Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 128
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
2.
J Periodontal Res ; 49(6): 845-54, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24620831

RESUMEN

BACKGROUND AND OBJECTIVE: Lipopolysaccharide (LPS)-mediated signaling in host cells involves Toll-like receptor 4 (TLR4) accessory molecules, including LPS-binding protein (LBP), cluster of differentiation 14 (CD14) and lymphocyte antigen 96 (MD-2). However, expression of these innate defense molecules in various compartments of the human periodontium is unclear. The aim of this study was to investigate the expression profile of TLR4 in human gingiva. MATERIAL AND METHODS: Human gingival biopsies were collected from healthy gingival or chronic periodontitis tissue. Primary gingival keratinocytes and fibroblasts were cultured. Immunohistochemical analysis for TLR4 was performed. Transcripts of TLR4, MD-2, CD14 and LBP, and their protein products, were examined using RT-PCR, immunoprecipitation and immunoblotting. The interactions between these molecules in keratinocytes and fibroblasts were investigated by co-immunoprecipitation. RESULTS: TLR4 immunoreactivity was found in healthy gingival epithelium and periodontitis tissue, and appeared to be lower in junctional epithelium ( p ≤ 0.01). Fibroblasts and inflammatory cells stained more strongly for TLR4 in diseased periodontal tissues (p < 0.001). Three TLR4 splicing variants, two MD-2 splicing variants and one CD14 mRNA were expressed by gingival keratinocytes and fibroblasts. Expression of TLR4, CD14 and MD-2 proteins was detected in keratinocytes and fibroblasts in vitro. TLR4 protein from gingival keratinocytes and fibroblasts could be co-immunoprecipitated with CD14 or MD-2, suggesting an association between the related molecules in vivo. LBP transcript was detected in gingival biopsies, but not in primary cultures of gingival keratinocytes or fibroblasts. CONCLUSION: TLR4, CD14 and MD-2, but not LBP, are expressed in human gingival keratinocytes and fibroblasts. The TLR4 expression level in the junctional epithelium appeared to be lowest within the periodontal epithelial barrier.


Asunto(s)
Periodontitis Crónica/inmunología , Encía/inmunología , Receptor Toll-Like 4/análisis , Proteínas de Fase Aguda/análisis , Adulto , Empalme Alternativo/genética , Pérdida de Hueso Alveolar/clasificación , Proteínas Portadoras/análisis , Células Cultivadas , Periodontitis Crónica/clasificación , Inserción Epitelial/inmunología , Epitelio/inmunología , Exones/genética , Femenino , Fibroblastos/inmunología , Encía/patología , Humanos , Inmunidad Innata/inmunología , Queratinocitos/inmunología , Leucocitos/inmunología , Receptores de Lipopolisacáridos/análisis , Receptores de Lipopolisacáridos/genética , Lipopolisacáridos/inmunología , Antígeno 96 de los Linfocitos/análisis , Antígeno 96 de los Linfocitos/genética , Masculino , Glicoproteínas de Membrana/análisis , Persona de Mediana Edad , Pérdida de la Inserción Periodontal/clasificación , Bolsa Periodontal/clasificación , Bolsa Periodontal/patología , Receptor Toll-Like 4/genética
3.
Nat Med ; 13(2): 147-9, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17206149

RESUMEN

Poor human-to-human transmission of influenza A H5N1 virus has been attributed to the paucity of putative sialic acid alpha2-3 virus receptors in the epithelium of the human upper respiratory tract, and thus to the presumed inability of the virus to replicate efficiently at this site. We now demonstrate that ex vivo cultures of human nasopharyngeal, adenoid and tonsillar tissues can be infected with H5N1 viruses in spite of an apparent lack of these receptors.


Asunto(s)
Subtipo H5N1 del Virus de la Influenza A/metabolismo , Gripe Humana/transmisión , Receptores de Superficie Celular/metabolismo , Sistema Respiratorio/virología , Acoplamiento Viral , Células Cultivadas , Epitelio/virología , Histocitoquímica , Hong Kong , Humanos , Subtipo H1N1 del Virus de la Influenza A/metabolismo , Subtipo H3N2 del Virus de la Influenza A/metabolismo , Gripe Humana/metabolismo , Fitohemaglutininas/metabolismo , Replicación Viral/fisiología
4.
Invest New Drugs ; 31(3): 567-75, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23143779

RESUMEN

Nasopharyngeal carcinoma (NPC) is endemic to Asia and over 40 % of NPC tissues harbor PIK3CA amplifications. This study characterized the preclinical activity of MK-2206, an oral allosteric inhibitor of AKT in 6 NPC cell lines: C666-1, HK1, HONE-1-EBV, HONE-1, CNE-2 and HNE-1. Exposure to increasing concentrations of MK-2206 resulted in over 95 % of growth inhibition in all NPC cell lines with IC50 values in the low micromolar range. Further experiments were performed in 3 representative NPC cell lines: CNE-2 (harbor PIK3CA mutation and most sensitive to MK-2206), C666-1 (carries PIK3CA amplification), and HONE-1-EBV (least sensitive to MK-2206). MK-2206 induced G0/G1 cycle arrest in all 3 cell lines, but could induce apoptosis only in CNE-2 cells. MK-2206 significantly abrogated AKT signaling in all 3 cell lines by inhibiting the activation of AKT and its downstream effectors (FKHR, GSK3ß and BAD). MK-2206 also reduced mTOR signaling by reducing activation of mTOR and its downstream 4E-BP1 and p70S6 kinase. MAPK activation was observed in HONE-1 and C666-1 cells, but not in CNE-2 cells following exposure to MK-2206. The addition of MK-2206 to cisplatin (but not with paclitaxel) has a supra-additive inhibitory effect on growth in vitro. In summary, MK-2206 can inhibit growth and abrogate AKT and mTOR signaling in NPC cell lines. This agent is currently being evaluated in a phase II study in metastatic NPC.


Asunto(s)
Antineoplásicos/administración & dosificación , Compuestos Heterocíclicos con 3 Anillos/administración & dosificación , Neoplasias Nasofaríngeas/metabolismo , Inhibidores de Proteínas Quinasas/administración & dosificación , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Carcinoma , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Cisplatino/administración & dosificación , Humanos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Carcinoma Nasofaríngeo , Paclitaxel/administración & dosificación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Serina-Treonina Quinasas TOR/metabolismo
5.
J Cell Biochem ; 112(1): 157-68, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21053361

RESUMEN

Epithelial-mesenchymal transition (EMT), characterized by cadherin switching, contributes to cancer metastasis. Our recent study showed that Id-1 (inhibitor of differentiation-1) promotes metastasis in esophageal cancer cells, but whether the invasive and metastatic dynamics can be induced early in the carcinogenesis process is still unclear. Immortalization is regarded as the initial stage in the malignant transformation of normal cells. In this study, we investigated the role and mechanisms of Id-1 in inducing EMT and cell invasiveness in immortalized esophageal epithelial cells. We found that immortalized epithelial cells expressed higher endogenous levels of Id-1 compared with normal cells. Ectopic Id-1 expression inhibited the differentiation of immortalized esophageal epithelial cells and promoted cadherin switching, which was accompanied by increased adhesiveness to extracellular matrix, cell motility, migratory potential and matrix metalloproteinase-dependent invasiveness. GTPase activity assays showed that over-expression or short-hairpin RNA knockdown of Id-1 led to corresponding changes in Rac1 activity, whereas RhoA activity was significantly decreased with Id-1 depletion. Inhibitors targeting Rac1, RhoA, and Rho kinase suppressed the invasiveness of Id-1-expressing NE2-hTERT cells. Knockdown of N-cadherin in Id-1-over-expressing cells inhibited cell invasiveness and down-regulated RhoA activity. These data suggest that the Id-1-induced invasive potential may be regulated through the N-cadherin-RhoA axis and Rac1 activation.


Asunto(s)
Cadherinas/metabolismo , Movimiento Celular , Transición Epitelial-Mesenquimal , Proteína 1 Inhibidora de la Diferenciación/metabolismo , Proteínas de Unión al GTP rho/metabolismo , Diferenciación Celular , Células Epiteliales/citología , Células Epiteliales/metabolismo , Células HeLa , Humanos , Transducción de Señal
6.
Acta Biomater ; 101: 168-182, 2020 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-31683015

RESUMEN

Invadosomes are invasive protrusions generated by cells which can secrete matrix metalloproteinases for focal digestion of extracellular matrix. They also aid invasive cancer cells in their transmigration through vascular endothelium. However, how the physical and chemical cues in a three-dimensional (3D) system signal the spatial localization of invadosomes remains largely unknown. Here we study the topographic guidance of invadosome formation in invasive nasopharyngeal cells under the stimulation of an inflammatory cytokine, TGF-ß1, using engineered gratings with different width and depth. We first report that TGF-ß1 can act as an external signal to upregulate the formation of invadosomes with a random distribution on a plane 2D surface. When the cells were seeded on parallel 3D gratings of 5 µm width and 1 µm depth, most of the invadosomes aligned to the edges of the gratings, indicating a topographic cue to the control of invadosome localization. While the number of invadosomes per cell were not upregulated when the cells were seeded on 3D topography, guidance of invadosomes localization to edges is correlated with cell migration directionality on 1 µm deep gratings. Invadosomes preferentially form at edges when the cells move at a lower speed and are guided along narrow gratings. The invadosomes forming at 3D edges also have a longer half-life than those forming on a plane surface. These data suggest that there are integrated biochemical and 3D geometric cues underlying the spatial regulation of invasive structures so as to elicit efficient invasion or metastasis of cells. STATEMENT OF SIGNIFICANCE: Nasopharyngeal cells were integrated with the biological cues and matrix topography to govern the activity and spatial distribution of invadosomes. The biochemical induction of invadosome formation by TGF-ß1 in nasopharyngeal cells was observed. When the cells were seeded on parallel 3D gratings, most of the invadosomes aligned to the edges of the gratings due to topographical induced invadosome localization. While the number of invadosomes per cell were not upregulated, guidance of invadosomes localization to edges is correlated with cell migration directionality on 1 µm deep gratings. Invadosomes preferentially form at edges with a higher stability when the cells are guided along narrow gratings. The integrated biochemical and 3D geometric cues could elicit efficient invasion or metastasis of cells.


