Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Dig Dis Sci ; 64(8): 2147-2157, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-30788686

RESUMO

BACKGROUND: Histone methylation, as an essential pattern of posttranslational modifications, contributes to multiple cancer-related biological processes. Dysregulation of histone methylation is now considered a biomarker for cancer prognosis. AIMS: This study investigated and evaluated the potential role of four histone lysine trimethylation markers as biomarkers for esophageal squamous cell carcinoma (ESCC) prognosis. METHODS: Tissue arrays were made from 135 paraffin-embedded ESCC samples and examined for histone markers by immunohistochemistry, and 10 pairs of cancer and noncancerous mucosa tissues from ESCC patients were investigated with Western blot. Chi-squared test, Kaplan-Meier analysis with log-rank test, and Cox proportional hazard trend analyses were performed to assess the prognostic values of the markers. RESULTS: Histone 3 lysine 4 trimethylation (H3K4me3), histone 3 lysine 9 trimethylation (H3K9me3), and histone 4 lysine 20 trimethylation (H4K20me3), but not histone 3 lysine 36 trimethylation (H3K36me3), showed stronger immunostaining signals in tumor tissues than in the corresponding adjacent non-neoplastic mucosa tissues. The expression patterns of H3K36me3, H3K9me3, and H4K20me3 correlated with tumor infiltrating depth, lymph node involvement, and pTNM stage. Low-scoring H3K9me3 and H4K20me3 predicted better prognosis, while H3K36me3 manifested the opposite trend. Poor prognosis occurred in ESCC patients with expression patterns of high levels of H3K9me3, high levels of H4K20me3, and low levels of H3K36me3 expression. CONCLUSIONS: H3K9me3, H4K20me3, and H3K36me3 showed a close relationship with clinical features and were considered independent risk factors for survival of ESCC patients. The combination of H3K9me3, H4K20me3, and H3K36me3 expression, rather than the expression of a single histone marker, is believed to further enhance evaluations of ESCC prognosis and management.


Assuntos
Biomarcadores Tumorais/análise , Metilação de DNA , Epigênese Genética , Neoplasias Esofágicas/química , Carcinoma de Células Escamosas do Esôfago/química , Histonas/análise , Processamento de Proteína Pós-Traducional , Adulto , Idoso , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/patologia , Carcinoma de Células Escamosas do Esôfago/genética , Carcinoma de Células Escamosas do Esôfago/patologia , Feminino , Humanos , Imuno-Histoquímica , Lisina , Masculino , Metilação , Pessoa de Meia-Idade , Valor Preditivo dos Testes , Prognóstico , Análise Serial de Tecidos
2.
Cell Physiol Biochem ; 46(6): 2215-2231, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29734189

RESUMO

BACKGROUND/AIMS: PITX1 has been identified as a potential tumor-suppressor gene in several malignant tumors. The molecular mechanism underlying PITX1, particularly its function as a transcription factor regulating gene expression during tumorigenesis, is still poorly understood. METHODS: The expression level and location of PITX1 were determined by quantitative reverse transcription PCR (qRT-PCR) and immunohistochemical staining in gastric cancer (GC). The effect of PITX1 on the GC cell proliferation and tumorigenesis was analyzed in vitro and in vivo. To explore how PITX1 suppresses cell proliferation, we used PITX1-ChIP-sequencing to measure genome-wide binding sites of PITX1 and assessed global function associations based on its putative target genes. ChIP-PCR, electrophoretic mobility shift assay, and promoter reporter assays examined whether PITX1 bound to PDCD5 and regulated its expression. The function of PDCD5 in GC cell apoptosis was further examined in vitro and in vivo. The relationship between the PITX1 protein level and GC patient prognosis was evaluated by the Kaplan-Meier estimator. Meanwhile, the expression level of miR-19a-3p, which is related to PITX1, was also detected by luciferase reporter assay, qRT-PCR, and western blotting. RESULTS: The expression level of PITX1 was decreased in GC tissues and cell lines. Elevated PITX1 expression significantly suppressed the cell proliferation of GC cells and tumorigenesis in vitro and in vivo. PITX1 knockdown blocked its inhibition of GC cell proliferation. PITX1 bound to whole genome-wide sites, with these targets enriched on genes with functions mainly related to cell growth and apoptosis. PITX1 bound to PDCD5, an apoptosis-related gene, during tumorigenesis, and cis-regulated PDCD5 expression. Increased PDCD5 expression in GC cells not only induced GC cell apoptosis, but also suppressed GC cell growth in vitro and in vivo. Moreover, PITX1 expression was regulated by miR-19a-3p. More importantly, a decreased level of PITX1 protein was correlated with poor GC patient prognosis. CONCLUSION: Decreased expression of PITX1 predicts shorter overall survival in GC patients. As a transcriptional activator, PITX1 regulates apoptosis-related genes, including PDCD5, during gastric carcinogenesis. These data indicate PDCD5 to be a novel and feasible therapeutic target for GC.


Assuntos
Proteínas Reguladoras de Apoptose/genética , Regulação para Baixo , Regulação Neoplásica da Expressão Gênica , MicroRNAs/genética , Proteínas de Neoplasias/genética , Fatores de Transcrição Box Pareados/genética , Neoplasias Gástricas/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Apoptose , Linhagem Celular Tumoral , Proliferação de Células , Feminino , Mucosa Gástrica/metabolismo , Humanos , Masculino , Camundongos Nus , Pessoa de Meia-Idade , Prognóstico , Estômago/patologia , Neoplasias Gástricas/diagnóstico , Neoplasias Gástricas/patologia , Ativação Transcricional
3.
Tumour Biol ; 37(1): 877-85, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26254611

RESUMO

Aberrant DNA methylation of CpG site is among the earliest and most frequent alterations in developmental process and diseases including cancer. To elucidate the functional preferred site of DNMTs, we analyzed the feature of distinct methylated sequences and established the defined relationship between DNMTs and preference genomic DNA sequences. Small interfering RNA (siRNA) construct of DNTM1, DNMT3A, and DNMT3B was transfected into the human hepatocellular carcinoma cell line SMMC-7721, respectively. Distinguishing methylated fragments pool was enriched by SHH method in cells which is knocked down DNMT1, DNMT3A, DNMT3B, separately. The defined binding transcription factors (TFs) containing of 5'CpG islands were obtained with bioinformatics software and website. In SMMC-7721 hepatocellular carcinoma (HCC) cell line, DNMT1, DNMT3A, and DNMT3B were specific suppressed by their corresponding siRNA construct, separately. A 46, 42, 67 distinctive methylated fragments from three different DNMTs were evaluated according to genomic DNA database. Those separated fragments were distributed among genomic DNA regions of all chromosome complements, including coding genes, repeat sequences, and genes with unknown function. The majority of coding genes contain CpG islands in their promoter region. Cluster analysis demonstrated all of preference sequences identified by three DNMTs shares their own conserved sequences. In depleting of different DNMTs cells, 80 % of 103 upregulation genes induced by DNMT1 knock-down contain CpG sites; 76 % of 25 upregulation genes induced by DNMT3A knock-down contain CpG sites; 63 % of 126 upregulation genes induced by DNMT3B knock-down contain CpG sites. Our findings suggested that distinctive DNMTs targeted DNA methylation site to their preference sequences, and this targeting might be associated with diverse roles of DNMTs in tumorigenesis. Meanwhile, the analysis of preference sequences provides an alternative way to find out the individual function of DNMTs.


Assuntos
Carcinoma Hepatocelular/genética , DNA (Citosina-5-)-Metiltransferases/genética , Metilação de DNA , Regulação Neoplásica da Expressão Gênica , Neoplasias Hepáticas/genética , Carcinoma Hepatocelular/metabolismo , Linhagem Celular Tumoral , Análise por Conglomerados , Ilhas de CpG , DNA (Citosina-5-)-Metiltransferase 1 , DNA Metiltransferase 3A , Epigênese Genética , Perfilação da Expressão Gênica , Humanos , Neoplasias Hepáticas/metabolismo , Hibridização de Ácido Nucleico , Regiões Promotoras Genéticas , RNA Interferente Pequeno/metabolismo , Regulação para Cima , DNA Metiltransferase 3B
4.
World J Surg Oncol ; 13: 101, 2015 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-25889078

RESUMO

BACKGROUND: MicroRNAs (miRNAs) play an important role in a tumor-suppressive or oncogenic manner in carcinogenesis. Alteration expression patterns of miRNAs in gastric cancer (GC) are associated with cancer initiation and progression. In the present study, we evaluated miR-29a-3p expression pattern and its function in gastric carcinogenesis. METHODS: The expression of miR-29a-3p in GC tissue samples and cell lines was detected by quantitative real-time PCR (qRT-PCR). After transfected with miR-29a-3p mimics or inhibitor, the cell proliferation, cell migration, and invasion ability were assessed by CCK-8 assay, wound healing assay, and Trans-well assay, respectively. The level of CDK2, CDK4, CDK6, and CyclinD1 were determined by qRT-PCR and Western blot. RESULTS: Compared with the corresponding non-tumor tissues, miR-29a-3p showed a significant down-regulated expression in tumor tissues. In vitro functional assays demonstrated that enforced miR-29a-3p expression inhibited cell proliferation by reducing the expression of CDK2, CDK4, and CDK6. Wound healing and Transwell assays revealed that miR-29a-3p suppressed tumor metastasis in GC. CONCLUSIONS: Our preliminary results suggest that altered expression of miR-29a-3p is involved in gastric cancer process. The present study provides the first insight into the specific role of miR-29a-3p in gastric carcinogenesis.


Assuntos
Movimento Celular , Proliferação de Células , Regulação Neoplásica da Expressão Gênica , MicroRNAs/genética , Neoplasias Gástricas/genética , Neoplasias Gástricas/patologia , Apoptose , Western Blotting , Regulação para Baixo , Humanos , Invasividade Neoplásica , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Tumorais Cultivadas
5.
Drug Deliv Transl Res ; 11(5): 1969-1982, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-33006741

RESUMO

Balancing the antitumor activity and systemic toxicity of tripterine still faces a big challenge due to the narrow therapeutic window. To address this issue, we report a microemulsion system based on tripterine, brucea oil, and glycyrrhizin, and dual modified with both transferrin and cell-penetrating peptide SA-R6H4 (Tf/SA-R6H4-TBG-MEs) for combinational and tumor-targeted cancer therapy. Such a microemulsion exhibited a spherical shape with a size of ~50 nm and a mildly-negative charge. The half-maximal inhibitory concentration (IC50) of Tf/SA-R6H4-TBG-MEs against ovarian cancer SKOV3 cells was 0.27 ± 0.43 µg tripterine/mL, which was 5.85 times lower than that of free tripterine. The cellular uptake of tripterine after treatment with Tf/SA-R6H4-TBG-MEs was 1.56 times higher than that of TBG-MEs (non-modified microemulsion). In pharmacokinetics studies, the area under the curve of Tf/SA-R6H4-TBG-MEs increased by 1.97 times compared with that of the physical mixture group. The tumoral accumulation of tripterine was significantly improved in Tf/SA-R6H4-TBG-MEs group than TBG-MEs-treated group. In antitumor efficacy in vivo, Tf/SA-R6H4-TBG-MEs exhibited the strongest inhibition of tumor growth and the longest survival period among all the groups, which is associated with the rational combination, microemulsion system, and dual modification with tumor-targeted ligands. Importantly, Tf/SA-R6H4-TBG-MEs significantly reduced the toxicity of tripterine against the liver and kidney. Our design provides a new approach for efficient and safe ovarian cancer therapy based on a multicomponent combination.


Assuntos
Peptídeos Penetradores de Células , Coix , Neoplasias Ovarianas , Linhagem Celular Tumoral , Emulsões , Humanos , Neoplasias Ovarianas/tratamento farmacológico , Transferrina
6.
BMC Med ; 8: 12, 2010 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-20128888

RESUMO

BACKGROUND: DNA-methyltransferase (DNMT)-3A plays an important role in the development of embryogenesis and the generation of aberrant methylation in carcinogenesis. The aim of this study was to investigate the role of a DNMT3A promoter genetic variant on its transcriptional activity and to evaluate the association between DNMT3A gene polymorphism and the susceptibility to gastric cancer (GC) and oesophagus carcinoma (EC) in the Chinese population. METHODS: We selected one of the single nucleotide polymorphisms (SNPs) -448A>G in the DNMT3A promoter region and evaluated its effect on activity using a luciferase assay. -448A>G polymorphisms of DNMT3A were determined by polymerase chain reaction/restriction fragment length polymorphism and confirmed by sequencing. The distribution of -448A>G polymorphisms was detected in 208 GC patients and 346 healthy controls matched for age and gender. The distribution of -448A>G polymorphisms was also detected in 96 EC patients and matched 241 healthy controls. The association of -448A>G polymorphisms of DNMT3A and the risk of GC and EC was evaluated by stratified analysis according to the patient's age and gender. RESULTS: In a promoter assay, carriage of the -448 A allele showed a significantly higher promoter activity (> two fold) compared with the -448G allele (P < 0.001). The allele frequency of -448A among GC patients and controls was 32.9% versus 19.9%, respectively. Overall, we found that, compared with GG carriers, the DNMT3A -448AA homozygotes has a > six fold increased risk of GC. Stratification analysis showed that AA homozygotes have a more profound risk in the subgroups of individuals at the age range G polymorphism is a novel functional SNP and contributes to its genetic susceptibility to GC. -448A>G can be used as a stratification marker to predict an individual's susceptibility to GC, especially in the subgroups of individuals at the age range G in EC can not be used as a prediction marker in order to evaluate an individual's susceptibility to EC.


Assuntos
DNA (Citosina-5-)-Metiltransferases/genética , Neoplasias Esofágicas/genética , Neoplasias Gástricas/genética , Estudos de Casos e Controles , DNA Metiltransferase 3A , Neoplasias Esofágicas/enzimologia , Feminino , Frequência do Gene , Predisposição Genética para Doença , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Reação em Cadeia da Polimerase , Polimorfismo de Nucleotídeo Único , Regiões Promotoras Genéticas , Análise de Sequência de DNA , Neoplasias Gástricas/enzimologia
7.
J Biomed Biotechnol ; 2010: 737535, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20467490

RESUMO

Promoter hypermethylation mediated by DNA methyltransferases (DNMTs) is the main reason for epigenetic inactivation of tumor suppressor genes (TSGs). Previous studies showed that DNMT1 and DNMT3B play an important role in CpG island methylation in tumorigenesis. Little is known about the role of DNMT3A in this process, especially in hepatocellular carcinoma (HCC). In the present study, increased DNMT3A expression in 3 out of 6 HCC cell lines and 16/25 (64%) HCC tissues implied that DNMT3A is involved in hepatocellular carcinogenesis. Depletion of DNMT3A in HCC cell line SMMC-7721 inhibited cell proliferation and decreased the colony formation (about 65%). Microarray data revealed that 153 genes were upregulated in DNMT3A knockdown cells and that almost 71% (109/153) of them contain CpG islands in their 5' region. 13 of them including PTEN, a crucial tumor suppressor gene in HCC, are genes involved in cell cycle and cell proliferation. Demethylation of PTEN promoter was observed in DNMT3A-depleted cells implying that DNMT3A silenced PTEN via DNA methylation. These results provide insights into the mechanisms of DNMT3A to regulate TSGs by an epigenetic approach in HCC.


Assuntos
Carcinoma Hepatocelular/metabolismo , Proliferação de Células , DNA (Citosina-5-)-Metiltransferases/genética , PTEN Fosfo-Hidrolase/metabolismo , Western Blotting , Ilhas de CpG , DNA (Citosina-5-)-Metiltransferases/metabolismo , Metilação de DNA , DNA Metiltransferase 3A , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Histocitoquímica , Humanos , Fígado/metabolismo , Fígado/patologia , PTEN Fosfo-Hidrolase/genética , Regiões Promotoras Genéticas , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Tumorais Cultivadas
8.
Exp Ther Med ; 17(5): 4046-4054, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-31007741

RESUMO

Resistance to chemotherapeutic drugs leads to a poor prognosis in gastric cancer (GC). The present study aimed to assess the association between pituitary homeobox paired homeodomain transcription 1 (PITX1) expression and the sensitivity of GC cells to the chemotherapeutic drugs 5-fluorouracil (5-FU) and cisplatin (CDDP). In the present study, the gastric cancer cell lines GES-1, AGS, BGC-823, MCG-803 and SGC-7901 were used. The expression of PITX1 was determined via reverse transcription-quantitative polymerase chain reaction in GC cell lines. AGS and BGC-823 cells, which exhibit a decreased PITX1 expression, were transfected with a PITX1 cDNA construct and its control vector. MCG-803 and SGC-7901 cells, which exhibit an increased PITX1 expression, were transfected with siRNA against PITX1 and its control scramble sequence. A Cell Counting kit-8 assay was performed to determine the impact of PITX1 expression on the sensitivity of GC cells to 5-FU and CDDP. The Cancer Genome Atlas database was used to analyze the expression of PITX1 with GC prognosis in the Asian population and to assess the potential mechanism of PITX1 in 5-FU and CDDP resistance. The results revealed that the overexpression of PIXT1 increased the sensitivity of GC cells to 5-FU/CDDP. The combination of 5-FU/CDDP and PITX1 overexpression also reduced the proliferation of GC cells. Additionally, PIXT1 knockdown decreased the sensitivity of GC cells to 5-FU/CDDP. TCGA data revealed that a lower expression of PITX1 is exhibited in Asian GC patients than in normal individuals. GC patients with a lower expression of PITX1 had a poor prognosis. The expression of PITX1 affected the sensitivity of GC cells to 5-FU/CDDP, indicating that PITX1 may increase the efficacy of treatment in GC patients.

9.
World J Gastroenterol ; 14(14): 2230-4, 2008 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-18407600

RESUMO

AIM: To investigate the relationship between 579 G>T polymorphisms in the DNMT3B gene, which is involved in de novo methylation and associated with the risk of esophagus cancer (EC) in Chinese. METHODS: DNMT3B 579 G>T genotypes were determined by PCR-RFLP in 194 EC patients and 210 healthy controls matched for age and sex, who did not receive radiotherapy or chemotherapy for newly diagnosed and histopathologically confirmed EC. RESULTS: In control subjects, the frequency of T/T and G/T genotypes, and T and G alleles was 81.4%, 18.1%, 90.05% and 9.55%, respectively. The distribution of genotypes and allelotypes in the EC patients was not significantly different from that in the controls. When stratified by sex and age, there was still no significant association between the risks of EC and GT and GG genotypes. This study also showed a distinct difference in the distribution of DNMT3B and single nucleotide polymorphism (SNP) between Chinese and Koreans. CONCLUSION: DNMT3B 579 G>T polymorphism may not be a stratification marker to predict the susceptibility to EC, at least in Chinese. DNMT3B promoter SNP is diverse in ethnic populations.


Assuntos
Carcinoma/diagnóstico , Carcinoma/genética , DNA (Citosina-5-)-Metiltransferases/genética , DNA (Citosina-5-)-Metiltransferases/fisiologia , Neoplasias Esofágicas/diagnóstico , Neoplasias Esofágicas/genética , Genótipo , Polimorfismo Genético , Alelos , Sequência de Bases , Estudos de Casos e Controles , China , Frequência do Gene , Humanos , Modelos Biológicos , Dados de Sequência Molecular , Polimorfismo de Fragmento de Restrição , Análise de Sequência de DNA , DNA Metiltransferase 3B
10.
Oncol Rep ; 40(2): 923-931, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-29845218

RESUMO

Cancer cell invasion and metastasis are the leading causes of the high mortality rates in patients with malignant tumors. There is accumulating evidence to indicate that dysregulated long non­coding RNAs (lncRNAs) may be involved in the progression of tumor invasion and metastasis. However, the regulatory mechanisms of the aberrant expression of lncRNAs remain largely unknown, although the roles of lncRNAs as drivers of tumor suppressive and oncogenic functions have appeared in prevalent cancer types in recent years. In the present study, we identified that the transcription factor, activating enhancer­binding protein 4 (TFAP4), acts as a key modulator of translation regulatory long non­coding RNA 1(TRERNA1), which has been proven to promote the invasion and metastasis of gastric cancer (GC) cells. We revealed that TRERNA1 was upregulated in gastric carcinogenesis and promoted cell migration and invasion in GC. Using bioinformatics analysis, we observed that there were several potential binding sites of TFAP4 in the promoter region of TRERNA1. The knockdown of TFAP4 significantly reduced the expression level of TRERNA1, whereas the ectopic expression of TFAP4 significantly increased the expression level of TRERNA1 in GC cell lines. Dual luciferase reporter assay combined with chromatin immunoprecipitation (ChIP) revealed that TFAP4 specifically regulated the transcriptional activity of TRERNA1 by binding to the E­box motifs in the TRERNA1 promoter. In addition, there was a positive correlation between the TFAP4 and TRERNA1 expression level in clinical GC cases, which also indicated that TFAP4 can directly modulate the expression of TRERNA1. In the present study, we provide a novel potential therapeutic target and strategy for GC.


Assuntos
Fator 4 Ativador da Transcrição/genética , Movimento Celular/genética , Invasividade Neoplásica/genética , RNA Longo não Codificante/genética , Neoplasias Gástricas/genética , Neoplasias Gástricas/patologia , Linhagem Celular Tumoral , Progressão da Doença , Feminino , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Masculino , Pessoa de Meia-Idade , Invasividade Neoplásica/patologia , Regiões Promotoras Genéticas/genética , Estômago/patologia , Regulação para Cima/genética
11.
Oncogene ; 37(32): 4358-4371, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29717263

RESUMO

DNA methyltransferase 3A (DNMT3A) has been recognised as a key element of epigenetic regulation in normal development, and the aberrant regulation of DNMT3A is implicated in multiple types of cancers, especially haematological malignancies. However, its clinical significance and detailed functional role in solid tumours remain unknown, although abnormal expression has gained widespread attention in these cancers. Here, we show that DNMT3A isoform b (DNMT3Ab), a member of the DNMT3A isoform family, is critical for directing epithelial-mesenchymal transition (EMT)-associated metastasis in gastric cancer (GC). DNMT3Ab is positively linked to tumour-node-metastasis (TNM) stage, lymph node metastasis and poor prognosis in GC patients. Overexpression of DNMT3Ab promotes GC cell migration and invasion as well as EMT through repression of E-cadherin. Meanwhile, DNMT3Ab promotes lung metastasis of GC in vivo. Mechanistic studies indicate that DNMT3Ab mediates the epigenetic inaction of the E-cadherin gene via DNA hypermethylation and histone modifications of H3K9me2 and H3K27me3. Depletion of DNMT3Ab effectively restores the expression of E-cadherin and reverses TGF-ß-induced EMT by reducing DNA methylation, H3K9me2 and H3K27me3 levels at the E-cadherin promoter. Importantly, DNMT3Ab cooperated with H3K9me2 and H3K27me3 contributes to the transcriptional regulation of E-cadherin in a Snail-dependent manner. Further, gene expression profiling analysis indicates that multiple metastasis-associated genes and oncogenic signalling pathways are regulated in response to DNMT3Ab overexpression. These results identify DNMT3Ab as a crucial regulator of metastasis-related genes in GC. Targeting the DNMT3Ab/Snail/E-cadherin axis may provide a promising therapeutic strategy in the treatment of metastatic GC with high DNMT3Ab expression.


Assuntos
Caderinas/genética , DNA (Citosina-5-)-Metiltransferases/genética , Metilação de DNA/genética , Transição Epitelial-Mesenquimal/genética , Isoformas de Proteínas/genética , Neoplasias Gástricas/genética , Movimento Celular/genética , DNA Metiltransferase 3A , Epigênese Genética/genética , Feminino , Regulação Neoplásica da Expressão Gênica/genética , Histonas/genética , Humanos , Metástase Linfática/genética , Masculino , Pessoa de Meia-Idade , Regiões Promotoras Genéticas/genética , Transdução de Sinais/genética , Fatores de Transcrição da Família Snail/genética , Neoplasias Gástricas/patologia , Transcrição Gênica/genética , Fator de Crescimento Transformador beta/genética
12.
Eur J Gastroenterol Hepatol ; 19(11): 952-61, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18049164

RESUMO

BACKGROUND: Hepatocellular carcinoma (HCC) is one of the most common causes of cancer-related mortality in the world; however, the molecular mechanisms leading to hepatocyte transformation, especially in epigenetic mechanisms (such as DNA methylation) are still poorly understood. DNA methyltransferase 1 (DNMT1) is the predominant maintenance methyltransferase gene required to maintain DNA methylation patterns in mammalian cells. AIM AND METHODS: To explore the role of DNMT1 in the regulation of expression of tumor-related genes in human HCC cells via DNA methylation of the regulatory CpG islands, we stably transfected expression constructs containing small interfering RNA (siRNA) of DNMT1 into the human HCC cell line, SMMC-7721. RESULTS: RNA interference knocked down specific DNMT1 protein expression, resulting in the demethylated promoter of CDH1 and the reexpression of CDH1 in 7721-pMT1. By contrast, promoter methylation and lack of gene expression were maintained when the cell lines had control constructs. Knock down of DNMT1 expression by siRNA induced the promoter of CDH1 demethylation and upregulated CDH1 transcription. High-density oligonucleotide gene expression microarrays were used to examine the effects of DNMT1 knock down on human HCC cells (SMMC-7721); these showed that a number of genes were induced in the DNMT1 knock down cell lines, including some important tumor-related genes such as PDCD4, DCN and PTGES except CDH1. Only approximately 78% of the induced genes have CpG islands within their 5' regions, suggesting that certain genes activated by DNMT1 siRNA might not have resulted from the direct inhibition of promoter methylation. CONCLUSION: In hepatocellular carcinoma, DNMT1 is necessary to maintain the methylation of CpG islands in certain tumor-related genes.


Assuntos
Caderinas/genética , Carcinoma Hepatocelular/genética , Ilhas de CpG , DNA (Citosina-5-)-Metiltransferases/genética , Regulação Neoplásica da Expressão Gênica , Neoplasias Hepáticas/genética , Antígenos CD , Proteínas Reguladoras de Apoptose/genética , Western Blotting/métodos , Carcinoma Hepatocelular/metabolismo , Linhagem Celular Tumoral , DNA (Citosina-5-)-Metiltransferase 1 , Metilação de DNA , DNA Complementar/análise , Decorina , Proteínas da Matriz Extracelular/genética , Perfilação da Expressão Gênica/métodos , Engenharia Genética/métodos , Humanos , Oxirredutases Intramoleculares/genética , Neoplasias Hepáticas/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Prostaglandina-E Sintases , Proteoglicanas/genética , Interferência de RNA , RNA Mensageiro/análise , RNA Interferente Pequeno/farmacologia , Proteínas de Ligação a RNA/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Transfecção/métodos
13.
Chin Med J (Engl) ; 120(18): 1626-31, 2007 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-17908484

RESUMO

BACKGROUND: Aberrant DNA methylation plays a key role in human carcinogenesis. 5-aza-2'-deoxycytidine inhibits DNA methylation and induces the expression of genes putatively silenced by promoter methylation in vitro. There are few studies of the biological and clinical significance of 5-aza-2'-deoxycytidine in human hepatocellular carcinoma. This study explored the mechanism of 5-aza-2'-deoxycytidine targeting transcriptional repressor complexes affecting global gene expression in hepatocellular carcinoma cell line. METHODS: High density oligonucleotide gene expression microarrays were used to examine the effects of 5-aza-2'-deoxycytidine treatments on human hepatocellular carcinoma cell line SMMC-7721. The 5' ends of the genes upregulated or downregulated in this manner were compared with BLAST database to determine whether they might have promoter CpG islands. Flow cytometry was used to detect stages of the cell cycle and apoptosis of SMMC-7721 after being treated with 5-aza-2'-deoxycytidine. RESULTS: Data obtained 3 days after 4 days of treatment with 5-aza-2'-deoxycytidine showed that more genes were induced in tumorigenic cells including genes that function in cell proliferation, differentiation, regulation of transcription, and cytokine signalling. Approximately 30% of induced genes did not have CpG islands within their 5' regions, suggesting that some genes activated by 5-aza-2'-deoxycytidine may not result from the direct inhibition of promoter methylation. This phenomenon may contribute to a number of upregulated genes involving regulation of transcription in the treated cell. Results showed that 100 micromol/L 5-aza-2'-deoxycytidine blocked cell cycle at S/G2-M phase increasing rate of apoptosis. Notably, we found differential expression of molecular action in the methylation although DNA methyltransferases did not show significant difference in the treated cell line. CONCLUSION: 5-aza-2'-deoxycytidine could restore some silenced genes expression independently of DNA methylation inhibition and expression of DNA methyltransferases.


Assuntos
Antimetabólitos Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Azacitidina/análogos & derivados , Carcinoma Hepatocelular/tratamento farmacológico , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Neoplasias Hepáticas/tratamento farmacológico , Azacitidina/farmacologia , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patologia , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Ilhas de CpG , Metilases de Modificação do DNA/genética , Decitabina , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patologia , Ativação Transcricional
14.
Oncol Rep ; 37(5): 2811-2818, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28339081

RESUMO

Hepatitis B virus (HBV) is mainly suspected to promote hepatocellular carcinoma (HCC) development by epigenetic alteration. The HBV X protein (HBx) plays a key role in the molecular pathogenesis of HBV-related HCC. However, the mechanism of HBx-mediated hepatocarcinogenesis remains to be elucidated. RIZ1 gene, a candidate HCC suppressor gene, is frequently found to be hypermethylated and downregulated in HCC. In the present study, we show that the expression of RIZ1 was downregulated in 65% HCC tissues. Decreased expression of RIZ1 was restored by 5'-Aza in MHCC-97H HCC cell lines. HBx recombinant transfection increased DNMT1 expression level and suppressed RIZ1 expression. Moreover, knockdown of DNMT1 by siRNA restored RIZ1 expression in HCC cell SMMC-7721 and reduced methylated CpG sites of RIZ1. ChIP results showed that DNMT1 protein could bind to RIZ1 promoter, and this interaction was further enhanced with the transfected HBX recombinant. Moreover, miR-152 was decreased and involved in upregulation of DNMT1 in HBx transfected cells, at least partly, contributed to the epigenetic inactivation of RIZ1. Taken together, our data found that HBx repressed RIZ1 expression via DNMT1, which offered a new mechanism of RIZ1 inactivation in HCC, except for the widely known DNA methylation. These results enriched the epigenetic mechanism by which HBx contributes to pathogenesis of HBV-HCC.


Assuntos
Carcinoma Hepatocelular/virologia , DNA (Citosina-5-)-Metiltransferases/metabolismo , Proteínas de Ligação a DNA/genética , Histona-Lisina N-Metiltransferase/genética , Neoplasias Hepáticas/virologia , MicroRNAs/genética , Proteínas Nucleares/genética , Transativadores/metabolismo , Fatores de Transcrição/genética , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Linhagem Celular Tumoral , DNA (Citosina-5-)-Metiltransferase 1 , DNA (Citosina-5-)-Metiltransferases/genética , Metilação de DNA , Proteínas de Ligação a DNA/metabolismo , Regulação para Baixo , Epigênese Genética , Técnicas de Silenciamento de Genes , Células Hep G2 , Vírus da Hepatite B/genética , Vírus da Hepatite B/metabolismo , Histona-Lisina N-Metiltransferase/metabolismo , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Recombinantes/metabolismo , Transativadores/genética , Fatores de Transcrição/metabolismo , Proteínas Virais Reguladoras e Acessórias
15.
Sci Rep ; 6: 23521, 2016 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-27009634

RESUMO

It is well accepted that HBx plays the major role in hepatocarcinogenesis associated with hepatitis B virus (HBV) infections. However, little was known about its role in regulating long noncoding RNAs (lncRNAs), a large group of transcripts regulating a variety of biological processes including carcinogenesis in mammalian cells. Here we report that HBx upregulates UCA1 genes and downregulates p27 genes in hepatic LO2 cells. Further studies show that the upregulated UCA1 promotes cell growth by facilitating G1/S transition through CDK2 in both hepatic and hepatoma cells. Knock down of UCA1 in HBx-expressing hepatic and hepatoma cells resulted in markedly increased apoptotic cells by elevating the cleaved caspase-3 and caspase-8. More importantly, UCA1 is found to be physically associated with enhancer of zeste homolog 2 (EZH2), which suppresses p27Kip1 through histone methylation (H3K27me3) on p27Kip1 promoter. We also show that knockdown of UCA1 in hepatoma cells inhibits tumorigenesis in nude mice. In a clinic study, UCA1 is found to be frequently up-regulated in HBx positive group tissues in comparison with the HBx negative group, and exhibits an inverse correlation between UCA1 and p27Kip1 levels. Our findings demonstrate an important mechanism of hepatocarcinogenesis through the signaling of HBx-UCA1/EZH2-p27Kip1 axis, and a potential target of HCC.


Assuntos
Carcinogênese/genética , Transformação Celular Viral/genética , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Neoplasias Hepáticas/genética , RNA Longo não Codificante/genética , Transativadores/genética , Animais , Proliferação de Células , Quinase 2 Dependente de Ciclina/genética , Inibidor de Quinase Dependente de Ciclina p27/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Hepáticas/virologia , Proteínas de Membrana , Camundongos , Camundongos Nus , Transplante de Neoplasias , Serina Endopeptidases , Transdução de Sinais , Transfecção , Proteínas Virais Reguladoras e Acessórias
16.
Yi Chuan Xue Bao ; 32(11): 1115-27, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16318277

RESUMO

Whether DNA methyltransferase 3B (DNMT3B) is deregulated in hepatocellular carcinoma cell lines is still unclear. The expression levels of DNMT3B protein in normal liver cell line, pericacinoma cell line and hepatocellular carcinoma cell lines were compared by both Western blotting and immunocytochemistry. Long-term downregulated DNMT3B in a hepatocellular carcinoma cell line SMMC-7721 was achieved using a RNAi recombinant plasmid. The suppression of DNMT3B induced by RNA interference was confirmed using semi-quantitative RT-PCR and Western blotting. High throughput cDNA microarray was used to analyze the expression profiling of downstream genes of DNMT3B displayed in the treated cell lines and control. In the result,DNMT3B in hepatocellular carcinoma cell lines was expressed at a significantly higher level compared to those in pericacinoma cell line and normal liver cell line. A specific DNMT3B siRNA stably expressed from a plasmid vector effectively suppressed the expression of DNMT3B in SMMC-7721 cell line. By microarray analysis,26 downregulated genes and 115 upregulated genes have been identified in the DNMT3B knockdown cell line,including some important developmental genes and tumor-related genes such as SNCG, NOTCH1, MBD3, WNT11, MAOA and FACL4. The discovery showed DNMT3B was over-expressed in most hepatocellular carcinoma cell lines examined and may be linked to the carcinogenesis of hepatocytes. An array of candidate genes that are involved in the action of DNMT3B have been identified,including those related to development.


Assuntos
DNA (Citosina-5-)-Metiltransferases/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Interferência de RNA/fisiologia , RNA Interferente Pequeno/farmacologia , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , DNA (Citosina-5-)-Metiltransferases/genética , Metilação de DNA , Regulação Neoplásica da Expressão Gênica/fisiologia , Humanos , Análise em Microsséries/métodos , DNA Metiltransferase 3B
17.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 22(2): 142-5, 2005 Apr.
Artigo em Zh | MEDLINE | ID: mdl-15793773

RESUMO

OBJECTIVE: To construct the specific stable expression and high efficiency small interfering RNA(siRNA) expression vector that can block DNMT1 gene function. METHODS: Using vector-based RNA interference technique, the authors constructed a vector to transcribe functional short interfering RNA (RNAi). After transfection by lipofectmine (TM) reagent, the treated cells were selected by G418. The expression levels of RNA and protein of DNMT1 were analyzed by reverse transcription polymerase chain reaction(RT-PCR) and Western blotting. The status of methylation of E-cadherin was analyzed by methylation-specific PCR(MSP). RESULTS: The expression level of endogenous DNMT1 mRNA in transfected SMMC-7721 cell lines with DNMT1 RNAi construct was 43% less than that in control cell 7721-pSU cell lines. The protein level in the former was about 10% less than that in the latter. The efficiency of the siRNA of DNMT1 was found to be higher than 90%. Demethylation of promoter of E-cadherin was obtained due to the inhibition of DNMT1. CONCLUSION: DNMT1 siRNA stable expressing vector was obtained by gene-recombined technology. There was no complete sameness between the levels of protein and RNA in gene silenced cell lines. The efficiency of the siRNA should be confirmed by Western-blotting.


Assuntos
DNA (Citosina-5-)-Metiltransferases/genética , Vetores Genéticos/genética , Interferência de RNA , RNA Interferente Pequeno/genética , Western Blotting , Linhagem Celular Tumoral , DNA (Citosina-5-)-Metiltransferase 1 , DNA (Citosina-5-)-Metiltransferases/metabolismo , Metilação de DNA , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Microscopia de Fluorescência , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção
18.
PLoS One ; 10(4): e0123926, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25874772

RESUMO

The de-regulation of the miR-29 family and DNA methyltransferase 3A (DNMT3A) is associated with gastric cancer (GC). While increasing evidence indicates miR-29b/c could regulate DNA methylation by targeting DNMT3A, it is currently unknown if epigenetic silencing of miR-29b/c via promoter hypermethylation in GC is caused by abnormal expression of DNMT3A. Thus, we aimed to evaluate whether cross-talk regulation exists between miR-29b/c and DNMT3A and whether it is associated with a malignant phenotype in GC. First, wound healing and Transwell assays revealed that miR-29b/c suppresses tumor metastasis in GC. A luciferase reporter assay demonstrated that DNMT3A is a direct target of miR-29b/c. We used bisulfite genomic sequencing to analyze the DNA methylation status of miR-29b/c. The percentage of methylated CpGs was significantly decreased in DNMT3A-depleted cells compared to the controls. Furthermore, the involvement of DNMT3A in promoting GC cell migration was associated with the promoter methylation-mediated repression of CDH1. In 50 paired clinical GC tissue specimens, decreased miR-29b/c was significantly correlated with the degree of differentiation and invasion of the cells and was negatively correlated with DNMT3A expression. Together, our preliminary results suggest that the following process may be involved in GC tumorigenesis. miR-29b/c suppresses the downstream gene DNMT3A, and in turn, miR-29b/c is suppressed by DNMT3A in a DNA methylation-dependent manner. The de-regulation of both of miR-29b/c and DNMT3A leads to the epigenetic silencing of CDH1 and contributes to the metastasis phenotype in GC. This finding reveals that DNA methylation-associated silencing of miR-29b/c is critical for GC development and thus may be a therapeutic target.


Assuntos
Caderinas/genética , DNA (Citosina-5-)-Metiltransferases/genética , Regulação Neoplásica da Expressão Gênica , MicroRNAs/genética , Neoplasias Gástricas/genética , Antígenos CD , Bioensaio , Caderinas/metabolismo , Carcinogênese/genética , Carcinogênese/metabolismo , Carcinogênese/patologia , Diferenciação Celular , Linhagem Celular Tumoral , Movimento Celular , Ilhas de CpG , DNA (Citosina-5-)-Metiltransferases/metabolismo , Metilação de DNA , DNA Metiltransferase 3A , Feminino , Inativação Gênica , Genes Reporter , Humanos , Luciferases/genética , Luciferases/metabolismo , Masculino , MicroRNAs/metabolismo , Estadiamento de Neoplasias , Transdução de Sinais , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patologia , Neoplasias Gástricas/cirurgia , Cicatrização
19.
World J Gastroenterol ; 9(7): 1455-9, 2003 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12854140

RESUMO

AIM: To prepare and characterize polyclonal antibodies against aldose reductase-like (ARL-1) protein. METHODS: ARL-1 gene was inserted into the E. coli expression vector pGEX-4T-1(His)(6)C and vector pQE-30. Recombinant ARL-1 proteins named ARL-(His)(6) and ARL-GST were expressed. They were purified by affinity chromatography. Sera from domestic rabbits immunized with ARL-(His) (6) were purified by CNBr-activated sepharose 4B coupled ARL-GST. Polyclonal antibodies were detected by Western blotting. RESULTS: Recombinant proteins of ARL-(His)(6) with molecular weight of 35.7 KD and ARL-GST with molecular weight of 60.8 KD were highly expressed. The expression levels of ARL-GST and ARL-(His)(6) were 15.1 % and 27.7 % among total bacteria proteins, respectively. They were soluble, predominantly in supernatant. After purification by non-denatured way, SDS-PAGE showed one band. In the course of polyclonal antibodies purification, only one elution peak could be seen. Western blotting showed positive signals in the two purified proteins and the bacteria transformed with pGEX-4T-1(His) (6) C-ARL and pQE-30-ARL individually. CONCLUSION: Polyclonal antibodies are purified and highly specific against ARL-1 protein. ARL-GST and ARL-(His) (6) are highly expressed and purified.


Assuntos
Aldeído Redutase/genética , Aldeído Redutase/imunologia , Anticorpos/imunologia , Especificidade de Anticorpos , Aldo-Ceto Redutases , Animais , Western Blotting , Clonagem Molecular , Escherichia coli , Expressão Gênica , Humanos , Plasmídeos , Coelhos , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia
20.
Asian Pac J Cancer Prev ; 14(11): 6261-5, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24377515

RESUMO

BACKGROUND: Recent studies have suggested that expression of the RAS protein activator like-1 gene (RASAL1) is decreased in gastric carcinoma tissues and cell lines, indicated a role in tumorigenesis and development of gastric cancer. Reduced expression of RASAL1 could result in aberrant increase of activity of RAS signaling pathways in cancer cells. However, the exact mechanism which induces down-regulation of the RASAL1 gene remains unclear. This study aimed to determine the methylation status and regulation of RASAL1 in gastric cancer. MATERIALS AND METHODS: Using the methylation-specific polymerase chain reaction (MSP), the methylation status of CpG islands in the RASAL1 promoter in gastric cancers and paired adjacent non-cancerous tissues from 40 patients was assessed and its clinicopathological significance was analyzed. The methylation status of RASAL1 in gastric cancer lines MKN-28, SGC-790l, BGC-823, as well as in normal gastric epithelial cell line GES-l was also determined after treatment with a DNA methyltransferase inhibitor, 5-aza-2'-doexycytidine (5-Aza-CdR). RAS activity (GAS-GTP) was assessed through a pull-down method, while protein levels of ERK1/2, a downstream molecule of RAS signaling pathways, were determined by Western blotting. RESULTS: The frequencies of RASAL1 promoter methylation in gastric cancer and paired adjacent non-cancerous tissues were 70% (28/40) and 30% (12/40) respectively (P<0.05). There were significantly correlations between RASAL1 promoter methylation with tumor differentiation, tumor size, invasive depth and lymph node metastasis in patients with gastric cancer (all P<0.05), but no correlation was found for age or gender. Promoter hypermethylation of the RASAL1 gene was detected in MKN-28, SGC-790l and BGC-823 cancer cells, but not in the normal gastric epithelial cell line GES-1. Elevated expression of the RASAL1 protein, a decreased RAS-GTP and p-ERK1/2 protein were detected in three gastric cancer cell lines after treatment with 5-Aza-CdR. CONCLUSIONS: Aberrant hypermethylation of the RASAL1 gene promoter frequently occurs in gastric cancer tissues and cells. In addition, the demethylating agent 5-Aza-CdR can reverse the hypermethylation of RASAL1 gene and up-regulate the expression of RASAL1 significantly in gastric cancer cells in vivo. Our study suggests that RASAL1 promoter methylation may have a certain relationship with the reduced RASAL1 expression in gastric cancer.


Assuntos
Metilação de DNA , Proteínas Ativadoras de GTPase/genética , Neoplasias Gástricas/genética , Neoplasias Gástricas/patologia , Linhagem Celular , Linhagem Celular Tumoral , Ilhas de CpG/genética , Células Epiteliais/patologia , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Metástase Linfática , Sistema de Sinalização das MAP Quinases/genética , Masculino , Pessoa de Meia-Idade , Regiões Promotoras Genéticas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA