this panel of DNA repair members, MLH1, involved in MMR, was significantly down-regulated in EBV-infected NPE cells through aberrant histone bivalent switches in a promoter hypermethylation-independent manner. Functionally, expression of MLH1 correlated closely with cisplatin sensitivity both in vitro and in vivo. Moreover, seven BER members with aberrant histone bivalent switches in the EBV-positive NPE cell lines were significantly enriched in pathway analysis in a promoter hypermethylation-independent manner. This observation is further validated by their down-regulation in EBV-infected NPE cells. The in vitro comet and apurinic/apyrimidinic site assays further confirmed that EBV-infected NPE cells showed reduced DNA damage repair responsiveness. These findings suggest the importance of EBV-associated aberrant histone bivalent switch in host cells in subsequent suppression of DNA damage repair genes in a methylation-independent manner.


Asunto(s)
Infecciones por Virus de Epstein-Barr/genética , Herpesvirus Humano 4/genética , Código de Histonas/genética , Histonas/genética , Islas de CpG/genética , Daño del ADN/genética , Metilación de ADN/genética , Reparación de la Incompatibilidad de ADN/genética , Reparación del ADN/genética , Células Epiteliales/metabolismo , Células Epiteliales/virología , Infecciones por Virus de Epstein-Barr/patología , Infecciones por Virus de Epstein-Barr/virología , Regulación de la Expresión Génica/genética , Herpesvirus Humano 4/patogenicidad , Recombinación Homóloga/genética , Humanos , Homólogo 1 de la Proteína MutL/genética , Nasofaringe/crecimiento & desarrollo , Nasofaringe/patología , Nasofaringe/virología , Regiones Promotoras Genéticas
2.
Cancer Biomark ; 21(3): 583-590, 2018 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-29278876

RESUMEN

BACKGROUND: Nidogen-2 (NID2), a secretory basement membrane protein, has been implicated as a potential biomarker in ovarian cancer and hepatocellular carcinoma. OBJECTIVE: In this study, we aimed to investigate the utility of detecting serum NID2 levels for identification of esophageal squamous cell carcinoma (ESCC) patients and prediction of poor survival outcome. METHODS: Using an in-house NID2 enzyme-linked immunosorbent assay (ELISA), serum samples from 101 ESCC patients and 50 healthy controls were screened for their NID2 levels. RESULTS: The serum NID2 levels in ESCC patients (median 24.4 µg/L) are significantly higher (p= 4.3e-09) than that of the healthy controls (median 15.85 µg/L). The receiver operating characteristic (ROC) curve demonstrated an area under the curve of 0.756. At the threshold of 17.95 µg/L, the sensitivity and specificity achieved are 0.76 and 0.63, respectively. Kaplan-Meier survival analysis revealed that patients with high serum NID2 levels (⩾ 32.6 µg/L) have significantly higher risk of death (HR = 1.984, 95% CI: 1.175-3.349; log-rank p-value = 0.012) compared to those with low serum NID2 levels (< 20.0 µg/L). CONCLUSIONS: In conclusion, we show that detecting the elevation of serum NID2 levels has potential diagnostic and prognostic value for ESCC patients.


Asunto(s)
Biomarcadores de Tumor , Carcinoma de Células Escamosas/sangre , Carcinoma de Células Escamosas/mortalidad , Moléculas de Adhesión Celular/sangre , Neoplasias Esofágicas/sangre , Neoplasias Esofágicas/mortalidad , Adulto , Anciano , Proteínas de Unión al Calcio , Estudios de Casos y Controles , Ensayo de Inmunoadsorción Enzimática , Carcinoma de Células Escamosas de Esófago , Humanos , Estimación de Kaplan-Meier , Persona de Mediana Edad , Clasificación del Tumor , Estadificación de Neoplasias , Pronóstico , Curva ROC
3.
Oncotarget ; 7(48): 78859-78871, 2016 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-27793011

RESUMEN

Nidogen-2 (NID2) is a key component of the basement membrane that stabilizes the extracellular matrix (ECM) network. The aim of the study is to analyze the functional roles of NID2 in the pathogenesis of nasopharyngeal carcinoma (NPC) and esophageal squamous cell carcinoma (ESCC). We performed genome-wide methylation profiling of NPC and ESCC and validated our findings using the methylation-sensitive high-resolution melting (MS-HRM) assay. Results showed that promoter methylation of NID2 was significantly higher in NPC and ESCC samples than in their adjacent non-cancer counterparts. Consistently, down-regulation of NID2 was observed in the clinical samples and cell lines of both NPC and ESCC. Re-expression of NID2 suppresses clonogenic survival and migration abilities of transduced NPC and ESCC cells. We showed that NID2 significantly inhibits liver metastasis. Mechanistic studies of signaling pathways also confirm that NID2 suppresses the EGFR/Akt and integrin/FAK/PLCγ metastasis-related pathways. This study provides novel insights into the crucial tumor metastasis suppression roles of NID2 in cancers.


Asunto(s)
Carcinoma de Células Escamosas/genética , Carcinoma/genética , Moléculas de Adhesión Celular/genética , Metilación de ADN , Neoplasias Esofágicas/genética , Silenciador del Gen , Neoplasias Nasofaríngeas/genética , Animales , Proteínas de Unión al Calcio , Carcinoma/metabolismo , Carcinoma/secundario , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/secundario , Moléculas de Adhesión Celular/metabolismo , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Receptores ErbB/metabolismo , Neoplasias Esofágicas/metabolismo , Neoplasias Esofágicas/patología , Carcinoma de Células Escamosas de Esófago , Femenino , Quinasa 1 de Adhesión Focal/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Integrinas/metabolismo , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/secundario , Ratones Endogámicos BALB C , Ratones Desnudos , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas/metabolismo , Neoplasias Nasofaríngeas/patología , Invasividad Neoplásica , Fosfolipasa C gamma/metabolismo , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Factores de Tiempo , Transfección , Carga Tumoral
4.
Proc Natl Acad Sci U S A ; 113(40): 11283-11288, 2016 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-27647909

RESUMEN

Nasopharyngeal carcinoma (NPC) is an epithelial malignancy with a unique geographical distribution. The genomic abnormalities leading to NPC pathogenesis remain unclear. In total, 135 NPC tumors were examined to characterize the mutational landscape using whole-exome sequencing and targeted resequencing. An APOBEC cytidine deaminase mutagenesis signature was revealed in the somatic mutations. Noticeably, multiple loss-of-function mutations were identified in several NF-κB signaling negative regulators NFKBIA, CYLD, and TNFAIP3 Functional studies confirmed that inhibition of NFKBIA had a significant impact on NF-κB activity and NPC cell growth. The identified loss-of-function mutations in NFKBIA leading to protein truncation contributed to the altered NF-κB activity, which is critical for NPC tumorigenesis. In addition, somatic mutations were found in several cancer-relevant pathways, including cell cycle-phase transition, cell death, EBV infection, and viral carcinogenesis. These data provide an enhanced road map for understanding the molecular basis underlying NPC.


Asunto(s)
Carcinoma/genética , Secuenciación del Exoma/métodos , Mutación con Pérdida de Función/genética , FN-kappa B/metabolismo , Neoplasias Nasofaríngeas/genética , Transducción de Señal/genética , Línea Celular Tumoral , Técnicas de Silenciamiento del Gen , Humanos , Tasa de Mutación , Inhibidor NF-kappaB alfa/metabolismo , Carcinoma Nasofaríngeo
5.
Chin Clin Oncol ; 5(2): 16, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27121876

RESUMEN

Nasopharyngeal carcinoma (NPC) is a unique epithelial malignancy that shows a remarkable geographical and ethic distribution. Multiple factors including predisposing genetic factors, environmental carcinogens, and Epstein-Barr virus (EBV) infection contribute to the accumulation of genetic and epigenetic alterations leading to NPC development. Emerging technologies now allow us to detailedly characterize and understand cancer genomes. Genome-wide studies show that typically NPC tumors are characterized as having comparatively low mutation rates, widespread hypermethylation, and frequent copy number alterations and chromosome abnormalities. In this review, we provide an updated overview of the genetic and epigenetic aberrations that likely drive nasopharyngeal tumor development and progression. We integrate the previous knowledge and novel findings from whole-exome sequencing (WES) and methylome studies in NPC, and further discuss the potential use of these findings to identify biomarkers for NPC diagnosis and prognosis.


Asunto(s)
Epigénesis Genética , Neoplasias Nasofaríngeas/diagnóstico , Neoplasias Nasofaríngeas/genética , Biomarcadores de Tumor/genética , Carcinoma , Metilación de ADN , Infecciones por Virus de Epstein-Barr/genética , Dosificación de Gen , Histonas/genética , Histonas/metabolismo , Humanos , Pérdida de Heterocigocidad , Mutación , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas/virología , Pronóstico
6.
Proc Natl Acad Sci U S A ; 113(12): 3317-22, 2016 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-26951679

RESUMEN

Multiple factors, including host genetics, environmental factors, and Epstein-Barr virus (EBV) infection, contribute to nasopharyngeal carcinoma (NPC) development. To identify genetic susceptibility genes for NPC, a whole-exome sequencing (WES) study was performed in 161 NPC cases and 895 controls of Southern Chinese descent. The gene-based burden test discovered an association between macrophage-stimulating 1 receptor (MST1R) and NPC. We identified 13 independent cases carrying the MST1R pathogenic heterozygous germ-line variants, and 53.8% of these cases were diagnosed with NPC aged at or even younger than 20 y, indicating that MST1R germline variants are relevant to disease early-age onset (EAO) (age of ≤20 y). In total, five MST1R missense variants were found in EAO cases but were rare in controls (EAO vs. control, 17.9% vs. 1.2%, P = 7.94 × 10(-12)). The validation study, including 2,160 cases and 2,433 controls, showed that the MST1R variant c.G917A:p.R306H is highly associated with NPC (odds ratio of 9.0). MST1R is predominantly expressed in the tissue-resident macrophages and is critical for innate immunity that protects organs from tissue damage and inflammation. Importantly, MST1R expression is detected in the ciliated epithelial cells in normal nasopharyngeal mucosa and plays a role in the cilia motility important for host defense. Although no somatic mutation of MST1R was identified in the sporadic NPC tumors, copy number alterations and promoter hypermethylation at MST1R were often observed. Our findings provide new insights into the pathogenesis of NPC by highlighting the involvement of the MST1R-mediated signaling pathways.


Asunto(s)
Exoma , Predisposición Genética a la Enfermedad , Neoplasias Nasofaríngeas/genética , Proteínas Tirosina Quinasas Receptoras/genética , Análisis de Secuencia , Adolescente , Adulto , Carcinoma , Estudios de Casos y Controles , Femenino , Humanos , Masculino , Persona de Mediana Edad , Carcinoma Nasofaríngeo , Adulto Joven
7.
Int J Cancer ; 138(1): 125-36, 2016 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-26205347

RESUMEN

Pan-histone deacetylase (HDAC) inhibitors, which inhibit 11 HDAC isoforms, are widely used to induce Epstein-Barr virus (EBV) lytic cycle in EBV-associated cancers in vitro and in clinical trials. Here, we hypothesized that inhibition of one or several specific HDAC isoforms by selective HDAC inhibitors could potently induce EBV lytic cycle in EBV-associated malignancies such as nasopharyngeal carcinoma (NPC) and gastric carcinoma (GC). We found that inhibition of class I HDACs, particularly HDAC-1, -2 and -3, was sufficient to induce EBV lytic cycle in NPC and GC cells in vitro and in vivo. Among a panel of selective HDAC inhibitors, the FDA-approved HDAC inhibitor romidepsin was found to be the most potent lytic inducer, which could activate EBV lytic cycle at ∼0.5 to 5 nM (versus ∼800 nM achievable concentration in patients' plasma) in more than 75% of cells. Upregulation of p21(WAF1) , which is negatively regulated by class I HDACs, was observed before the induction of EBV lytic cycle. The upregulation of p21(WAF1) and induction of lytic cycle were abrogated by a specific inhibitor of PKC-δ but not the inhibitors of PI3K, MEK, p38 MAPK, JNK or ATM pathways. Interestingly, inhibition of HDAC-1, -2 and -3 by romidepsin or shRNA knockdown could confer susceptibility of EBV-positive epithelial cells to the treatment with ganciclovir (GCV). In conclusion, we demonstrated that inhibition of class I HDACs by romidepsin could potently induce EBV lytic cycle and mediate enhanced cell death with GCV, suggesting potential application of romidepsin for the treatment of EBV-associated cancers.


Asunto(s)
Antivirales/farmacología , Depsipéptidos/farmacología , Ganciclovir/farmacología , Herpesvirus Humano 4/efectos de los fármacos , Herpesvirus Humano 4/fisiología , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/metabolismo , Replicación Viral/efectos de los fármacos , Acetilación , Animales , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Carcinoma , Muerte Celular/efectos de los fármacos , Línea Celular , Proliferación Celular/efectos de los fármacos , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Relación Dosis-Respuesta a Droga , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Células Epiteliales/virología , Histona Desacetilasas/genética , Histonas/metabolismo , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas/tratamiento farmacológico , Neoplasias Nasofaríngeas/metabolismo , Neoplasias Nasofaríngeas/patología , Neoplasias Nasofaríngeas/virología , Proteína Quinasa C-delta/metabolismo , Transducción de Señal/efectos de los fármacos , Neoplasias Gástricas/tratamiento farmacológico , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología , Neoplasias Gástricas/virología , Activación Viral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Int J Cancer ; 138(1): 160-70, 2016 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-26227166

RESUMEN

Tumor suppressor genes (TSGs) play a prominent role in cancer and are important in the development of nasopharyngeal carcinoma (NPC), which is endemic in Southern China as well as Southeast Asia. Apart from TSGs, aberrant signalling pathways are also commonly associated with tumor progression. Unsurprisingly, the NF-κB pathway is frequently associated with angiogenesis and promoting tumor growth and development. Functional complementation studies using microcell-mediated chromosome transfer helped to identify IKBB as a putative TSG in NPC. IKBB, an inhibitor of NF-κB, has recently been shown to be inversely associated with tumor growth and metastasis via inactivation of the NF-κB pathway, but its suppressive role is still only poorly understood. This study takes the lead in revealing the suppressive role of IKBB in NPC. IKBB is silenced in the majority of NPC tumor tissues in all stages. Its suppressive role is substantiated by perturbation in tumor formation, cell migration and angiogenesis. Interestingly, IKBB not only affects the 'seed', but also influences the 'soil' by downregulating the transcriptional level of proangiogenic factors Rantes, Upar, IL6, and IL8. For the first time, our data establish the importance of a novel tumor suppressive IKBB gene in abrogating angiogenesis in NPC via the NF-κB signalling pathway, which is likely mediated by crosstalk with the Akt/Gsk3ß signalling pathway.


Asunto(s)
Proteínas I-kappa B/metabolismo , FN-kappa B/metabolismo , Neoplasias Nasofaríngeas/metabolismo , Transducción de Señal , Proteínas Supresoras de Tumor/metabolismo , Adulto , Anciano , Carcinoma , Línea Celular Tumoral , Movimiento Celular/genética , Citocinas/genética , Citocinas/metabolismo , Regulación hacia Abajo , Femenino , Regulación Neoplásica de la Expresión Génica , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Humanos , Proteínas I-kappa B/genética , Masculino , Persona de Mediana Edad , FN-kappa B/genética , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas/genética , Neoplasias Nasofaríngeas/mortalidad , Neoplasias Nasofaríngeas/patología , Estadificación de Neoplasias , Neovascularización Patológica/genética , Pronóstico , Unión Proteica , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Supresoras de Tumor/genética
9.
Oncotarget ; 6(15): 13434-47, 2015 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-25970784

RESUMEN

Protein Tyrosine Phosphatase, Receptor Type G (PTPRG) was identified as a candidate tumor suppressor gene in nasopharyngeal carcinoma (NPC). PTPRG induces significant in vivo tumor suppression in NPC. We identified EGFR as a PTPRG potential interacting partner and examined this interaction. Dephosphorylation of EGFR at EGFR-Y1068 and -Y1086 sites inactivated the PI3K/Akt signaling cascade and subsequent down-regulation of downstream pro-angiogenic and -invasive proteins (VEGF, IL6, and IL8) and suppressed tumor cell proliferation, angiogenesis, and invasion. The effect of Akt inhibition in NPC cells was further validated by Akt knockdown experiments in the PTPRG-down-regulated NPC cell lines. Our results suggested that inhibition of Akt in NPC cells induces tumor suppression at both the in vitro and in vivo levels, and also importantly, in vivo metastasis. In conclusion, we confirmed the vital role of PTPRG in inhibiting Akt signaling with the resultant suppression of in vivo tumorigenesis and metastasis.


Asunto(s)
Neoplasias Nasofaríngeas/enzimología , Neoplasias Nasofaríngeas/patología , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Tirosina Fosfatasas Clase 5 Similares a Receptores/metabolismo , Animales , Carcinogénesis , Carcinoma , Línea Celular Tumoral , Proliferación Celular , Genes Supresores de Tumor , Xenoinjertos , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas/genética , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Tirosina Fosfatasas Clase 5 Similares a Receptores/genética , Transducción de Señal
10.
Cancer Med ; 4(7): 1079-90, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25924914

RESUMEN

Altered patterns of DNA methylation are key features of cancer. Nasopharyngeal carcinoma (NPC) has the highest incidence in Southern China. Aberrant methylation at the promoter region of tumor suppressors is frequently reported in NPC; however, genome-wide methylation changes have not been comprehensively investigated. Therefore, we systematically analyzed methylome data in 25 primary NPC tumors and nontumor counterparts using a high-throughput approach with the Illumina HumanMethylation450 BeadChip. Comparatively, we examined the methylome data of 11 types of solid tumors collected by The Cancer Genome Atlas (TCGA). In NPC, the hypermethylation pattern was more dominant than hypomethylation and the majority of de novo methylated loci were within or close to CpG islands in tumors. The comparative methylome analysis reveals hypermethylation at chromosome 6p21.3 frequently occurred in NPC (false discovery rate; FDR=1.33 × 10(-9) ), but was less obvious in other types of solid tumors except for prostate and Epstein-Barr virus (EBV)-positive gastric cancer (FDR<10(-3) ). Bisulfite pyrosequencing results further confirmed the aberrant methylation at 6p in an additional patient cohort. Evident enrichment of the repressive mark H3K27me3 and active mark H3K4me3 derived from human embryonic stem cells were found at these regions, indicating both DNA methylation and histone modification function together, leading to epigenetic deregulation in NPC. Our study highlights the importance of epigenetic deregulation in NPC. Polycomb Complex 2 (PRC2), responsible for H3K27 trimethylation, is a promising therapeutic target. A key genomic region on 6p with aberrant methylation was identified. This region contains several important genes having potential use as biomarkers for NPC detection.


Asunto(s)
Cromosomas Humanos Par 6 , Metilación de ADN , Neoplasias Nasofaríngeas/genética , Transcriptoma , Carcinoma , Biología Computacional , Islas de CpG , Epigénesis Genética , Epigenómica/métodos , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Histonas/metabolismo , Humanos , Metaanálisis como Asunto , Metilación , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas/metabolismo , Neoplasias Nasofaríngeas/patología , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patología , Regiones Promotoras Genéticas
11.
PLoS One ; 10(5): e0127239, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25974126

RESUMEN

NF-κB is a well-characterized transcription factor, widely known as a key player in tumor-derived inflammation and cancer development. Herein, we present the functional and molecular relevance of the canonical NF-κB p65 subunit in nasopharyngeal carcinoma (NPC). Loss- and gain-of-function approaches were utilized to reveal the functional characteristics of p65 in propagating tumor growth, tumor-associated angiogenesis, and epithelial-to-mesenchymal transition in NPC cells. Extracellular inflammatory stimuli are critical factors that trigger the NF-κB p65 signaling; hence, we investigated the components of the tumor microenvironment that might potentially influence the p65 signaling pathway. This led to the identification of an extracellular matrix (ECM) protein that was previously reported as a candidate tumor suppressor in NPC. Our studies on the Latent Transforming Growth Factor-ß Binding Protein 2 (LTBP2) protein provides substantial evidence that it can modulate the p65 transcriptional activity. Re-expression of LTBP2 elicits tumor suppressive effects that parallel the inactivation of p65 in NPC cells. LTBP2 was able to reduce phosphorylation of p65 at Serine 536, inhibit nuclear localization of active phosphorylated p65, and impair the p65 DNA-binding ability. This results in a consequential down-regulation of p65-related gene expression. Therefore, the data suggest that the overall up-regulation of p65 expression and the loss of this candidate ECM tumor suppressor are milestone events contributing to NPC development.


Asunto(s)
Carcinoma/metabolismo , Proteínas de Unión a TGF-beta Latente/metabolismo , FN-kappa B/metabolismo , Neoplasias Nasofaríngeas/metabolismo , Factor de Transcripción ReIA/metabolismo , Carcinoma/patología , Línea Celular Tumoral , Movimiento Celular/fisiología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Neoplasias Nasofaríngeas/patología , Fosforilación , Transducción de Señal , Microambiente Tumoral
12.
Chin J Cancer ; 33(11): 556-68, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25367335

RESUMEN

The interplay between host cell genetics and Epstein-Barr virus (EBV) infection contributes to the development of nasopharyngeal carcinoma (NPC). Understanding the host genetic and epigenetic alterations and the influence of EBV on cell signaling and host gene regulation will aid in understanding the molecular pathogenesis of NPC and provide useful biomarkers and targets for diagnosis and therapy. In this review, we provide an update of the oncogenes and tumor suppressor genes associated with NPC, as well as genes associated with NPC risk including those involved in carcinogen detoxification and DNA repair. We also describe the importance of host genetics that govern the human leukocyte antigen (HLA) complex and immune responses, and we describe the impact of EBV infection on host cell signaling changes and epigenetic regulation of gene expression. High-power genomic sequencing approaches are needed to elucidate the genetic basis for inherited susceptibility to NPC and to identify the genes and pathways driving its molecular pathogenesis.


Asunto(s)
Epigénesis Genética , Infecciones por Virus de Epstein-Barr , Predisposición Genética a la Enfermedad , Neoplasias Nasofaríngeas/etiología , Carcinoma , Genes Supresores de Tumor , Herpesvirus Humano 4/genética , Humanos , Carcinoma Nasofaríngeo , Oncogenes , Transducción de Señal
13.
BMC Cell Biol ; 14: 44, 2013 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-24073846

RESUMEN

BACKGROUND: A few reports suggested that low levels of Wnt signaling might drive cell reprogramming, but these studies could not establish a clear relationship between Wnt signaling and self-renewal networks. There are ongoing debates as to whether and how the Wnt/ß-catenin signaling is involved in the control of pluripotency gene networks. Additionally, whether physiological ß-catenin signaling generates stem-like cells through interactions with other pathways is as yet unclear. The nasopharyngeal carcinoma HONE1 cells have low expression of ß-catenin and wild-type expression of p53, which provided a possibility to study regulatory mechanism of stemness networks induced by physiological levels of Wnt signaling in these cells. RESULTS: Introduction of increased ß-catenin signaling, haploid expression of ß-catenin under control by its natural regulators in transferred chromosome 3, resulted in activation of Wnt/ß-catenin networks and dedifferentiation in HONE1 hybrid cell lines, but not in esophageal carcinoma SLMT1 hybrid cells that had high levels of endogenous ß-catenin expression. HONE1 hybrid cells displayed stem cell-like properties, including enhancement of CD24(+) and CD44(+) populations and generation of spheres that were not observed in parental HONE1 cells. Signaling cascades were detected in HONE1 hybrid cells, including activation of p53- and RB1-mediated tumor suppressor pathways, up-regulation of Nanog-, Oct4-, Sox2-, and Klf4-mediated pluripotency networks, and altered E-cadherin expression in both in vitro and in vivo assays. qPCR array analyses further revealed interactions of physiological Wnt/ß-catenin signaling with other pathways such as epithelial-mesenchymal transition, TGF-ß, Activin, BMPR, FGFR2, and LIFR- and IL6ST-mediated cell self-renewal networks. Using ß-catenin shRNA inhibitory assays, a dominant role for ß-catenin in these cellular network activities was observed. The expression of cell surface markers such as CD9, CD24, CD44, CD90, and CD133 in generated spheres was progressively up-regulated compared to HONE1 hybrid cells. Thirty-four up-regulated components of the Wnt pathway were identified in these spheres. CONCLUSIONS: Wnt/ß-catenin signaling regulates self-renewal networks and plays a central role in the control of pluripotency genes, tumor suppressive pathways and expression of cancer stem cell markers. This current study provides a novel platform to investigate the interaction of physiological Wnt/ß-catenin signaling with stemness transition networks.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Neoplasias Nasofaríngeas/metabolismo , Proteínas de Neoplasias/metabolismo , Células Madre Neoplásicas/metabolismo , Transducción de Señal/genética , beta Catenina/metabolismo , Antígenos CD/genética , Antígenos CD/metabolismo , Carcinoma , Línea Celular Tumoral , Proliferación Celular , Transición Epitelial-Mesenquimal/genética , Perfilación de la Expresión Génica , Redes Reguladoras de Genes , Humanos , Células Híbridas/metabolismo , Células Híbridas/patología , Factor 4 Similar a Kruppel , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas/genética , Neoplasias Nasofaríngeas/patología , Proteínas de Neoplasias/genética , Células Madre Neoplásicas/patología , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Esferoides Celulares/metabolismo , Esferoides Celulares/patología , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , beta Catenina/antagonistas & inhibidores , beta Catenina/genética
14.
Mol Cancer Ther ; 12(8): 1393-401, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23686835

RESUMEN

Nasopharyngeal carcinoma is a cancer with its highest prevalence among the southern Chinese and is rare elsewhere in the world. The main treatment modalities include chemotherapy and radiotherapy. However, tumor chemoresistance often limits the efficacy of nasopharyngeal carcinoma treatment and reduces survival rates. Thus, identifying new selective chemotherapeutic drugs for nasopharyngeal carcinoma treatment is needed. In this current study, the antitumor efficacy of a polo-like kinase inhibitor, Ro5203280, was investigated. Ro5203280 induces tumor suppression both in vitro and in vivo. An inhibitory effect was observed with the highly proliferating cancer cell lines tested, but not with the nontumorigenic cell line. Real-time cell proliferation and fluorescence-activated cell sorting (FACS) analysis, together with immunohistochemical (IHC), immunofluorescence, and Annexin V staining assays, were used to evaluate the impact of drug treatment on cell cycle and apoptosis. Ro5203280 induces G2-M cell-cycle arrest and apoptosis. Western blotting shows it inhibits PLK1 phosphorylation and downregulates the downstream signaling molecule, Cdc25c, and upregulates two important mitosis regulators, Wee1 and Securin, as well as the DNA damage-related factor Chk2 in vitro and in vivo. In vivo tumorigenicity assays with Ro5203280 intravenous injection showed its potent ability to inhibit tumor growth in mice, with no observable signs of toxicity. These findings suggest the potential usefulness of Ro5203280 as a chemotherapeutic targeting drug for nasopharyngeal carcinoma treatment.


Asunto(s)
Antineoplásicos/farmacología , Benzamidas/farmacología , Proteínas de Ciclo Celular/antagonistas & inhibidores , Compuestos Heterocíclicos con 2 Anillos/farmacología , Neoplasias Nasofaríngeas/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/química , Apoptosis/efectos de los fármacos , Benzamidas/administración & dosificación , Benzamidas/química , Carcinoma , Puntos de Control del Ciclo Celular/efectos de los fármacos , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Compuestos Heterocíclicos con 2 Anillos/administración & dosificación , Compuestos Heterocíclicos con 2 Anillos/química , Humanos , Ratones , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas/tratamiento farmacológico , Neoplasias Nasofaríngeas/patología , Inhibidores de Proteínas Quinasas/administración & dosificación , Inhibidores de Proteínas Quinasas/química , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Huso Acromático/efectos de los fármacos , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto , Quinasa Tipo Polo 1
15.
Semin Cancer Biol ; 22(2): 87-95, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22154888

RESUMEN

The identification of cancer genes in sporadic cancers has been recognized as a major challenge in the field. It is clear that deletion mapping, genomic sequencing, comparative genomic hybridization, or global gene expression profiling alone would not have easily identified candidate tumor suppressor genes (TSGs) from the huge array of lost regions or genes observed in nasopharyngeal carcinoma (NPC). In addition, the epigenetically silenced genes would not have been recognized by the mapping of deleted regions. In this review, we describe how functional approaches using monochromosome transfer may be used to circumvent the above problems and identify TSGs in NPC. A few examples of selected NPC TSGs and their functional roles are reviewed. They regulate a variety of gene functions including cell growth and proliferation, adhesion, migration, invasion, epithelial-mesenchymal transition, metastasis, and angiogenesis. These studies show the advantages of using functional approaches for identification of TSGs.


Asunto(s)
Mapeo Cromosómico/métodos , Predisposición Genética a la Enfermedad/genética , Neoplasias Nasofaríngeas/genética , Proteínas Supresoras de Tumor/genética , Animales , Carcinoma , Fusión Celular/métodos , Movimiento Celular/genética , Humanos , Células Híbridas/metabolismo , Ratones , Carcinoma Nasofaríngeo
16.
Proc Natl Acad Sci U S A ; 108(20): 8390-5, 2011 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-21540330

RESUMEN

Chromosome 14 was transferred into tumorigenic nasopharyngeal carcinoma and esophageal carcinoma cell lines by a microcell-mediated chromosome transfer approach. Functional complementation of defects present in the cancer cells suppressed tumor formation. A candidate tumor-suppressor gene, cysteine-rich intestinal protein 2 (CRIP2), located in the hot spot for chromosomal loss at 14q32.3, was identified as an important candidate gene capable of functionally suppressing tumor formation. Previous studies have shown that CRIP2 is associated with development. To date, no report has provided functional evidence supporting a role for CRIP2 in tumor development. The present study provides unequivocal evidence that CRIP2 can functionally suppress tumorigenesis. CRIP2 is significantly down-regulated in nasopharyngeal carcinoma cell lines and tumors. CRIP2 reexpression functionally suppresses in vivo tumorigenesis and angiogenesis; these effects are induced by its transcription-repressor capability. It interacts with the NF-κB/p65 to inhibit its DNA-binding ability to the promoter regions of the major proangiogenesis cytokines critical for tumor progression, including IL6, IL8, and VEGF. In conclusion, we provide compelling evidence that CRIP2 acts as a transcription repressor of the NF-κB-mediated proangiogenic cytokine expression and thus functionally inhibits tumor formation and angiogenesis.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Transformación Celular Neoplásica/genética , Citocinas/genética , FN-kappa B/metabolismo , Neovascularización Patológica/genética , Transcripción Genética , Proteínas Supresoras de Tumor/genética , Proteínas Angiogénicas/análisis , Línea Celular , Línea Celular Tumoral , Cromosomas Humanos Par 14 , Citocinas/fisiología , Humanos , Proteínas con Dominio LIM , Proteínas Represoras/fisiología
17.
Cancer Res ; 70(13): 5567-76, 2010 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-20551050

RESUMEN

ADAMTS metalloprotease family member ADAMTS9 maps to 3p14.2 and shows significant associations with the aerodigestive tract cancers esophageal squamous cell carcinoma (ESCC) and nasopharyngeal carcinoma (NPC). However, the functional impact of ADAMTS9 on cancer development has not been explored. In this study, we evaluated the hypothesized antiangiogenic and tumor-suppressive functions of ADAMTS9 in ESCC and NPC, in stringent tumorigenicity and Matrigel plug angiogenesis assays. ADAMTS9 activation suppressed tumor formation in nude mice. Conversely, knockdown of ADAMTS9 resulted in clones reverting to the tumorigenic phenotype of parental cells. In vivo angiogenesis assays revealed a reduction in microvessel numbers in gel plugs injected with tumor-suppressive cell transfectants. Similarly, conditioned medium from cell transfectants dramatically reduced the tube-forming capacity of human umbilical vein endothelial cells. These activities were associated with a reduction in expression levels of the proangiogenic factors MMP9 and VEGFA, which were consistently reduced in ADAMTS9 transfectants derived from both cancers. Taken together, our results indicate that ADAMTS9 contributes an important function in the tumor microenvironment that acts to inhibit angiogenesis and tumor growth in both ESCC and NPC.


Asunto(s)
Proteínas ADAM/biosíntesis , Neoplasias Esofágicas/irrigación sanguínea , Neoplasias Esofágicas/enzimología , Neoplasias Nasofaríngeas/irrigación sanguínea , Neoplasias Nasofaríngeas/enzimología , Proteínas ADAM/genética , Proteínas ADAM/metabolismo , Proteína ADAMTS9 , Animales , Regulación hacia Abajo , Células Endoteliales/citología , Activación Enzimática , Neoplasias Esofágicas/genética , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Metaloproteinasa 9 de la Matriz/biosíntesis , Metaloproteinasa 9 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Neoplasias Nasofaríngeas/genética , Neovascularización Patológica/enzimología , Neovascularización Patológica/genética , Neovascularización Patológica/patología , Transfección , Factor A de Crecimiento Endotelial Vascular/biosíntesis , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
18.
Int J Cancer ; 127(2): 304-12, 2010 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-19921696

RESUMEN

THY1 was previously identified as a candidate tumor suppressor gene (TSG) associated with lymph node metastases in nasopharyngeal carcinoma (NPC) through functional studies. It was identified by oligonucleotide microarray analysis as an interesting differentially expressed gene. However, direct functional evidence is still lacking for THY1 being a TSG in NPC, as in vivo tumorigenicity assays have not been previously reported in our last study of THY1. In this study, a tetracycline-inducible expression vector, pETE-Bsd, was used to obtain stable transfectants of THY1. The stringent in vivo tumorigenicity assay results show that the activation of THY1 suppresses tumor formation of HONE1 cells in nude mice, and the tumor formation ability was restored in the presence of doxycycline (a tetracycline analog), when the gene is shut off. Functional inactivation of this gene is observed in all the tumors derived from the tumorigenic transfectant. The tumor suppressive effect could be repressed by knockdown of THY1 expression in nontumorigenic microcell hybrids. Further studies indicate that expression of THY1 inhibits HONE1 cell growth in vitro by arresting cells in G(0)/G(1) phase. It greatly reduces the ability for anchorage-independent growth. The invasiveness of HONE1 cells was also inhibited by the expression of THY1. These findings suggest that THY1 is a TSG in NPC, which is involved in invasion and shows an association with tumor metastasis. Taken together, THY1 clearly plays an important functional role in tumor suppression in NPC.


Asunto(s)
Movimiento Celular , Regulación Neoplásica de la Expresión Génica/fisiología , Genes Supresores de Tumor/fisiología , Neoplasias Nasofaríngeas/patología , Antígenos Thy-1/fisiología , Animales , Western Blotting , Adhesión Celular , Ciclo Celular , Proliferación Celular , Femenino , Humanos , Metástasis Linfática , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Neoplasias Nasofaríngeas/genética , Neoplasias Nasofaríngeas/metabolismo , Invasividad Neoplásica , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Proc Natl Acad Sci U S A ; 106(34): 14478-83, 2009 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-19667180

RESUMEN

Chromosome 14 allelic loss is common in nasopharyngeal carcinoma (NPC) and may reflect essential tumor suppressor gene loss in tumorigenesis. An intact chromosome 14 was transferred to an NPC cell line using a microcell-mediated chromosome transfer approach. Microcell hybrids (MCHs) containing intact exogenously transferred chromosome 14 were tumor suppressive in athymic mice, demonstrating that intact chromosome 14 NPC MCHs are able to suppress tumor growth in mice. Comparative analysis of these MCHs and their derived tumor segregants identified 4 commonly eliminated tumor-suppressive CRs. Here we provide functional evidence that a gene, Mirror-Image POLydactyly 1 (MIPOL1), which maps within a single 14q13.1-13.3 CR and that hitherto has been reported to be associated only with a developmental disorder, specifically suppresses in vivo tumor formation. MIPOL1 gene expression is down-regulated in all NPC cell lines and in approximately 63% of NPC tumors via promoter hypermethylation and allelic loss. SLC25A21 and FOXA1, 2 neighboring genes mapping to this region, did not show this frequent down-regulated gene expression or promoter hypermethylation, precluding possible global methylation effects and providing further evidence that MIPOL1 plays a unique role in NPC. The protein localizes mainly to the nucleus. Re-expression of MIPOL1 in the stable transfectants induces cell cycle arrest. MIPOL1 tumor suppression is related to up-regulation of the p21(WAF1/CIP1) and p27(KIP1) protein pathways. This study provides compelling evidence that chromosome 14 harbors tumor suppressor genes associated with NPC and that a candidate gene, MIPOL1, is associated with tumor development.


Asunto(s)
Cromosomas Humanos Par 14/genética , Neoplasias Nasofaríngeas/genética , Proteínas Supresoras de Tumor/genética , Animales , Ciclo Celular , Línea Celular Transformada , Línea Celular Tumoral , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina , Metilación de ADN , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Técnicas de Transferencia de Gen , Genes Supresores de Tumor , Predisposición Genética a la Enfermedad , Humanos , Inmunohistoquímica , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones , Ratones Desnudos , Neoplasias Nasofaríngeas/metabolismo , Neoplasias Nasofaríngeas/patología , Neoplasias Experimentales/genética , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Reacción en Cadena de la Polimerasa/métodos , Transfección/instrumentación , Transfección/métodos , Trasplante Heterólogo , Proteínas Supresoras de Tumor/metabolismo
20.
Cancer Res ; 68(19): 8137-45, 2008 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-18829573

RESUMEN

Functional studies to identify the potential role of a chromosome 3p14-21 gene, protein tyrosine phosphatase receptor type G (PTPRG), were performed. PTPRG was identified as a candidate tumor suppressor gene (TSG) in nasopharyngeal carcinoma (NPC) by differential gene profiling of tumorigenic and nontumorigenic NPC chromosome 3 microcell hybrids (MCH). Down-regulation of this gene was found in tumor segregants when compared with their corresponding tumor-suppressive MCHs, as well as in NPC cell lines and tumor biopsies. Promoter hypermethylation and loss of heterozygosity were found to be important mechanisms contributing to PTPRG silencing. PTPRG overexpression in NPC cell lines induces growth suppression and reduced anchorage-independent growth in vitro. This is the first study to use a tetracycline-responsive vector expression system to study PTPRG stable transfectants. Results indicate its ability to induce significant tumor growth suppression in nude mice under conditions activating transgene expression. These studies now provide functional evidence indicating critical interactions of PTPRG in the extracellular matrix milieu induce cell arrest and changes in cell cycle status. This is associated with inhibition of pRB phosphorylation through down-regulation of cyclin D1. These novel findings enhance our current understanding of how PTPRG may contribute to tumorigenesis.


Asunto(s)
Carcinoma/genética , Neoplasias Nasofaríngeas/genética , Proteínas Tirosina Fosfatasas Clase 5 Similares a Receptores/fisiología , Animales , Células Cultivadas , Cromosomas Humanos Par 3 , Metilación de ADN , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Genes Supresores de Tumor/fisiología , Genes cdc/fisiología , Humanos , Pérdida de Heterocigocidad , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteínas Tirosina Fosfatasas Clase 5 Similares a Receptores/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...