Asunto(s)
Células Epiteliales/metabolismo , Matriz Extracelular/metabolismo , Modelos Biológicos , Neoplasias Nasofaríngeas/metabolismo , Podosomas/metabolismo , Factor de Crecimiento Transformador beta1/farmacología , Línea Celular Tumoral , Células Epiteliales/patología , Humanos , Neoplasias Nasofaríngeas/patología , Invasividad Neoplásica , Metástasis de la Neoplasia , Podosomas/patología
7.
J Virol ; 82(22): 11318-30, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18753196

RESUMEN

The production of virus-like particles (VLPs) constitutes a relevant and safe model to study molecular determinants of virion egress. The minimal requirement for the assembly of VLPs for the coronavirus responsible for severe acute respiratory syndrome in humans (SARS-CoV) is still controversial. Recent studies have shown that SARS-CoV VLP formation depends on either M and E proteins or M and N proteins. Here we show that both E and N proteins must be coexpressed with M protein for the efficient production and release of VLPs by transfected Vero E6 cells. This suggests that the mechanism of SARS-CoV assembly differs from that of other studied coronaviruses, which only require M and E proteins for VLP formation. When coexpressed, the native envelope trimeric S glycoprotein is incorporated onto VLPs. Interestingly, when a fluorescent protein tag is added to the C-terminal end of N or S protein, but not M protein, the chimeric viral proteins can be assembled within VLPs and allow visualization of VLP production and trafficking in living cells by state-of-the-art imaging technologies. Fluorescent VLPs will be used further to investigate the role of cellular machineries during SARS-CoV egress.


Asunto(s)
Proteínas de la Nucleocápside/metabolismo , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/fisiología , Proteínas del Envoltorio Viral/metabolismo , Proteínas de la Matriz Viral/metabolismo , Ensamble de Virus , Animales , Chlorocebus aethiops , Proteínas M de Coronavirus , Proteínas de la Nucleocápside de Coronavirus , Humanos , Glicoproteínas de Membrana/metabolismo , Microscopía Electrónica de Transmisión , Proteínas de la Nucleocápside/genética , Glicoproteína de la Espiga del Coronavirus , Células Vero , Proteínas del Envoltorio Viral/genética , Proteínas de la Matriz Viral/genética , Proteínas Viroporinas , Virosomas/metabolismo , Virosomas/ultraestructura
8.
Oncogene ; 26(21): 3069-80, 2007 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-17099724

RESUMEN

Epstein-Barr virus (EBV) infection is closely associated with nasopharyngeal carcinoma (NPC) and can be detected in early premalignant lesions of nasopharyngeal epithelium. The latent membrane protein 1 (LMP1) is an oncoprotein encoded by the EBV and is believed to play a role in transforming premalignant nasopharyngeal epithelial cells into cancer cells. RASSF1A is a tumor-suppressor gene commonly inactivated in many types of human cancer including NPC. In this study, we report a novel function of LMP1, in down-regulating RASSF1A expression in human epithelial cells. Downregulation of RASSF1A expression by LMP1 is dependent on the activation of intracellular signaling of NF-kappaB involving the C-terminal activating regions (CTARs) of LMP1. LMP1 expression also suppresses the transcriptional activity of the RASSF1A core promoter. RASSF1A stabilizes microtubules and regulates mitotic events. Aberrant mitotic spindles and chromosome aberrations are reported phenotypes in RASSF1A inactivated cells. In this study, we observed that LMP1 expression in human epithelial cells could induce aberrant mitotic spindles, disorganized interphase microtubules and aneuploidy. LMP1 expression could also suppress microtubule dynamics as exemplified by tracking movements of the growing tips of microtubules in live cells by transfecting EGFP-tagged EB1 into cells. The aberrant mitotic spindles and interphase microtubule organization induced by LMP1 could be rescued by transfecting RASSF1A expression plasmid into cells. Downregulation of RASSF1A expression by LMP1 may facilitate its role in transformation of premalignant nasopharyngeal epithelial cells into cancer cells.


Asunto(s)
Aberraciones Cromosómicas , Regulación hacia Abajo/genética , Células Epiteliales/metabolismo , Células Epiteliales/patología , Microtúbulos/metabolismo , Proteínas Supresoras de Tumor/antagonistas & inhibidores , Proteínas Supresoras de Tumor/genética , Proteínas de la Matriz Viral/fisiología , Línea Celular , Línea Celular Transformada , Línea Celular Tumoral , Células HeLa , Humanos , Microtúbulos/patología , FN-kappa B/fisiología , Proteínas Supresoras de Tumor/biosíntesis
9.
Oncogene ; 26(1): 148-57, 2007 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-16799631

RESUMEN

A gene critical to esophageal cancer has been identified. Functional studies using microcell-mediated chromosome transfer of intact and truncated donor chromosomes 3 into an esophageal cancer cell line and nude mouse tumorigenicity assays were used to identify a 1.61 Mb tumor suppressive critical region (CR) mapping to chromosome 3p14.2. This CR is bounded by D3S1600 and D3S1285 microsatellite markers. One candidate tumor suppressor gene, ADAMTS9, maps to this CR. Further studies showed normal expression levels of this gene in tumor-suppressed microcell hybrids, levels that were much higher than observed in the recipient cells. Complete loss or downregulation of ADAMTS9 gene expression was found in 15 out of 16 esophageal carcinoma cell lines. Promoter hypermethylation was detected in the cell lines that do not express this gene. Re-expression of ADAMTS9 was observed after demethylation drug treatment, confirming that hypermethylation is involved in gene downregulation. Downregulation of ADAMTS9 was also found in 43.5 and 47.6% of primary esophageal tumor tissues from Hong Kong and from the high-risk region of Henan, respectively. Thus, this study identifies and provides functional evidence for a CR associated with tumor suppression on 3p14.2 and provides the first evidence that ADAMTS9, mapping to this region, may contribute to esophageal cancer development.


Asunto(s)
Proteínas ADAM/genética , Carcinoma de Células Escamosas/genética , Cromosomas Humanos Par 3 , Neoplasias Esofágicas/genética , Genes Supresores de Tumor , Proteína ADAMTS9 , Secuencia de Bases , Mapeo Cromosómico , ADN , Metilación de ADN , Regulación Neoplásica de la Expresión Génica , Humanos , Hibridación Fluorescente in Situ , Datos de Secuencia Molecular
10.
Oncogene ; 26(40): 5877-88, 2007 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-17384685

RESUMEN

By comparative DNA fingerprinting, we identified a 357-bp DNA fragment frequently amplified in esophageal squamous cell carcinomas (ESCC). This fragment overlaps with an expressed sequence tag mapped to 7q22. Further 5' and 3'-rapid amplification of cDNA ends revealed that it is part of a novel, single-exon gene with full-length mRNA of 2052 bp and encodes a nuclear protein of 109 amino acids ( approximately 15 kDa). This gene, designated as gene amplified in esophageal cancer 1 (GAEC1), was located within a 1-2 Mb amplicon at 7q22.1 identified by high-resolution 1 Mb array-comparative genomic hybridization in 6/10 ESCC cell lines. GAEC1 was ubiquitously expressed in normal tissues including esophageal and gastrointestinal organs; with amplification and overexpression in 6/10 (60%) ESCC cell lines and 34/99 (34%) primary tumors. Overexpression of GAEC1 in 3T3 mouse fibroblasts caused foci formation and colony formation in soft agar, comparable to H-ras and injection of GAEC1-transfected 3T3 cells into athymic nude mice formed undifferentiated sarcoma in vivo, indicating that GAEC1 is a transforming oncogene. Although no significant correlation was observed between GAEC1 amplification and clinicopathological parameters and prognosis, our study demonstrated that overexpressed GAEC1 has tumorigenic potential and suggest that overexpressed GAEC1 may play an important role in ESCC pathogenesis.


Asunto(s)
Carcinoma de Células Escamosas/genética , Núcleo Celular/metabolismo , Cromosomas Humanos Par 7 , Neoplasias Esofágicas/genética , Regulación Neoplásica de la Expresión Génica , Proteínas Nucleares/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Carcinoma de Células Escamosas/metabolismo , Línea Celular Tumoral , Neoplasias Esofágicas/metabolismo , Humanos , Ratones , Ratones Desnudos , Modelos Genéticos , Datos de Secuencia Molecular , Trasplante de Neoplasias , Proteínas Nucleares/biosíntesis
11.
Gene Ther ; 15(3): 191-202, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17972920

RESUMEN

To induce RNA interference (RNAi), either small interfering RNAs (siRNAs) are directly introduced into the cell or short hairpin RNAs (shRNAs) are expressed from a DNA vector. At present, shRNAs are commonly synthesized by RNA polymerase III (Pol III) promoters of the H1 and U6 RNAs. In this study, we designed and characterized a new set of plasmid vectors driven by promoters of the Epstein-Barr virus (EBV)-encoded small RNAs (EBERs). The EBERs are the most abundant transcript in infected cells and they are transcribed by Pol III. We showed that the EBER promoters were able to drive the expression of shRNA fusion transcripts. siRNAs processed from these fusion transcripts specifically and effectively inhibited the expression of homologous reporter or endogenous genes in various types of cells. The partial EBER sequences in the fusion transcripts did not activate double-stranded RNA-dependent protein kinase or suppress RNAi. In nasopharyngeal carcinoma cells, the EBER2 promoter was stronger than the H1 and U6 promoters in shRNA synthesis, leading to more effective knockdown of the target genes. Taken together, our findings suggest that the EBER promoters fundamentally different from those of H1 and U6 can be used to drive the intracellular expression of shRNAs for effective silencing of target genes in mammalian cells and particularly, in EBV-infected cells.


Asunto(s)
Terapia Genética/métodos , Herpesvirus Humano 4/genética , Neoplasias Nasofaríngeas/terapia , Regiones Promotoras Genéticas , Interferencia de ARN , ARN Interferente Pequeño , Línea Celular Tumoral , Estudios de Factibilidad , Expresión Génica , Silenciador del Gen , Ingeniería Genética , Vectores Genéticos/farmacología , Humanos , Neoplasias Nasofaríngeas/metabolismo , ARN Polimerasa III/genética , ARN Mensajero/análisis , Proteínas Recombinantes de Fusión/genética
12.
Int J Oncol ; 33(4): 713-24, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18813784

RESUMEN

Mutation of the p53 gene is a common event in human cancer. Interestingly, p53 mutation is uncommon in nasopharyngeal carcinoma (NPC). The DeltaNp63 has been postulated to have a dominant-negative effect on the function of the p53 gene and may play a role in the pathogenesis of nasopharyngeal carcinoma. Immortalization is a common property of cancer cells and is believed to be an early event in carcinogenesis. At present, the relationship between DeltaNp63 and immortalization is poorly understood. In this study, we defined the expression profile of p63 and its various isoforms in primary and immortalized nasopharyngeal epithelial cells. Also, we elucidated some events regulating the expression of p63. Elevated expression of p63 was generally detected in both primary and immortalized nasopharyngeal epithelial cells at their proliferation stage and the predominant isoform of p63 expressed was DeltaNp63alpha. p63 expression was suppressed upon cellular senescence of primary nasopharyngeal epithelial cells and induction of terminal differentiation in immortalized nasopharyngeal epithelial cells. Expression of DeltaNp63 alone was able to drive clonal proliferation in primary nasopharyngeal cells in culture while downregulation of DeltaNp63 induced cellular apoptosis. All these results support a role of DeltaNp63 in proliferation and immortalization which facilitates pathogenesis of nasopharyngeal carcinoma. TGFbeta and retinoic acid downregulated the expression of p63 in immortalized nasopharyngeal epithelial cells and may play a role in regulating differentiation in squamous epithelial cells with potential applications in prevention and treatment of nasopharyngeal carcinoma.


Asunto(s)
Carcinoma de Células Escamosas/tratamiento farmacológico , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Regulación Neoplásica de la Expresión Génica , Mutación , Neoplasias Nasofaríngeas/tratamiento farmacológico , Transactivadores/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Carcinoma de Células Escamosas/patología , Línea Celular , Proliferación Celular , Senescencia Celular , ADN Complementario/metabolismo , Humanos , Modelos Biológicos , ARN/metabolismo , Retroviridae/genética , Factores de Transcripción
13.
Histopathology ; 52(2): 167-74, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18184265

RESUMEN

AIMS: Hydatidiform mole (HM) is the most common type of gestational trophoblastic disease. A proportion of patients with HM develop gestational trophoblastic neoplasia (GTN) requiring chemotherapy. The aim was to identify differentially expressed genes that are associated with development of GTN. METHODS AND RESULTS: Using cDNA microarray, differential expression of prostate stem cell antigen (PSCA) was identified in HMs that developed GTN compared with those that spontaneously regressed. Significant overexpression of PSCA RNA (P = 0.037) and protein (P < 0.05) in aggressive HM was verified by real-time polymerase chain reaction (PCR) and immunohistochemical analysis in 10 first-trimester placentas, 36 HM that subsequently regressed and 11 HM that developed GTN. A high level of PSCA expression was also found in three choriocarcinomas and three placental site trophoblastic tumours. A positive correlation was observed between PSCA expression and proliferation and apoptotic indices as assessed by Ki67 (P = 0.01), mcm7 (P = 0.001) and M30 (P = 0.016), as well as p53 (P < 0.01), p21(WAF1/CIP1) (P < 0.01) and mdm2 (P = 0.002) expression. CONCLUSIONS: Overexpression of PSCA is associated with development of GTN in HM. PSCA probably plays a role in the regulation of cell growth through p53-related signaling pathways.


Asunto(s)
Enfermedad Trofoblástica Gestacional/metabolismo , Mola Hidatiforme/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias de la Próstata/metabolismo , Adolescente , Adulto , Antígenos de Neoplasias , Biomarcadores de Tumor/metabolismo , Proliferación Celular , ADN de Neoplasias/genética , Femenino , Proteínas Ligadas a GPI , Regulación Neoplásica de la Expresión Génica , Enfermedad Trofoblástica Gestacional/genética , Enfermedad Trofoblástica Gestacional/patología , Humanos , Mola Hidatiforme/genética , Mola Hidatiforme/patología , Masculino , Glicoproteínas de Membrana/genética , Persona de Mediana Edad , Proteínas de Neoplasias/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Embarazo , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Transducción de Señal/genética , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
14.
Oncogene ; 25(7): 1070-80, 2006 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-16247458

RESUMEN

Protocadherins constitute the largest subgroup in the cadherin superfamily of cell adhesion molecules. Their major functions are poorly understood, although some are implicated in nervous system development. As tumor-specific promoter methylation is a marker for tumor suppressor genes (TSG), we searched for epigenetically inactivated TSGs using methylation-subtraction combined with pharmacologic demethylation, and identified the PCDH10 CpG island as a methylated sequence in nasopharyngeal carcinoma (NPC). PCDH10 is broadly expressed in all normal adult and fetal tissues including the epithelia, though at different levels. It resides at 4q28.3--a region with hemizygous deletion detected by array-CGH in NPC cell lines; however, PCDH10 itself is not located within the deletion. In contrast, its transcriptional silencing and promoter methylation were frequently detected in multiple carcinoma cell lines in a biallelic way, including 12/12 nasopharyngeal, 13/16 esophageal, 3/4 breast, 5/5 colorectal, 3/4 cervical, 2/5 lung and 2/8 hepatocellular carcinoma cell lines, but not in any immortalized normal epithelial cell line. Aberrant methylation was further frequently detected in multiple primary carcinomas (82% in NPC, 42-51% for other carcinomas), but not normal tissues. The transcriptional silencing of PCDH10 could be reversed by pharmacologic demethylation with 5-aza-2'-deoxycytidine or genetic demethylation with double knockout of DNMT1 and DNMT3B, indicating a direct epigenetic mechanism. Ectopic expression of PCDH10 strongly suppressed tumor cell growth, migration, invasion and colony formation. Although the epigenetic and genetic disruptions of several classical cadherins as TSGs have been well documented in tumors, this is the first report that a widely expressed protocadherin can also function as a TSG that is frequently inactivated epigenetically in multiple carcinomas.


Asunto(s)
Cadherinas/genética , Carcinoma/genética , Epigénesis Genética , Neoplasias Esofágicas/genética , Genes Supresores de Tumor , Neoplasias Nasofaríngeas/genética , Azacitidina/análogos & derivados , Azacitidina/farmacología , Movimiento Celular/genética , ADN (Citosina-5-)-Metiltransferasa 1 , ADN (Citosina-5-)-Metiltransferasas/antagonistas & inhibidores , Metilación de ADN/efectos de los fármacos , Decitabina , Silenciador del Gen/efectos de los fármacos , Humanos , Regiones Promotoras Genéticas , Protocadherinas , Eliminación de Secuencia , Transcripción Genética/genética , ADN Metiltransferasa 3B
15.
Oncogene ; 25(2): 310-6, 2006 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-16116475

RESUMEN

RASSF1A is a tumor suppressor gene on 3p21.3 frequently inactivated by promoter hypermethylation in nasopharyngeal carcinoma (NPC). To identify RASSF1A target genes in NPC, we have investigated the expression profile of the stable RASSF1A transfectants and controls by high-density oligonucleotide array. A total of 57 genes showed differential expression in the RASSF1A-expressing cells. These RASSF1A target genes were involved in multiple cellular regulatory processes such as transcription, signal transduction, cell adhesion and RNA processing. The RASSF1A-modulated expression of eight selected genes with the highest fold changes (ATF5, TCRB, RGS1, activin betaE, HNRPH1, HNRPD, Id2 and CKS2) by RASSF1A was confirmed in both stable and transient transfectants. Compared with the RASSF1A transfectants, an inverse expression pattern of activin betaE, Id2 and ATF5 was shown in the immortalized nasopharyngeal epithelial cells treated with siRNA against RASSF1A. The findings imply that the expression of activin betaE, Id2 and ATF5 was tightly regulated by RASSF1A and may associate with its tumor suppressor function. Strikingly, overexpression of Id2 is common in NPC and RASSF1A-induced repression of Id2 was mediated by the overexpression of activin betaE. The results suggest a novel RASSF1A pathway in which both activin betaE and Id2 are involved.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Subunidades beta de Inhibinas/metabolismo , Proteína 2 Inhibidora de la Diferenciación/metabolismo , Neoplasias Nasofaríngeas/genética , Transducción de Señal , Proteínas Supresoras de Tumor/metabolismo , Biomarcadores de Tumor/genética , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Humanos , Subunidades beta de Inhibinas/genética , Proteína 2 Inhibidora de la Diferenciación/genética , Neoplasias Nasofaríngeas/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección , Células Tumorales Cultivadas , Proteínas Supresoras de Tumor/antagonistas & inhibidores , Proteínas Supresoras de Tumor/genética
16.
Int J Mol Med ; 19(6): 915-23, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17487424

RESUMEN

Esophageal squamous cell carcinoma (ESCC) shows high frequency and mortality in Asian regions, including China. Previous analysis of genomic DNA of ESCC using comparative genomic hybridization indicated that amplification of the chromosome 5p regions is a common event in ESCC cell lines and patient cases of Hong Kong Chinese origin, and the results suggested that the genes located in the chromosome 5p regions may play crucial roles in the molecular pathogenesis of ESCC. Our previous studies on ESCC confirmed the tumorigenic and overexpression properties of a novel gene JS-1 located in chromosome 5p15.2 upstream to delta-catenin. In the present study, another novel gene JK-1 which is located at 5p15.1 downstream to delta-catenin was characterized for its roles in the pathogenesis of ESCC. Thirteen ESCC cell lines and 30 surgical specimens of esophageal tumors were studied for the overexpression of JK-1 using multiplex RT-PCR analysis. The transforming capacity of overexpression of JK-1 was also investigated by transfecting NIH 3T3 and HEK 293 cells with the expression vector cloned with JK-1, followed by the soft agar and foci formation assays. JK-1 was overexpressed in 9/13 (69%) of the ESCC cell lines and 9/30 (30%) of the ESCC patient cases. Both NIH 3T3 and HEK 293 cells acquired the properties of anchorage-dependent and -independent growth when JK-1 was overexpressed. Most significantly, subcutaneous sarcomas were formed in all (3/3) the athymic nude mice after NIH 3T3 cells overexpressing JK-1 were injected subcutaneously. Our results thus indicated that JK-1 is commonly overexpressed in ESCC and has a prominent capacity to transform normal cells. Our overall results thus provide the first evidence that the overexpression of JK-1 and its transforming capacity in normal cells may play a critical role in the molecular pathogenesis of ESCC.


Asunto(s)
Carcinoma de Células Escamosas/genética , Cromosomas Humanos Par 5 , Neoplasias Esofágicas/genética , Regulación Neoplásica de la Expresión Génica , Genes Relacionados con las Neoplasias , Proteínas de Neoplasias/genética , Anciano , Animales , Pruebas de Carcinogenicidad , Mapeo Cromosómico , Femenino , Humanos , Péptidos y Proteínas de Señalización Intracelular , Masculino , Proteínas de la Membrana , Ratones , Ratones Desnudos , Persona de Mediana Edad , Células 3T3 NIH , Oncogenes/fisiología , Células Tumorales Cultivadas
17.
Int J Mol Med ; 19(6): 953-60, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17487429

RESUMEN

Previous studies have shown that the anomalous fruit extract of Gleditsia sinensis (GSE) exhibited apoptotic properties in various solid and non-solid tumors in vitro. However, the inhibitory actions of GSE on oncogenic expression and telomerase activity in esophageal squamous cell carcinoma (ESCC) have not been studied before. In the present study, the anti-cancer effects of GSE were demonstrated in three ESCC cell lines (HKESC-1, HKESC-2 and SLMT-1) by MTS and anchorage-independent clongen-icity assays, expression studies on oncogenes at 11q13 (CCND1, INT2, FGF4 and EMS1) and real-time quantitative telomeric repeat amplification protocol assay to show the inhibitory effect of GSE on telomerase in ESCC. The means of MTS50 of GSE for the ESCC cell lines and non-tumor NIH 3T3 cells were 21 and 163 microg/ml respectively. The anchorage-independent clongenicity assay showed that SLMT-1 cells lost their colony-forming potential which was dose-dependent to GSE. Moreover, GSE demonstrated dose-dependent suppression on the expression of INT2, EMS1 and FGF4, and inhibition of telomerase activity in the ESCC cell lines. Our overall results thus provide the first evidence that the anti-cancer effects of GSE on ESCC involve the suppression of oncogenic expression and inhibition of telomerase activity. Our findings also offer a new opportunity for the future development of GSE as a novel anti-cancer agent for ESCC and possibly for other cancers.


Asunto(s)
Carcinoma de Células Escamosas/genética , Neoplasias Esofágicas/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Gleditsia , Extractos Vegetales/farmacología , Telomerasa/antagonistas & inhibidores , Telomerasa/genética , Animales , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Neoplasias Esofágicas/patología , Frutas/química , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Gleditsia/química , Humanos , Ratones , Células 3T3 NIH , Telomerasa/metabolismo
18.
Cell Death Differ ; 24(6): 1133, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-27636105

RESUMEN

Retraction to: Cell Death Differ 2016;23(9):1471­1482. doi:10.1038/cdd.2016.32

19.
Oncogene ; 36(28): 3986-4000, 2017 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-28288140

RESUMEN

Esophageal cancer is one of the most lethal cancers worldwide with poor survival and limited therapeutic options. The discovery of microRNAs created a new milestone in cancer research. miR-377 is located in chromosome region 14q32, which is frequently deleted in esophageal squamous cell carcinoma (ESCC), but the biological functions, clinical significance and therapeutic implication of miR-377 in ESCC are largely unknown. In this study, we found that miR-377 expression was significantly downregulated in tumor tissue and serum of patients with ESCC. Both tumor tissue and serum miR-377 expression levels were positively correlated with patient survival. Higher serum miR-377 expression was inversely associated with pathologic tumor stage, distant metastasis, residual tumor status and chemoradiotherapy resistance. The roles of miR-377 in suppressing tumor initiation and progression, and the underlying molecular mechanisms were investigated. Results of in vitro and in vivo experiments showed that miR-377 overexpression inhibited the initiation, growth and angiogenesis of ESCC tumors as well as metastatic colonization of ESCC cells, whereas silencing of miR-377 had opposite effects. Mechanistically, miR-377 regulated CD133 and VEGF by directly binding to their 3' untranslated region. Moreover, systemic delivery of formulated miR-377 mimic not only suppressed tumor growth in nude mice but also blocked tumor angiogenesis and metastasis of ESCC cells to the lungs without overt toxicity to mice. Collectively, our study established that miR-377 plays a functional and significant role in suppressing tumor initiation and progression, and may represent a promising non-invasive diagnostic and prognostic biomarker and therapeutic strategy for patients with ESCC.


Asunto(s)
Antígeno AC133/genética , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patología , Transformación Celular Neoplásica/genética , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/patología , MicroARNs/fisiología , Factor A de Crecimiento Endotelial Vascular/genética , Adulto , Anciano , Anciano de 80 o más Años , Animales , Carcinoma de Células Escamosas/diagnóstico , Carcinoma de Células Escamosas/mortalidad , Estudios de Casos y Controles , Línea Celular Tumoral , Progresión de la Enfermedad , Regulación hacia Abajo/genética , Neoplasias Esofágicas/diagnóstico , Neoplasias Esofágicas/mortalidad , Carcinoma de Células Escamosas de Esófago , Femenino , Regulación Neoplásica de la Expresión Génica , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos NOD , Ratones Desnudos , Ratones SCID , Persona de Mediana Edad
20.
Cancer Lett ; 238(1): 90-103, 2006 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-16054293

RESUMEN

Epstein-Barr virus (EBV) infection has been implicated in the carcinogenesis of several types of human cancer, including gastric cancer. In contrast to two other EBV-related malingancies, nasopharyngeal carcinoma and Hodgkins Lympomain which the latent membrane protein (LMP)-1 is often detected, in gastric cancer, BARF1, one of the early EBV genes, is frequently expressed in EBV-positive specimens. This indicates that expression of BARF1 may play a positive role in the development of gastric cancer. The aim of this study was to investigate the effect of BARF1 expression in gastric cancer cells. First, a retroviral vector containing the full length BARF1 gene was transfected into an EBV negative gastric cancer cell line, BGC823, and stable transfectants expressing ectopic BARF1 were generated. Microarray analysis was then performed and gene expression profiles were analysed and compared between the cells expressing ectopic BARF1 and the vector control. In addition, the effect of BARF1 on gastric cancer cell proliferation and apoptosis was investigated by MTT assay, DAPI staining, flow cytometry as well as Western blotting. We found that expression of BARF1 in gastric cancer cells led to significant alterations of gene expression, especially genes related to proliferation and apoptosis. In addition, the BARF1 expressing cells were more resistant to apoptosis induced by a commonly used anticancer drug, taxol. This chemo-protective effect of BARF1 was associated with increased Bcl-2 and Bax ratio and decreased expression of cleaved PARP, but not alterations in cell proliferation. Our results suggest that BARF1 expression in gastric cancer cells may provide a protective role against apoptosis through an increased Bcl-2 to Bax ratio, thus promoting cancer cell survival.


Asunto(s)
Apoptosis/genética , Neoplasias Gástricas/metabolismo , Proteínas Virales/fisiología , Antineoplásicos Fitogénicos/farmacología , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/genética , Técnicas de Transferencia de Gen , Vectores Genéticos/genética , Humanos , Análisis de Secuencia por Matrices de Oligonucleótidos , Paclitaxel/farmacología , Neoplasias Gástricas/tratamiento farmacológico , Neoplasias Gástricas/genética , Proteínas Virales/genética , Proteínas Virales/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA