Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cancer ; 119(2): 304-12, 2013 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-22833454

RESUMO

BACKGROUND: Aberrant methylation of tumor-related genes has been reported in Epstein-Barr virus (EBV)-associated gastric cancers. This study sought to profile EBV-driven hypermethylation in EBV-infected cells. METHODS: The EBV-positive AGS gastric cancer cell line (AGS-EBV) and EBV-negative AGS cells were used in this study. DNA methyltransferase-3b (DNMT3b) activity was assessed by EpiQuick activity assay, and genome-wide DNA methylation profiles were assessed by methyl-DNA immunoprecipitation microarray assay. RESULTS: EBV infection was confirmed in AGS-EBV cells by EBV-encoded RNA in situ hybridization. Expression and activity of DNA methyltransferase-3b (DNMT3b) was significantly increased in AGS-EBV compared to AGS. Ectopic expression of LMP2A (latent membrane protein 2A) in AGS increased activity of DNMT3b. A total of 1065 genes were differentially methylated by EBV infection (fold-changes ≥ 2, P < .05) in AGS-EBV compared to AGS cells. The majority of the differentially methylated genes (83.2%, 886 of 1065 genes) had cytosine-guanine dinucleotide (CpG) hypermethylation in AGS-EBV (fold-changes 2.43∼65.2) versus that found in AGS cells. Gene ontology analysis revealed that hypermethylated genes were enriched in the important cancer pathways (≥ 10 genes each, P ≤ .05) including mitogen-activated protein kinase signaling, cell adhesion molecules, wnt signaling pathway, and so forth. Six novel hypermethylated candidates (IL15RA, REC8, SSTR1, EPHB6, MDGA2, and SCARF2) were further validated. Higher levels of DNA methylation were confirmed for all these genes in AGS-EBV cells by bisulfite genomic sequencing. Furthermore, these candidates were silenced or down-regulated in AGS-EBV cells, but can be restored by demethylation treatment. CONCLUSIONS: EBV infection in AGS cells induced aberrant CpG hypermethylation of 886 genes involving in important cancer-related pathways. Induction of promoter methylation by EBV is regulated by up-regulation of DNMT3b through LMP2A.


Assuntos
Metilação de DNA , Infecções por Vírus Epstein-Barr/virologia , Herpesvirus Humano 4/fisiologia , Neoplasias Gástricas/genética , Proteínas de Ciclo Celular/genética , Linhagem Celular Tumoral , Ilhas de CpG , DNA (Citosina-5-)-Metiltransferases/genética , DNA (Citosina-5-)-Metiltransferases/metabolismo , Ativação Enzimática , Epigênese Genética , Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Genoma Humano , Interações Hospedeiro-Patógeno , Humanos , Regiões Promotoras Genéticas , Receptores Proteína Tirosina Quinases/genética , Receptores da Família Eph , Receptores de Interleucina-15/genética , Receptores de Somatostatina/genética , Análise de Sequência de DNA , Neoplasias Gástricas/virologia , Proteínas da Matriz Viral/genética , Proteínas da Matriz Viral/metabolismo , DNA Metiltransferase 3B
2.
Cancer ; 119(6): 1217-26, 2013 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-23212831

RESUMO

BACKGROUND: Previous reports from these authors found that activation of peroxisome proliferator-activated receptor gamma (PPARγ) suppressed hepatocellular carcinoma (HCC). This study sought to identify the molecular target of PPARγ and characterize its antitumor effect in HCC. METHODS: Optimal PPARγ binding activity was obtained using the PPARγ agonist rosiglitazone (100 µM) as determined by enzyme-linked immunosorbent assay. Under PPARγ activation, 114 PPARγ downstream targets associated with cancer development were identified by oligonucleotide microarray and Gene Ontology analysis. Among them, Cbp/p300-interacting transactivator, with Glu/Asp-rich carboxy-terminal domain, 2 (CITED2) was the most prominent PPARγ-bound target, as determined by chromatin immunoprecipitation-polymerase chain reaction. RESULTS: CITED2 messenger RNA and protein was significantly down-regulated in primary HCCs compared with their adjacent nontumor tissues. PPARγ induced expression of CITED2 in HCC cell lines after adenovirus-PPARγ transduction. The biological function of CITED2 was evaluated by loss- and gain-of-function assays. CITED2 knockdown in the hepatocyte cell line LO2 and HCC cell line Hep3B significantly increased cell viability and clonogenicity, and promoted G1 -S phase transition in both cell lines. In contrast, ectopic expression of CITED2 in HepG2 and BEL7404 HCC cell lines significantly suppressed cell growth. The tumor suppressive effect of CITED2 was associated with up-regulation of cyclin-dependent kinase inhibitors p15(INK4B) , p21(Wat1/Cip1) , p27(Kip1) , antiproliferative regulator interferon alpha 1, proapoptotic mediators including tumor necrosis factor receptor superfamily member 1A (TNFRSF1A), TNFRSF25, caspase-8, granzyme A, and the tumor suppressor gene maspin. CITED2 was also associated with the down-regulation of cell cycle regulator cyclin D1, oncogene telomerase reverse transcriptase, and proinvasion/metastasis gene matrix metallopeptidase 2. CONCLUSIONS: CITED2 is a direct effector of PPARγ for tumor suppression. Cancer 2013. © 2012 American Cancer Society.


Assuntos
Carcinoma Hepatocelular/metabolismo , PPAR gama/metabolismo , Proteínas Repressoras/fisiologia , Transativadores/fisiologia , Carcinoma Hepatocelular/patologia , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células , Quinases Ciclina-Dependentes/metabolismo , Regulação para Baixo , Técnicas de Silenciamento de Genes , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Rosiglitazona , Tiazolidinedionas/farmacologia , Transfecção
3.
Cancer ; 118(4): 924-36, 2012 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-21717425

RESUMO

BACKGROUND: The role of Epstein-Barr virus (EBV) infection in gastric carcinogenesis remains largely unknown. The authors studied the effects of zinc finger E-box binding factor 1 (ZEB1) on latent-lytic switch of EBV infection in gastric cancer and explored the importance of EBV in gastric carcinogenesis. METHODS: Loss or gain of ZEB1 function was obtained by ZEB1 small-interfering RNA (siRNA) knock-down or forced ZEB1 re-expression. Cell growth was evaluated by cell viability and colony formation assays, and the cell cycle was assessed by flow cytometry. EBV was detected using quantitative polymerase chain reaction (PCR) and in situ hybridization analyses. RESULTS: ZEB1 knock-down in a latent EBV-infected gastric cancer cell line (YCC10) increased lytic gene BamHI W leftward reading frame 1 (BZLF1) expression and decreased the expression of latent gene EB nuclear antigen 1 (EBNA1) concomitant with the inhibition of cell viability (P < .05) and S-phase DNA synthesis (P < .01). ZEB1 depletion combined with ganciclovir revealed a further reduction in cell viability (P < .001). ZEB1 knock-down induced cell apoptosis and the up-regulation of caspase 3 and poly(adenosine diphosphate-ribose) polymerase cleavage. Conversely, ectopic overexpression of ZEB1 in a lytic EBV-infected gastric cancer cell line (AGS-EBV) inhibited BZLF1 promoter (Zp) activity, BZLF1 expression, and apoptosis and promoted cell growth. EBV infection was detected in 11.3% (80 of 711) of gastric cancers. The presence of EBV was associated with age, men, and intestinal type cancer. CONCLUSIONS: ZEB1 was confirmed as a key mediator of the latent-lytic switch of EBV-associated gastric cancer, a distinct subtype with different clinicopathologic features. The current results indicated that inhibition of ZEB1 may be a potential target for EBV-associated gastric cancer therapy.


Assuntos
Infecções por Vírus Epstein-Barr/complicações , Herpesvirus Humano 4/metabolismo , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/virologia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Adulto , Idoso , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Apoptose , Linhagem Celular Tumoral , Sobrevivência Celular , China , DNA Viral/metabolismo , Infecções por Vírus Epstein-Barr/epidemiologia , Infecções por Vírus Epstein-Barr/metabolismo , Antígenos Nucleares do Vírus Epstein-Barr/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Herpesvirus Humano 4/genética , Proteínas de Homeodomínio/efeitos dos fármacos , Humanos , Incidência , Masculino , Pessoa de Meia-Idade , RNA Interferente Pequeno/farmacologia , Estudos Retrospectivos , Neoplasias Gástricas/tratamento farmacológico , Transativadores/metabolismo , Fatores de Transcrição/efeitos dos fármacos , Homeobox 1 de Ligação a E-box em Dedo de Zinco
4.
Cancer ; 118(4): 947-59, 2012 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-21717426

RESUMO

BACKGROUND: By using genome-wide methylation screening, the authors identified ring finger protein 180 (RNF180) as preferentially methylated in cancer. This study was undertaken to clarify its structure and functional role in gastric cancer. METHODS: The transcription start site and core functional promoter region of RNF180 were revealed by 5' rapid amplification of cDNA ends and luciferase activity assays. Promoter methylation was detected by combined bisulfite restriction analysis and bisulfite genomic sequencing. Cell growth was detected by colony formation assay, apoptosis by annexin V assay, and RNF180 target genes by cDNA microarray. RESULTS: The authors revealed the transcription start site of RNF180 gene and identified the functional core promoter region (-202/+372) in the CpG island, which could be silenced by in vitro methylation assay. RNF180 was silenced in 6 of 7 gastric cancer cell lines and significantly down-regulated in primary gastric cancers compared with adjacent normal tissues (P = .001). Loss of gene expression was associated with promoter methylation. Re-expression of RNF180 suppressed cell growth (P < .001) and induced apoptosis (P < .05), which were mediated by up-regulating the antiproliferation regulators MTSS1 and CDKN2A and the proapoptotic mediator TIMP3. Promoter methylation of RNF180 was detected in 76% (150 of 198) of primary gastric cancers and 55% (11 of 20) of intestinal metaplasia, but in none of 23 normal gastric tissues. Methylated RNF180 DNA was detected in the plasma of 56% of gastric cancer patients, but not in healthy controls (P = .003). Patients with low or loss of RNF180 expression had significantly poorer overall survival. CONCLUSIONS: RNF180 is a novel potential tumor suppressor in gastric carcinogenesis and has potential clinical utility as a biomarker for gastric cancer patients.


Assuntos
Apoptose/genética , Apoptose/fisiologia , Proliferação de Células , Domínios RING Finger/genética , Domínios RING Finger/fisiologia , Neoplasias Gástricas/genética , Neoplasias Gástricas/fisiopatologia , Idoso , Sequência de Bases , Estudos de Casos e Controles , Linhagem Celular Tumoral , Ilhas de CpG/genética , Metilação de DNA , DNA de Neoplasias/sangue , DNA de Neoplasias/genética , Regulação para Baixo , Feminino , Mucosa Gástrica/metabolismo , Mucosa Gástrica/patologia , Humanos , Técnicas In Vitro , Masculino , Pessoa de Meia-Idade , Dados de Sequência Molecular , Prognóstico , Neoplasias Gástricas/mortalidade , Taxa de Sobrevida , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/fisiologia
5.
Cancer Invest ; 29(8): 564-72, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21843047

RESUMO

We investigated the association of Interleukin (IL)-6-6331 polymorphisms with susceptibility to gastric cancer in 375 patients with gastric cancer and 386 age- and gender-matched healthy controls. After adjustment for the potential confounding effects of gender and age, IL-6-6331TC genotype was associated with a decreased risk of gastric cancer compared with the CC genotype. Further stratification analyses indicated that the protective effect of TC genotype was also observed in poorly differentiated gastric cancer, noncardia gastric cancer, and intestinal-type gastric cancer, respectively. These results suggest that the IL-6-6331 polymorphism is involved in susceptibility to developing gastric cancer.


Assuntos
Povo Asiático/genética , Predisposição Genética para Doença/genética , Interleucina-6/genética , Polimorfismo Genético , Neoplasias Gástricas/genética , Adulto , Idoso , Estudos de Casos e Controles , Feminino , Genótipo , Humanos , Masculino , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Neoplasias Gástricas/patologia
6.
Hepatology ; 51(6): 2008-19, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20512989

RESUMO

UNLABELLED: Although peroxisome proliferator-activated receptor gamma (PPARgamma) agonist have been shown to inhibit hepatocellular carcinoma (HCC) development, the role of PPARgamma in hepatocarcinogenesis remains unclear. We investigated the therapeutic efficacy of PPARgamma against HCC. PPARgamma-deficient (PPARgamma(+/-)) and wild-type (PPARgamma(+/+)) littermates were used in a diethylnitrosamine (DEN)-induced HCC model and treated with PPARgamma agonist (rosiglitazone) or the vehicle alone for 8 months. The effects of PPARgamma on HCC cell growth and apoptosis were examined using PPARgamma-expressing adenovirus (Ad-PPARgamma). PPARgamma(+/-) mice were more susceptible to DEN-induced HCC than PPARgamma(+/+) mice (94% versus 62%, P < 0.05), and rosiglitazone significantly reduced the incidence of HCC in PPARgamma(+/+) mice (vehicle 62% versus treatment 24%, P < 0.01), but not in PPARgamma(+/-) mice, indicating that PPARgamma suppresses hepatocellular carcinogenesis. A pronounced expression of PPARgamma was observed in a HCC cell line (Hep3B) infected with Ad-PPARgamma. Such induction markedly suppressed HCC cell viability (P < 0.01). Further, Hep3B infection with Ad-PPARgamma revealed a decreased proportion of cells in S-phase (12.92% versus 11.58%, P < 0.05), with arrest at G(2)/M phase (38.2% versus 55.68%, P < 0.001), and there was concomitant phosphorylation of the key G(2)/M phase inhibitors cdc25C and cdc2. PPARgamma overexpression increased cell apoptosis (21.47% versus 35.02%, P < 0.01), mediated by both extrinsic (Fas and tumor necrosis factor-alpha) and intrinsic (caspase-9, caspase-3, caspase-7, and poly[ADP-ribose] polymerase) pathways. Moreover, PPARgamma directly induced a putative tumor suppressor gene, growth differentiation factor-15. CONCLUSION: Loss of one PPARgamma allele is sufficient to enhance susceptibility to HCC. PPARgamma suppresses tumor cell growth through reducing cell proliferation and inducing G(2)/M phase arrest, apoptosis, and up-regulating growth differentiation factor-15. Thus, PPARgamma acts as a tumor-suppressor gene in the liver.


Assuntos
Carcinoma Hepatocelular/prevenção & controle , Hipoglicemiantes/uso terapêutico , Neoplasias Hepáticas Experimentais/prevenção & controle , PPAR gama/metabolismo , Tiazolidinedionas/uso terapêutico , Adenoviridae , Alquilantes , Animais , Apoptose , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Ciclo Celular , Linhagem Celular Tumoral , Proliferação de Células , Dietilnitrosamina , Perfilação da Expressão Gênica , Fator 15 de Diferenciação de Crescimento/metabolismo , Humanos , Neoplasias Hepáticas Experimentais/genética , Neoplasias Hepáticas Experimentais/metabolismo , Masculino , Camundongos , Camundongos Knockout , Análise de Sequência com Séries de Oligonucleotídeos , PPAR gama/agonistas , PPAR gama/genética , Rosiglitazona , Regulação para Cima
7.
Neoplasia ; 7(3): 200-6, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15799820

RESUMO

We investigated the epigenetic silencing and genetic changes of the RAS-associated domain family 1A (RASSF1A) gene and the O6-methylguanine-DNA methyltransferase (MGMT) gene in retinoblastoma. We extracted DNA from microdissected tumor and normal retina tissues of the same patient in 68 retinoblastoma cases. Promoter methylation in RASSF1A and MGMT was analyzed by methylation-specific PCR, RASSF1A sequence alterations in all coding exons by direct DNA sequencing, and RASSF1A expression by RT-PCR. Cell cycle staging was analyzed by flow cytometry. We detected RASSF1A promoter hypermethylation in 82% of retinoblastoma, in tumor tissues only but not in adjacent normal retinal tissue cells. There was no expression of RASSF1A transcripts in all hypermethylated samples, but RASSF1A transcripts were restored after 5-aza-2'-deoxycytidine treatment with no changes in cell cycle or apoptosis. No mutation in the RASSF1A sequence was found. MGMT hypermethylation was present in 15% of the retinoblastoma samples, and the absence of MGMT hypermethylation was associated (P = .002) with retinoblastoma at advanced Reese-Ellsworth tumor stage. Our results revealed a high RASSF1A hypermethylation frequency in retinoblastoma. The correlation of MGMT inactivation by promoter hypermethylation with lower-stage diseases indicated that MGMT hypermethylation provides useful prognostic information. Epigenetic mechanism plays an important role in the progression of retinoblastoma.


Assuntos
Metilação de DNA , O(6)-Metilguanina-DNA Metiltransferase/genética , Regiões Promotoras Genéticas , Retinoblastoma/genética , Proteínas Supressoras de Tumor/genética , Ciclo Celular , Linhagem Celular Tumoral , Proliferação de Células , Ilhas de CpG , DNA/metabolismo , Análise Mutacional de DNA , Progressão da Doença , Éxons , Citometria de Fluxo , Humanos , Lasers , Mutação , Retina/metabolismo , Retinoblastoma/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Análise de Sequência de DNA
8.
Invest Ophthalmol Vis Sci ; 45(10): 3404-9, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15452042

RESUMO

PURPOSE: To investigate the link between microsatellite instability and epigenetic silencing of the MLH1 gene in the human retinoblastoma genome. METHODS: Methylation at the 5' region of MLH1 was studied, along with its protein expression level by using immunohistochemical staining in 51 retinoblastoma tumors and 2 retinoblastoma cell lines. Also assessed was the genomic stability of 26 retinoblastoma DNAs from microdissected tumor tissue and matched normal retina tissue obtained from the same patient by microsatellite instability (MSI) analysis. The National Cancer Institute-designed reference panel, and 85 markers on chromosomes 1, 6, 9, and 13 were used. RESULTS: Hypermethylation of the MLH1 promoter was detected in the WERI-Rb1 cell line and in 34 (67%) of the 51 tumors, but not in cell line Y79 and the other 17 tumors. MLH1 hypermethylation was associated with null MLH1 protein expression (P < 0.0005) and with well-differentiated histology (P < 0.05). MSI at three markers (D2S123, D6S470, and D13S265) was frequently identified among 26 retinoblastoma specimens with matched normal DNA. Among these 26 retinoblastomas, high-frequency MSI (MSI-H) tumors were detected in 19% (5/26) and low-frequency MSI (MSI-L) in another 19% (5/26). The remaining 62% (15/26) were genetically stable (MSS). MSI status (MSS, MSI-L, and MSI-H) was not associated with MLH1 promoter hypermethylation (P = 0.088; Kruskal-Wallis test). CONCLUSIONS: Epigenetic silencing of the DNA repair gene MLH1 by promoter hypermethylation is a frequent event in retinoblastoma. The results showed that somatic genetic changes involving MSI occur in a subset of retinoblastoma and implicated the presence of a defective DNA mismatch repair pathway resulting in MSI in retinoblastoma.


Assuntos
Metilação de DNA , DNA de Neoplasias/genética , Instabilidade Genômica , Repetições de Microssatélites/genética , Proteínas de Neoplasias/genética , Neoplasias da Retina/genética , Retinoblastoma/genética , Proteínas Adaptadoras de Transdução de Sinal , Pareamento Incorreto de Bases/genética , Proteínas de Transporte , Pré-Escolar , Reparo do DNA/genética , Epigênese Genética , Feminino , Inativação Gênica , Humanos , Técnicas Imunoenzimáticas , Lactente , Masculino , Proteína 1 Homóloga a MutL , Proteínas de Neoplasias/metabolismo , Proteínas Nucleares , Reação em Cadeia da Polimerase , Regiões Promotoras Genéticas/genética , Neoplasias da Retina/metabolismo , Retinoblastoma/metabolismo , Células Tumorais Cultivadas
9.
Cancer Gene Ther ; 11(5): 325-32, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-15044958

RESUMO

Pigment epithelium-derived factor (PEDF) is a potent inhibitor of angiogenesis and an inducer of neural differentiation. We previously reported the loss of PEDF expression in glioma progression. In this study, we investigated whether PEDF overexpression could suppress glioma growth and invasion. Glioma cell line U251 was stably transfected with a full-length human PEDF expression vector. The expression and release of various cytokines and angiogenic factors into the medium were analyzed by real-time reverse transcription-polymerase chain reaction, enzyme-linked immunosorbent assay, and gelatin zymography. Apoptosis was checked by terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling. Growth inhibition was evaluated by using the in vitro Matrigel invasion. Tumorigenicity was examined in vivo by subcutaneous xenotransplantation into severe combined immunodeficient mice. In U251 cells overexpressing PEDF, thrombospondin-1 protein was upregulated (5.3-fold more), but the production of vascular endothelial growth factor (VEGF) (1.8-fold less) and basic fibroblast growth factor (2.5-fold less) was lower than in cells transfected with the vector only. PEDF also downregulated the production of matrix metalloproteinase-9. Conditioned medium collected from the PEDF-transfected U251 cells showed a significant reduction of VEGF expression. In vitro invasiveness was reduced by approximately 40%. PEDF expression prevented the growth of transfected cells and caused a significant increase in the percentage of cells undergoing apoptosis (50.4% in PEDF-transfected cells). Furthermore, the size of xenotransplants was significantly smaller. In conclusion, PEDF overexpression decreased malignancy, and this might be attributed to the promotion of apoptosis and the regulation of expression of angiogenic effectors. Thus, treatment with PEDF may be useful in patients with malignant gliomas. However, the mechanism of apoptosis induction needs to be investigated.


Assuntos
Apoptose/genética , Proteínas do Olho/biossíntese , Proteínas do Olho/genética , Glioma/genética , Glioma/patologia , Fatores de Crescimento Neural/biossíntese , Fatores de Crescimento Neural/genética , Serpinas/biossíntese , Serpinas/genética , Animais , Diferenciação Celular/genética , Divisão Celular/genética , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Terapia Genética , Glioma/metabolismo , Humanos , Metaloproteases/biossíntese , Camundongos , Camundongos SCID , Invasividade Neoplásica/genética , Invasividade Neoplásica/prevenção & controle , Transplante de Neoplasias , Neovascularização Patológica/genética , Trombospondina 1/biossíntese , Fator A de Crescimento do Endotélio Vascular/biossíntese
10.
Mol Vis ; 9: 502-7, 2003 Oct 07.
Artigo em Inglês | MEDLINE | ID: mdl-14551532

RESUMO

PURPOSE: To explore the presence of common genetic alterations in retinoblastoma and to localize the altered genomic regions. METHODS: Genetic analysis included determinations of the loss of heterozygosity (LOH) and microsatellite instability (MSI) on chromosomes 19, 20, 21, 22, and X. Investigations were carried out among 15 microdissected retinoblastoma tumors and corresponding genomic DNA specimens. RESULTS: Among the 15 retinoblastoma cases, 73% (11/15) showed genome instability (LOH and/or MSI) at one or more loci on the 5 chromosomes, although loci with recurrent LOH was infrequent. The loss of a single allele was more frequent in chromosomes 19 (33%) and 20 (27%) than the other 3 chromosomes. Five loci with recurrent allelic loss were identified, among them the most frequent allelic losses were between D19S902 and D19S571 on 19q13 and were identified in 3 out of the 15 tumor specimens. The results suggested that gene loci in the 19q13 region may be associated with tumor development in retina. In addition, 3 specimens showed moderate frequency of LOH and/or MSI in more than 6 microsatellite markers, indicating genomic instability to occur at least in a subset of retinoblastoma. CONCLUSIONS: Our results provide the first evidence of LOH in chromosomes 19 and 20 in retinoblastoma. They also support the proposition that presence of genome instability in retinoblastoma may play a role in the tumorigenesis or progression of retinoblastoma.


Assuntos
Cromossomos Humanos/genética , Perda de Heterozigosidade , Neoplasias da Retina/genética , Retinoblastoma/genética , Pré-Escolar , Cromossomos Humanos Par 19/genética , Cromossomos Humanos Par 20/genética , Cromossomos Humanos Par 21/genética , Cromossomos Humanos Par 22/genética , Cromossomos Humanos X/genética , DNA de Neoplasias/genética , Feminino , Humanos , Lactente , Masculino , Repetições de Microssatélites/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...