Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Aging (Albany NY) ; 13(2): 1633-1648, 2020 12 28.
Artigo em Inglês | MEDLINE | ID: mdl-33378746

RESUMO

Histone modifications, specifically in the lysine residues of histone H3, have been implicated in lifespan regulation in several model organisms. Our previous studies showed that growth hormone (GH) treatment during early life can dramatically influence lifespan in long-lived Ames dwarf mice. However, the effects of this hormonal intervention on epigenetic modifications have never been examined. In this study, we sought to compare tissue-specific histone H3 lysine methylation and acetylation markers in Ames dwarf and wild type (WT) mice and to determine how these markers are affected by early-life GH intervention. Ames dwarf mice exhibited suppressed H3K4me in both hepatic and brain tissues, while showing elevated H3K27me in the brain. Early-life GH intervention significantly altered the histone H3 markers in those tissues. Furthermore, early GH intervention increased expression of histone H3 acetylation at multiple lysine residues in a tissue-specific manner. This included changes in H3K14ac and H3K18ac in the liver and brain, H3K18ac in visceral adipose tissue and H3K9ac, H3K14ac and H3K27ac in subcutaneous adipose tissue. This study serves as an initial, but important step in elucidating the epigenetic mechanisms by which hormonal signals during early life can influence aging and longevity in mammals.


Assuntos
Encéfalo/efeitos dos fármacos , Nanismo Hipofisário/metabolismo , Epigênese Genética/efeitos dos fármacos , Hormônio do Crescimento/farmacologia , Histonas/efeitos dos fármacos , Fígado/efeitos dos fármacos , Acetilação/efeitos dos fármacos , Animais , Encéfalo/metabolismo , DNA (Citosina-5-)-Metiltransferase 1/efeitos dos fármacos , DNA (Citosina-5-)-Metiltransferase 1/genética , DNA (Citosina-5-)-Metiltransferases/efeitos dos fármacos , DNA (Citosina-5-)-Metiltransferases/genética , DNA Metiltransferase 3A , Modelos Animais de Doenças , Nanismo Hipofisário/genética , Proteína Potenciadora do Homólogo 2 de Zeste , Hormônio do Crescimento/deficiência , Código das Histonas/efeitos dos fármacos , Histonas/metabolismo , Proteínas de Homeodomínio/genética , Terapia de Reposição Hormonal , Gordura Intra-Abdominal/efeitos dos fármacos , Gordura Intra-Abdominal/metabolismo , Fígado/metabolismo , Longevidade/genética , Metilação/efeitos dos fármacos , Camundongos , Gordura Subcutânea/efeitos dos fármacos , Gordura Subcutânea/metabolismo , DNA Metiltransferase 3B
2.
Int J Neuropsychopharmacol ; 23(4): 257-267, 2020 04 23.
Artigo em Inglês | MEDLINE | ID: mdl-32124922

RESUMO

BACKGROUND: Although the action mechanism of antineoplastic agents is different, oxaliplatin, paclitaxel, or bortezomib as first-line antineoplastic drugs can induce painful neuropathy. In rodents, mechanical allodynia is a common phenotype of painful neuropathy for 3 chemotherapeutics. However, whether there is a common molecular involved in the different chemotherapeutics-induced painful peripheral neuropathy remains unclear. METHODS: Mechanical allodynia was tested by von Frey hairs following i.p. injection of vehicle, oxaliplatin, paclitaxel, or bortezomib in Sprague-Dawley rats. Reduced representation bisulfite sequencing and methylated DNA immunoprecipitation were used to detect the change of DNA methylation. Western blot, quantitative polymerase chain reaction, chromatin immunoprecipitation, and immunohistochemistry were employed to explore the molecular mechanisms. RESULTS: In 3 chemotherapeutic models, oxaliplatin, paclitaxel, or bortezomib accordantly upregulated the expression of transient receptor potential cation channel, subfamily C6 (TRPC6) mRNA and protein without affecting the DNA methylation level of TRPC6 gene in DRG. Inhibition of TRPC6 by using TRPC6 siRNA (i.t., 10 consecutive days) relieved mechanical allodynia significantly following application of chemotherapeutics. Furthermore, the downregulated recruitment of DNA methyltransferase 3 beta (DNMT3b) at paired box protein 6 (PAX6) gene led to the hypomethylation of PAX6 gene and increased PAX6 expression. Finally, the increased PAX6 via binding to the TPRC6 promoter contributes to the TRPC6 increase and mechanical allodynia following chemotherapeutics treatment. CONCLUSIONS: The TRPC6 upregulation through DNMT3b-mediated PAX6 gene hypomethylation participated in mechanical allodynia following application of different chemotherapeutic drugs.


Assuntos
Antineoplásicos/farmacologia , DNA (Citosina-5-)-Metiltransferases/efeitos dos fármacos , Metilação de DNA/efeitos dos fármacos , Gânglios Espinais/efeitos dos fármacos , Expressão Gênica/efeitos dos fármacos , Hiperalgesia/induzido quimicamente , Neuralgia/induzido quimicamente , Fator de Transcrição PAX6/efeitos dos fármacos , Canais de Cátion TRPC/efeitos dos fármacos , Animais , Bortezomib/farmacologia , Modelos Animais de Doenças , Hiperalgesia/tratamento farmacológico , Hiperalgesia/etiologia , Masculino , Neuralgia/complicações , Oxaliplatina/farmacologia , Paclitaxel/farmacologia , Ratos , Ratos Sprague-Dawley , Canais de Cátion TRPC/antagonistas & inibidores , Regulação para Cima/efeitos dos fármacos , DNA Metiltransferase 3B
3.
Biochemistry (Mosc) ; 84(1): 62-70, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30927527

RESUMO

Olivomycin A is a highly active antitumor drug that belongs to the family of aureolic acid antibiotics. The antitumor effect of olivomycin A is related to its ability to bind to the DNA minor groove in GC-rich regions as Mg2+-coordinated complexes. Characterization of cellular targets of olivomycin A and its mechanism of action is crucial for the successful application of this antibiotic in clinical practice and development of semi-synthetic derivatives with improved pharmacological properties. Previously, we have shown that minor groove ligands are able to disrupt the key epigenetic process of DNA methylation. In this paper, we have studied the impact of olivomycin A and its improved semi-synthetic analogue N,N-dimethylaminoethylamide of 1'-des-(2,3-dihydroxy-n-butyroyl)-1'-carboxy-olivomycin A (olivamide) on the functioning of de novo DNA methyltransferase Dnmt3a (enzyme that carries out methylation of cytosine residues in the DNA CG-sites in eukaryotic cells) using an in vitro system consisting of the murine Dnmt3a catalytic domain and a 30-mer DNA duplex containing four consecutive GC pairs. We have shown that olivomycin A and olivamide inhibit Dnmt3a with IC50 of 6 ± 1 and 7.1 ± 0.7 µM, respectively. Neither olivomycin A nor olivamide interfered with the formation of the specific enzyme-substrate complex; however, olivomycin A prevented formation of the covalent DNA-Dnmt3a intermediate that is necessary for the methylation reaction to proceed. The inhibitory effects of olivomycin A and olivamide can be explained by the disruption of the enzyme catalytic loop movement through the DNA minor groove (the reaction stage that precedes the covalent bond formation between DNA and the enzyme). The results of this work indicate the epigenetic contribution to the antitumor effect of aureolic acid group antibiotics.


Assuntos
Antibióticos Antineoplásicos/farmacologia , DNA (Citosina-5-)-Metiltransferases/efeitos dos fármacos , Animais , Metilação de DNA/efeitos dos fármacos , DNA Metiltransferase 3A , Camundongos , Olivomicina/química , Olivomicina/farmacologia , Ligação Proteica/efeitos dos fármacos
4.
Br J Haematol ; 186(1): 91-100, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30891745

RESUMO

Sickle cell disease (SCD) affects over 2 million people worldwide with high morbidity and mortality in underdeveloped countries. Therapeutic interventions aimed at reactivating fetal haemoglobin (HbF) is an effective approach for improving survival and ameliorating the clinical severity of SCD. A class of agents that inhibit DNA methyltransferase (DNMT) activity show promise as HbF inducers because off-target effects are not observed at low concentrations. However, these compounds are rapidly degraded by cytidine deaminase when taken by oral administration, creating a critical barrier to clinical development for SCD. We previously demonstrated that microRNA29B (MIR29B) inhibits de novo DNMT synthesis, therefore, the goal of our study was to determine if MIR29 mediates HbF induction. Overexpression of MIR29B in human KU812 cells and primary erythroid progenitors significantly increased the percentage of HbF positive cells, while decreasing the expression of DNMT3A and the HBG repressor MYB. Furthermore, HBG promoter methylation levels decreased significantly following MIR29B overexpression in human erythroid progenitors. We subsequently, observed higher MIR29B expression in SCD patients with higher HbF levels compared to those with low HbF. Our findings provide evidence for the ability of MIR29B to induce HbF and supports further investigation to expand treatment options for SCD.


Assuntos
Anemia Falciforme/genética , Epigênese Genética/efeitos dos fármacos , Hemoglobina Fetal/genética , MicroRNAs/fisiologia , Ativação Transcricional/efeitos dos fármacos , gama-Globinas/genética , Linhagem Celular , Células Cultivadas , DNA (Citosina-5-)-Metiltransferases/biossíntese , DNA (Citosina-5-)-Metiltransferases/efeitos dos fármacos , DNA Metiltransferase 3A , Metilases de Modificação do DNA/biossíntese , Metilases de Modificação do DNA/efeitos dos fármacos , Células Precursoras Eritroides/metabolismo , Hemoglobina Fetal/metabolismo , Humanos , MicroRNAs/antagonistas & inibidores
5.
Eur J Neurosci ; 50(2): 1843-1870, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30585666

RESUMO

Evidence in humans and rodents suggests that perinatal exposure to selective serotonin reuptake inhibitor (SSRI) antidepressants can have serious long-term consequences in offspring exposed in utero or infancy via breast milk. In spite of this, there is limited knowledge of how perinatal SSRI exposure impacts brain development and adult behaviour. Children exposed to SSRIs in utero exhibit increased internalizing behaviour and abnormal social behaviour between the ages of 3 and 6, and increased risk of depression in adolescence; however, the neurobiological changes underlying this behaviour are poorly understood. In rodents, perinatal SSRI exposure perturbs hippocampal gene expression and alters adult emotional behaviour (including increased depression-like behaviour). The present study demonstrates that perinatal exposure to the SSRI paroxetine leads to DNA hypomethylation and reduces DNA methyltransferase 3a (Dnmt3a) mRNA expression in the hippocampus during the second and third weeks of life. Next-generation sequencing identified numerous differentially methylated genomic regions, including altered methylation and transcription of several dendritogenesis-related genes. We then tested the hypothesis that transiently decreasing Dnmt3a expression in the early postnatal hippocampus would mimic the behavioural effects of perinatal SSRI exposure. We found that siRNA-mediated knockdown of Dnmt3a in the dentate gyrus during the second to third week of life produced greater depression-like behaviour in adult female (but not male) offspring, akin to the behavioural consequences of perinatal SSRI exposure. Overall, these data suggest that perinatal SSRI exposure may increase depression-like behaviours, at least in part, through reduced Dnmt3a expression in the developing hippocampus.


Assuntos
Antidepressivos/efeitos adversos , Comportamento Animal/efeitos dos fármacos , DNA (Citosina-5-)-Metiltransferases/efeitos dos fármacos , Giro Denteado , Depressão/induzido quimicamente , Expressão Gênica/efeitos dos fármacos , Paroxetina/efeitos adversos , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Inibidores Seletivos de Recaptação de Serotonina/efeitos adversos , Animais , Animais Recém-Nascidos , DNA Metiltransferase 3A , Giro Denteado/efeitos dos fármacos , Giro Denteado/crescimento & desenvolvimento , Giro Denteado/metabolismo , Modelos Animais de Doenças , Feminino , Masculino , Gravidez , RNA Interferente Pequeno , Ratos , Ratos Sprague-Dawley , Fatores Sexuais
6.
Psychiatry Clin Neurosci ; 72(4): 255-265, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28990703

RESUMO

AIM: Early life stress can induce epigenetic changes through genetic and environmental interactions and is a risk factor for depression. Brain-derived neurotrophic factor (BDNF) has been implicated in the pathophysiology of depression and antidepressant drug action. We investigated epigenetic changes at the BDNF exon I promoter in the hippocampus of adult rats subjected to maternal separation (MS) during early life and treated with an antidepressant drug as adults. METHODS: Rat pups were subjected to MS from postnatal day 1 to 21 and received chronic escitalopram (ESC) as adults. We assessed the effects of MS and ESC on BDNF exon I and DNA methyltransferases (DNMT) mRNA levels (quantitative reverse-transcription polymerase chain reaction), acetylated histone H3, and MeCP2 binding to the BDNF promoter I (chromatin immunoprecipitation followed by real-time polymerase chain reaction), and BDNF protein levels (enzyme-linked immunosorbent assay). RESULTS: The levels of BDNF protein, exon I mRNA, histone H3 acetylation, and DNMT1 and DNMT3a mRNA were altered in the MS group compared with the control group. Significant decreases were observed in the BDNF protein, exon I mRNA, and histone H3 acetylation levels and there were significant increases in DNMT1 and DNMT3a mRNA levels. The comparison between the MS + ESC and MS groups revealed significant increases in BDNF protein, exon I mRNA, and histone H3 acetylation levels and significant decreases in MeCP2 and DNMT1 and DNMT3a mRNA levels. CONCLUSION: These findings indicate that MS induced epigenetic changes at the BDNF exon I promoter and these changes were prevented by antidepressant drug treatment during adulthood.


Assuntos
Antidepressivos de Segunda Geração/farmacologia , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Citalopram/farmacologia , DNA (Citosina-5-)-Metiltransferase 1/metabolismo , DNA (Citosina-5-)-Metiltransferases/metabolismo , Epigênese Genética/fisiologia , Hipocampo/metabolismo , Histonas/metabolismo , Privação Materna , Proteína 2 de Ligação a Metil-CpG/metabolismo , RNA Mensageiro/metabolismo , Acetilação , Animais , Fator Neurotrófico Derivado do Encéfalo/efeitos dos fármacos , DNA (Citosina-5-)-Metiltransferase 1/efeitos dos fármacos , DNA (Citosina-5-)-Metiltransferases/efeitos dos fármacos , DNA Metiltransferase 3A , Epigênese Genética/efeitos dos fármacos , Éxons , Feminino , Hipocampo/efeitos dos fármacos , Histonas/efeitos dos fármacos , Masculino , Proteína 2 de Ligação a Metil-CpG/efeitos dos fármacos , Gravidez , Regiões Promotoras Genéticas , RNA Mensageiro/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley
7.
Orv Hetil ; 158(10): 380-385, 2017 Mar.
Artigo em Húngaro | MEDLINE | ID: mdl-28270006

RESUMO

INTRODUCTION: Nowadays, the food industry more often uses different type of additives during the food production. AIM: Our aim was to examine the monosodium-glutamate's effect (in animal experiment) on DNA-methyltransferases in gene expression patterns of mRNA levels. MATERIALS AND METHOD: In the investigation we used 24 (n=24) CD1 type female mice. The animals were fed with different equivalent human doses of the tested substance. After autopsy, mRNA was isolated from different tissues (lung, liver, kidney, spleen). DNMT1, DNMT3A and DNMT3B levels were determined by Quantitative Real-Time PCR. RESULTS: DNMT1 significantly suppressed the gene expression in all the three treated groups (p<0.05). The DNMT3A expression patterns showed significant decreasing tendency in the 1. and 2. treated groups of the lung tissue (p<0,05) and 1, 2, 3. groups of liver and kidney tissues (p<0,05). CONCLUSIONS: Our results shows that the monosodium glutamate, suppressed the DNMT1 and DNMT3A gene expression - on mRNA levels of several organs - in mice. It can be a similar chemopreventive effect to epigallo-catechin-gallate's, curcumin's, genistein's, likopine's and rezveratrol's effects. In this case it can be possible that the MSG has anticarcinogenic effects. Orv. Hetil., 2017, 158(10), 380-385.


Assuntos
DNA (Citosina-5-)-Metiltransferases/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Glutamato de Sódio/farmacologia , Animais , DNA (Citosina-5-)-Metiltransferase 1 , DNA Metiltransferase 3A , Modelos Animais de Doenças , Feminino , Camundongos , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , DNA Metiltransferase 3B
8.
Cell Reprogram ; 17(5): 404-14, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26035741

RESUMO

The present study was undertaken to examine the effect of Scriptaid treatment on histone acetylation, DNA methylation, expression of genes related to histone acetylation, and development of buffalo somatic cell nuclear transfer (SCNT) embryos. Treatment of buffalo SCNT embryos with 500 nM Scriptaid for 24 h resulted in a significant increase in the blastocyst formation rate (28.2% vs. 13.6%, p<0.05). Meanwhile, treatment of buffalo SCNT embryos with Scriptaid also resulted in higher acetylation levels of H3K18 and lower methylation levels of global DNA at the blastocyst stage, which was similar to fertilized counterparts. The expression levels of CBP, p300, HAT1, Dnmt1, and Dnmt3a in SCNT embryos treated with Scriptaid were significantly lower than the control group at the eight-cell stage (p<0.05), but the expression of HAT1 and Dnmt1a was higher than the control group at the blastocyst stage (p<0.05). When 96 blastocysts developed from Scriptaid-treated SCNT embryos were transferred into 48 recipients, 11 recipients (22.9%) became pregnant, whereas only one recipient (11.1%) became pregnant following transfer of 18 blastocysts developed from untreated SCNT embryos into nine recipients. These results indicate that treatment of buffalo SCNT embryos with Scriptaid can improve their developmental competence, and this action is mediated by resulting in a similar histone acetylation level and global DNA methylation level compared to in vitro-fertilized embryos through regulating the expression pattern of genes related to histone acetylation and DNA methylation.


Assuntos
Búfalos/embriologia , Metilação de DNA/efeitos dos fármacos , Desenvolvimento Embrionário/efeitos dos fármacos , Inibidores de Histona Desacetilases/farmacologia , Hidroxilaminas/farmacologia , Técnicas de Transferência Nuclear , Quinolinas/farmacologia , Acetilação , Animais , Blastocisto/efeitos dos fármacos , Blastocisto/metabolismo , Búfalos/metabolismo , Proteína de Ligação a CREB/efeitos dos fármacos , Proteína de Ligação a CREB/genética , DNA (Citosina-5-)-Metiltransferase 1 , DNA (Citosina-5-)-Metiltransferases/efeitos dos fármacos , DNA (Citosina-5-)-Metiltransferases/genética , DNA Metiltransferase 3A , Feminino , Expressão Gênica , Histona Acetiltransferases/efeitos dos fármacos , Histona Acetiltransferases/genética , Histonas/efeitos dos fármacos , Histonas/metabolismo , Gravidez
9.
Mutagenesis ; 30(2): 287-96, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25392149

RESUMO

To investigate polypeptide from Chlamy Farreri (PCF)'s protective effect against skin cancer, we used a cellular model of ultraviolet B (UVB)-induced malignant transformation. The human keratinocyte cell line HaCaT was repeatly exposed to UVB (10 mJ/cm(2), 20 times) and malignant transformation was confirmed by Gimesa staining, cell cycle analysis and various assays [anchorage independent growth, matrix metalloproteinase-9 (MMP9) activity, plating efficiency]. The malignant transformation was found to be effectively prevented by PCF pretreatment (2.84mM for 2h prior to each UVB exposure). We investigated the mechanism of PCF-mediated action by determining its effect on DNA methylation status of the tumour suppressor genes [P16 and ras association domain family 1 A (RASSF1A)] in the UVB-transformed cells. Both genes were found to be hypermethylated by chronic UVB exposure. The expression levels of P16, RASSF1A, DNA methyltransferases (DNMTs) and DNA damage inducible protein a (GADD45a) were measured by reverse transcriptase-polymerase chain reaction and western blotting. While chronic UVB exposure was found to suppress the expression of P16 and RASSF1A, it enhanced the expression of DNMT3b. In the early phase of UVB-induced malignant transformation, the GADD45a expression was increased, however, it declined with a continued irradiation of the cells. The UVB-induced DNA hypermethylation of P16 and RASSF1A and subsequent gene silencing was reversed by PCF treatment. The inhibition of DNMTs expression suggested that PCF blocked DNA methylation and thereby the silencing of tumour suppressor genes. Furthermore, the PCF-mediated substantial increase in GADD45a expression indicated that PCF promoted demethylation of tumour suppressor genes via GADD45a induction.


Assuntos
Transformação Celular Neoplásica/efeitos dos fármacos , Queratinócitos/efeitos da radiação , Neoplasias Induzidas por Radiação/prevenção & controle , Pectinidae , Peptídeos/farmacologia , Neoplasias Cutâneas/prevenção & controle , Raios Ultravioleta , Animais , Proteínas de Ciclo Celular/efeitos dos fármacos , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/efeitos da radiação , Linhagem Celular , Transformação Celular Neoplásica/genética , Inibidor p16 de Quinase Dependente de Ciclina , DNA (Citosina-5-)-Metiltransferases/efeitos dos fármacos , DNA (Citosina-5-)-Metiltransferases/genética , DNA (Citosina-5-)-Metiltransferases/efeitos da radiação , Metilação de DNA/efeitos dos fármacos , Metilação de DNA/efeitos da radiação , Expressão Gênica/efeitos dos fármacos , Expressão Gênica/efeitos da radiação , Humanos , Queratinócitos/efeitos dos fármacos , Proteínas de Neoplasias/efeitos dos fármacos , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/efeitos da radiação , Neoplasias Induzidas por Radiação/genética , Proteínas Nucleares/efeitos dos fármacos , Proteínas Nucleares/genética , Proteínas Nucleares/efeitos da radiação , Substâncias Protetoras/farmacologia , Neoplasias Cutâneas/genética , Proteínas Supressoras de Tumor/efeitos dos fármacos , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/efeitos da radiação , DNA Metiltransferase 3B
10.
AAPS J ; 16(4): 727-35, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24830944

RESUMO

Nrf2 is a crucial transcription factor that controls a critical anti-oxidative stress defense system and is implicated in skin homeostasis. Apigenin (API), a potent cancer chemopreventive agent, protects against skin carcinogenesis and elicits multiple molecular signaling pathways. However, the potential epigenetic effect of API in skin cancer chemoprotection is not known. In this study, bisulfite genomic DNA sequencing and methylated DNA immunoprecipitation were utilized to investigate the demethylation effect of API at 15 CpG sites in the Nrf2 promoter in mouse skin epidermal JB6 P + cells. In addition, qPCR and Western blot analyses were performed to evaluate the mRNA and protein expression of Nrf2 and the Nrf2 ARE downstream gene, NQO1. Finally, the protein expression levels of DNA methyltransferases (DNMTs) and histone deacetylases (HDACs) were evaluated using API and the DNMT/HDAC inhibitor 5-aza/ trichostatin A. Our results showed that API effectively reversed the hypermethylated status of the 15 CpG sites in the Nrf2 promoter in a dose-dependent manner. API enhanced the nuclear translocation of Nrf2 and increased the mRNA and protein expression of Nrf2 and the Nrf2 downstream target gene, NQO1. Furthermore, API reduced the expression of the DNMT1, DNMT3a, and DNMT3b epigenetic proteins as well as the expression of some HDACs (1-8). Taken together, our results showed that API can restore the silenced status of Nrf2 in skin epidermal JB6 P + cells by CpG demethylation coupled with attenuated DNMT and HDAC activity. These results may provide new therapeutic insights into the prevention of skin cancer by dietary phytochemicals.


Assuntos
Apigenina/farmacologia , Epiderme/metabolismo , Epigênese Genética , Fator 2 Relacionado a NF-E2/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Ativação Metabólica , Animais , Linhagem Celular , DNA (Citosina-5-)-Metiltransferases/biossíntese , DNA (Citosina-5-)-Metiltransferases/efeitos dos fármacos , Metilação de DNA/efeitos dos fármacos , Células Epidérmicas , Histona Desacetilase 1/biossíntese , Histona Desacetilase 1/efeitos dos fármacos , Camundongos , Fator 2 Relacionado a NF-E2/genética , Estresse Oxidativo/genética , Inibidores da Síntese de Proteínas/farmacologia , Transdução de Sinais/genética
11.
Curr Alzheimer Res ; 9(5): 555-62, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22272629

RESUMO

Late onset Alzheimer's disease (LOAD) is typical of the majority of Alzheimer's disease (AD) cases (~90%), and has no clear genetic association. Previous studies from our lab suggest that an epigenetic component could be involved. Developmental exposure of primates and rodents to lead (Pb) predetermined the expression of AD-related genes, such as the amyloid-ß precursor protein (AßPP), later in life. In addition to AßPP, the preponderance of genes that were reprogrammed was rich in CpG dinucleotides implicating DNA methylation and chromatin restructuring in their regulation. To examine the involvement of epigenetic intermediates in Pb-induced alterations in gene expression, differentiated SH-SY5Y cells were exposed to a series of Pb concentrations (5-100 µM) for 48 h and were analyzed for the protein expression of AßPP, ß-site amyloid precursor protein cleaving enzyme 1 (BACE1), specificity protein 1 and 3 (Sp1, Sp3) and epigenetic intermediates like DNA methyltransferase 1, 3a (Dnmt1, Dnmt3a) and methyl CpG binding protein 2 (MeCP2) involved in DNA methylation six days after the exposure had ceased. Western blot analysis indicated a significant latent elevation in AD biomarkers as well as the transcription factors Sp1 and Sp3, accompanied by a significant reduction in the protein levels of DNA methylating enzymes. RT-PCR analysis of Dnmt1, Dnmt3a and MeCP2 indicated a significant down-regulation of the mRNA levels. These data suggest that Pb interferes with DNA methylating capacity in these cells, thus altering the expression of AD-related genes.


Assuntos
Doença de Alzheimer/etiologia , Metilação de DNA/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Chumbo/toxicidade , Proteínas do Tecido Nervoso/efeitos dos fármacos , Doença de Alzheimer/metabolismo , Secretases da Proteína Precursora do Amiloide/efeitos dos fármacos , Secretases da Proteína Precursora do Amiloide/genética , Secretases da Proteína Precursora do Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/efeitos dos fármacos , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Ácido Aspártico Endopeptidases/efeitos dos fármacos , Ácido Aspártico Endopeptidases/genética , Ácido Aspártico Endopeptidases/metabolismo , Biomarcadores/metabolismo , Células Cultivadas , DNA (Citosina-5-)-Metiltransferases/efeitos dos fármacos , DNA (Citosina-5-)-Metiltransferases/genética , DNA (Citosina-5-)-Metiltransferases/metabolismo , Exposição Ambiental , Epigênese Genética/efeitos dos fármacos , Humanos , Proteína 2 de Ligação a Metil-CpG/efeitos dos fármacos , Proteína 2 de Ligação a Metil-CpG/genética , Proteína 2 de Ligação a Metil-CpG/metabolismo , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , RNA Mensageiro/análise , Fatores de Transcrição Sp/efeitos dos fármacos , Fatores de Transcrição Sp/genética , Fatores de Transcrição Sp/metabolismo
12.
Int J Neuropsychopharmacol ; 15(5): 669-79, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21682946

RESUMO

P11 (S100A10) has been associated with the pathophysiology of depression both in human and rodent models. Different types of antidepressants have been shown to increase P11 levels in distinct brain regions and P11 gene therapy was recently proven effective in reversing depressive-like behaviours in mice. However, the molecular mechanisms that govern P11 gene expression in response to antidepressants still remain elusive. In this study we report decreased levels of P11, associated with higher DNA methylation in the promoter region, in the prefrontal cortex of the Flinders Sensitive Line (FSL) genetic rodent model of depression. This hypermethylated pattern was reversed to normal, as indicated by the control line, after chronic administration of escitalopram (a selective serotonin reuptake inhibitor; SSRI). The escitalopram-induced hypomethylation was associated with both an increase in P11 gene expression and a reduction in mRNA levels of two DNA methyltransferases that have been shown to maintain DNA methylation in adult forebrain neurons (Dnmt1 and Dnmt3a). In conclusion, our data further support a role for P11 in depression-like states and suggest that this gene is controlled by epigenetic mechanisms that can be affected by antidepressant treatment.


Assuntos
Anexina A2/genética , Citalopram/farmacologia , Transtorno Depressivo/tratamento farmacológico , Epigenômica , Proteínas S100/genética , Inibidores Seletivos de Recaptação de Serotonina/farmacologia , Animais , Anexina A2/metabolismo , DNA (Citosina-5-)-Metiltransferase 1 , DNA (Citosina-5-)-Metiltransferases/efeitos dos fármacos , DNA Metiltransferase 3A , Transtorno Depressivo/genética , Transtorno Depressivo/metabolismo , Modelos Animais de Doenças , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Córtex Pré-Frontal/efeitos dos fármacos , RNA Mensageiro/biossíntese , Ratos , Proteínas S100/metabolismo
13.
Postepy Biochem ; 57(1): 24-32, 2011.
Artigo em Polonês | MEDLINE | ID: mdl-21735817

RESUMO

DNA methyltransferase 1 (DNMT1) is one of three enzymes independently coded in mammalian cells. It catalyses postreplicative synthesis of 5-methylcytosine in DNA. The aim of this modification is regulation of gene expression characteristic for the given organism. DNMT1 maintains methylation pattern by copying it from maternal to daughter stand. S-adenosylo-L-methionina is a donor of methyl group in his process. Disturbance in methylation level results in changes in cell differentiation and finally to tumor transformation. Hypermethylation of promotor or first exon of tumor suppression gene results in silencing of its transcription. While hypomethylation of regulatory sequence of protooncogene and retrotransposon make them transcriptionally active. DNMT1 as a key enzyme in maintaing of proper methylation pattern is a attractive target in anti-tumor therapy.


Assuntos
5-Metilcitosina/biossíntese , DNA (Citosina-5-)-Metiltransferases/efeitos dos fármacos , DNA (Citosina-5-)-Metiltransferases/metabolismo , Neoplasias/tratamento farmacológico , Neoplasias/enzimologia , Animais , DNA (Citosina-5-)-Metiltransferase 1 , Metilação de DNA , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias/genética
14.
Cancer Res ; 71(5): 1904-11, 2011 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-21239472

RESUMO

Thiopurines including 6-thioguanine ((S)G), 6-mercaptopurine, and azathioprine are effective anticancer agents with remarkable success in clinical practice, especially in effective treatment of acute lymphoblastic leukemia (ALL). (S)G is understood to act as a DNA hypomethylating agent in ALL cells, however, the underlying mechanism leading to global cytosine demethylation remains unclear. Here we report that (S)G treatment results in reactivation of epigenetically silenced genes in T leukemia cells. Bisulfite genomic sequencing revealed that (S)G treatment universally elicited demethylation in the promoters and/or first exons of the genes that were reactivated. (S)G treatment also attenuated the expression of histone lysine-specific demethylase 1 (LSD1), thereby stimulating lysine methylation of the DNA methylase DNMT1 and triggering its degradation via the ubiquitin-proteasomal pathway. Taken together, our findings reveal a previously uncharacterized but vital mechanistic link between (S)G treatment and DNA hypomethylation.


Assuntos
Antimetabólitos Antineoplásicos/farmacologia , DNA (Citosina-5-)-Metiltransferases/metabolismo , Expressão Gênica/efeitos dos fármacos , Inativação Gênica/efeitos dos fármacos , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Complexo de Endopeptidases do Proteassoma/metabolismo , Tioguanina/farmacologia , Western Blotting , Linhagem Celular Tumoral , Cromatografia Líquida de Alta Pressão , DNA (Citosina-5-)-Metiltransferase 1 , DNA (Citosina-5-)-Metiltransferases/efeitos dos fármacos , Metilação de DNA , Humanos , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
15.
J Clin Invest ; 120(2): 521-32, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20093774

RESUMO

DNA methyltransferase 1 (DNMT1) catalyzes DNA methylation and is overexpressed in many human diseases, including cancer. The tobacco-specific carcinogen NNK also induces DNA methylation. However, the role of DNMT1-mediated methylation in tobacco carcinogenesis remains unclear. Here we used human and mouse lung cancer samples and cell lines to determine a mechanism whereby NNK induced DNMT1 expression and activity. We determined that in a human lung cell line, glycogen synthase kinase 3beta (GSK3beta) phosphorylated DNMT1 to recruit beta-transducin repeat-containing protein (betaTrCP), resulting in DNMT1 degradation, and that NNK activated AKT, inhibiting GSK3beta function and thereby attenuating DNMT1 degradation. NNK also induced betaTrCP translocation to the cytoplasm via the heterogeneous nuclear ribonucleoprotein U (hnRNP-U) shuttling protein, resulting in DNMT1 nuclear accumulation and hypermethylation of the promoters of tumor suppressor genes. Fluorescence immunohistochemistry (IHC) of lung adenomas from NNK-treated mice and tumors from lung cancer patients that were smokers were characterized by disruption of the DNMT1/betaTrCP interaction and DNMT1 nuclear accumulation. Importantly, DNMT1 overexpression in lung cancer patients who smoked continuously correlated with poor prognosis. We believe that the NNK-induced DNMT1 accumulation and subsequent hypermethylation of the promoter of tumor suppressor genes may lead to tumorigenesis and poor prognosis and provide an important link between tobacco smoking and lung cancer. Furthermore, this mechanism may also be involved in other smoking-related human diseases.


Assuntos
DNA (Citosina-5-)-Metiltransferases/biossíntese , Genes Supressores de Tumor/efeitos dos fármacos , Neoplasias Pulmonares/enzimologia , Nitrosaminas/toxicidade , Animais , Carcinógenos/toxicidade , Linhagem Celular , Linhagem Celular Tumoral , DNA (Citosina-5-)-Metiltransferase 1 , DNA (Citosina-5-)-Metiltransferases/efeitos dos fármacos , Metilação de DNA/efeitos dos fármacos , Humanos , Camundongos , Fosforilação , Prognóstico , Fumar/efeitos adversos
16.
Int J Cancer ; 126(11): 2520-33, 2010 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-19856314

RESUMO

Epigenetic silencing of gluthathione-S-transferase pi (GSTP1) is recognized as being a molecular hallmark of human prostate cancer. We investigated the effects of green tea polyphenols (GTPs) on GSTP1 re-expression and further elucidated its mechanism of action and long-term safety, compared with nucleoside-analog inhibitor of DNA methyltransferase (DNMT), 5-aza-2'-deoxycitidine. Exposure of human prostate cancer LNCaP cells to 1-10 microg/ml of GTP for 1-7 days caused a concentration- and time-dependent re-expression of GSTP1, which correlated with DNMT1 inhibition. Methyl-specific-PCR and sequencing revealed extensive demethylation in the proximal GSTP1 promoter and regions distal to the transcription factor binding sites. GTP exposure in a time-dependent fashion diminished the mRNA and protein levels of MBD1, MBD4 and MeCP2; HDAC 1-3 and increased the levels of acetylated histone H3 (LysH9/18) and H4. Chromatin immunoprecipitation assays demonstrated that cells treated with GTP have reduced MBD2 association with accessible Sp1 binding sites leading to increased binding and transcriptional activation of the GSTP1 gene. Exposure of cells to GTP did not result in global hypomethylation, as demonstrated by methyl-specific PCR for LINE-1 promoter; rather GTP promotes maintenance of genomic integrity. Furthermore, exposure of cells to GTP did not cause activation of the prometaststic gene S100P, a reverse response noted after exposure of cells to 5-aza-2'deoxycitidine. Our results, for the first time, demonstrate that GTP has dual potential to alter DNA methylation and chromatin modeling, the 2 global epigenetic mechanisms of gene regulation and their lack of toxicity makes them excellent candidates for the chemoprevention of prostate cancer.


Assuntos
Flavonoides/farmacologia , Glutationa S-Transferase pi/genética , Fenóis/farmacologia , Regiões Promotoras Genéticas/efeitos dos fármacos , Regiões Promotoras Genéticas/genética , Neoplasias da Próstata/genética , Linhagem Celular Tumoral , DNA (Citosina-5-)-Metiltransferase 1 , DNA (Citosina-5-)-Metiltransferases/efeitos dos fármacos , DNA (Citosina-5-)-Metiltransferases/genética , Metilação de DNA/genética , Células Epiteliais/fisiologia , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Inativação Gênica , Guanosina Trifosfato/farmacologia , Histonas/isolamento & purificação , Humanos , Imunoglobulinas/genética , Imuno-Histoquímica , Masculino , Polifenóis , Próstata/citologia , Próstata/fisiologia , Neoplasias da Próstata/enzimologia , Neoplasias da Próstata/patologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Chá/química
17.
Cancer Res ; 69(10): 4277-85, 2009 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-19417133

RESUMO

Reactivation of silenced tumor suppressor genes by 5-azacytidine (Vidaza) and its congener 5-aza-2'-deoxycytidine (decitabine) has provided an alternate approach to cancer therapy. We have shown previously that these drugs selectively and rapidly induce degradation of the maintenance DNA methyltransferase (DNMT) 1 by a proteasomal pathway. Because the toxicity of these compounds is largely due to their incorporation into DNA, it is critical to explore novel, nonnucleoside compounds that can effectively reactivate the silenced genes. Here, we report that a quinoline-based compound, designated SGI-1027, inhibits the activity of DNMT1, DNMT3A, and DNMT3B as well M. SssI with comparable IC(50) (6-13 micromol/L) by competing with S-adenosylmethionine in the methylation reaction. Treatment of different cancer cell lines with SGI-1027 resulted in selective degradation of DNMT1 with minimal or no effects on DNMT3A and DNMT3B. At a concentration of 2.5 to 5 micromol/L (similar to that of decitabine), complete degradation of DNMT1 protein was achieved within 24 h without significantly affecting its mRNA level. MG132 blocked SGI-1027-induced depletion of DNMT1, indicating the involvement of proteasomal pathway. Prolonged treatment of RKO cells with SGI-1027 led to demethylation and reexpression of the silenced tumor suppressor genes P16, MLH1, and TIMP3. Further, this compound did not exhibit significant toxicity in a rat hepatoma (H4IIE) cell line. This study provides a novel class of DNA hypomethylating agents that have the potential for use in epigenetic cancer therapy.


Assuntos
Azacitidina/toxicidade , Metilação de DNA/efeitos dos fármacos , Inativação Gênica/efeitos dos fármacos , Genes Supressores de Tumor/efeitos dos fármacos , Quinolinas/toxicidade , Animais , Neoplasias da Mama , Carcinoma Hepatocelular , Linhagem Celular Tumoral , Neoplasias do Colo , DNA (Citosina-5-)-Metiltransferase 1 , DNA (Citosina-5-)-Metiltransferases/antagonistas & inibidores , DNA (Citosina-5-)-Metiltransferases/efeitos dos fármacos , DNA (Citosina-5-)-Metiltransferases/genética , DNA (Citosina-5-)-Metiltransferases/isolamento & purificação , DNA Metiltransferase 3A , Metilases de Modificação do DNA/metabolismo , Feminino , Células HeLa , Humanos , Neoplasias Hepáticas , Camundongos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , DNA Metiltransferase 3B
18.
Carcinogenesis ; 29(6): 1267-75, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18204074

RESUMO

Nickel (Ni) compounds are potent carcinogens and can induce malignant transformation of rodent and human cells. To uncover the molecular mechanisms of nickel sulfide (NiS)-induced cell transformation, we investigated epigenetic alterations in a set of DNA repair genes. The silencing of the O(6)-methylguanine DNA methyltransferase (MGMT) gene locus and upregulation of DNA methyltransferase 1 (DNMT1) expression was specifically detected in NiS-transformed human bronchial epithelial (16HBE) cells. In addition, we noted epigenetic alterations including DNA hypermethylation, reduced histone H4 acetylation and a decrease in the ratio of Lys-9 acetylated/methylated histone H3 at the MGMT CpG island in NiS-transformed 16HBE cells. Meanwhile, we identified concurrent binding of methyl-CpG-binding protein 2, methylated DNA-binding domain protein 2 and DNMT1 to the CpG island of the MGMT promoter, demonstrating that these components collaborate to maintain MGMT methylation in NiS-transformed cells. Moreover, depletion of DNMT1 by introduction of a small hairpin RNA construct into NiS-transformed cells resulted in a 30% inhibition of cell proliferation and led to increased MGMT gene expression by reversion of the epigenetic modifications at the MGMT promoter region. MGMT suppression and hypermethylation at the CpG island of the MGMT promoter occurred 6 days after NiS treatment, indicating that epigenetic modifications of MGMT might be an early event in tumorigenesis. Taken together, these observations demonstrate that epigenetic silencing of MGMT is associated with DNA hypermethylation, histone modifications and DNMT1 upregulation, which contribute to NiS-induced malignant transformation.


Assuntos
Carcinógenos/toxicidade , Transformação Celular Neoplásica/genética , DNA (Citosina-5-)-Metiltransferases/genética , Metilases de Modificação do DNA/genética , Enzimas Reparadoras do DNA/genética , Inativação Gênica , Níquel/toxicidade , Proteínas Supressoras de Tumor/genética , Western Blotting , Linhagem Celular , DNA (Citosina-5-)-Metiltransferase 1 , DNA (Citosina-5-)-Metiltransferases/efeitos dos fármacos , DNA (Citosina-5-)-Metiltransferases/metabolismo , Metilação de DNA , Metilases de Modificação do DNA/efeitos dos fármacos , Metilases de Modificação do DNA/metabolismo , Enzimas Reparadoras do DNA/efeitos dos fármacos , Enzimas Reparadoras do DNA/metabolismo , Expressão Gênica , Histonas/genética , Humanos , Imunoprecipitação , RNA Mensageiro/análise , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transcrição Gênica , Proteínas Supressoras de Tumor/efeitos dos fármacos , Proteínas Supressoras de Tumor/metabolismo
19.
Blood ; 111(4): 2364-73, 2008 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-18083845

RESUMO

Bortezomib reversibly inhibits 26S proteasomal degradation, interferes with NF-kappaB, and exhibits antitumor activity in human malignancies. Zinc finger protein Sp1 transactivates DNMT1 gene in mice and is functionally regulated through protein abundance, posttranslational modifications (ie, ubiquitination), or interaction with other transcription factors (ie, NF-kappaB). We hypothesize that inhibition of proteasomal degradation and Sp1/NF-kappaB-mediated transactivation may impair aberrant DNA methyltransferase activity. We show here that, in addition to inducing accumulation of polyubiquitinated proteins and abolishment of NF-kappaB activities, bortezomib decreases Sp1 protein levels, disrupts the physical interaction of Sp1/NF-kappaB, and prevents binding of the Sp1/NF-kappaB complex to the DNMT1 gene promoter. Abrogation of Sp1/NF-kappaB complex by bortezomib causes transcriptional repression of DNMT1 gene and down-regulation of DNMT1 protein, which in turn induces global DNA hypomethylation in vitro and in vivo and re-expression of epigenetically silenced genes in human cancer cells. The involvement of Sp1/NF-kappaB in DNMT1 regulation is further demonstrated by the observation that Sp1 knockdown using mithramycin A or shRNA decreases DNMT1 protein levels, which instead are increased by Sp1 or NF-kappaB overexpression. Our results unveil the Sp1/NF-kappaB pathway as a modulator of DNA methyltransferase activity in human cancer and identify bortezomib as a novel epigenetic-targeting drug.


Assuntos
Antineoplásicos/farmacologia , Ácidos Borônicos/farmacologia , DNA (Citosina-5-)-Metiltransferases/metabolismo , Metilação de DNA/efeitos dos fármacos , Inativação Gênica/efeitos dos fármacos , Leucemia Mieloide Aguda/genética , NF-kappa B/fisiologia , Pirazinas/farmacologia , Transcrição Gênica/efeitos dos fármacos , Bortezomib , DNA (Citosina-5-)-Metiltransferases/efeitos dos fármacos , Humanos , Proteínas Quinases/efeitos dos fármacos , Proteínas Quinases/genética , Proteínas Quinases/metabolismo
20.
Environ Health Perspect ; 115(10): 1454-9, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17938735

RESUMO

BACKGROUND: Aberrant DNA methylation is common in carcinogenesis. The typical pattern appears to involve reduced expression of maintenance DNA methyltransferase, DNMT1, inducing genomic hypomethylation, whereas increased expression of de novo DNMT3a or 3b causes gene-specific hypermethylation. OBJECTIVES: During cadmium-induced malignant transformation, an unusual pattern of genomic hypermethylation occurred that we studied to provide insight into the roles of specific DNMTs in oncogenesis. METHODS: Gene expression and DNA methylation were assessed in control and chronic cadmium-transformed prostate epithelial cells (CTPE) using reverse transcription-polymerase chain reaction (RT-PCR), Western blot analysis, methylation-specific PCR, and methyl acceptance assay. RESULTS: During the 10-weeks of cadmium exposure that induced malignant transformation, progressive increases in generalized DNMT enzymatic activity occurred that were associated with over-expression of DNMT3b without changes in DNMT1 expression. Increased DNMT3b expression preceded increased DNMT enzymatic activity. Procainamide, a specific DNMT1 inhibitor, reversed cadmium-induced genomic DNA hypermethylation. Reduced expression of the tumor suppressor genes, RASSF1A and p16, began about the time DNMT3b overexpression first occurred and progressively decreased thereafter. RASSF1A and p16 promoter regions were heavily methylated in CTPE cells, indicating silencing by hypermethylation, while the DNA demethylating agent, 5-aza-2'-deoxycytidine, reversed this silencing. DNMT1 inhibition only modestly increased RASSF1A and p16 expression in CTPE cells and did not completely reverse silencing. CONCLUSIONS: These data indicate that DNMT3b overexpression can result in generalized DNA hypermethylation and gene silencing but that DNMT1 is required to maintain these effects. The pattern of genomic DNA hypermethylation together with up-regulation of DNMT3b may provide a unique set of biomarkers to specifically identify cadmium-induced human prostate cancers.


Assuntos
Cádmio/toxicidade , Carcinógenos Ambientais/toxicidade , Transformação Celular Neoplásica , Metilação de DNA/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Próstata , Estudos de Casos e Controles , Transformação Celular Neoplásica/induzido quimicamente , Transformação Celular Neoplásica/genética , Células Cultivadas , DNA (Citosina-5-)-Metiltransferase 1 , DNA (Citosina-5-)-Metiltransferases/efeitos dos fármacos , DNA (Citosina-5-)-Metiltransferases/metabolismo , Células Epiteliais/efeitos dos fármacos , Perfilação da Expressão Gênica , Genes Supressores de Tumor/efeitos dos fármacos , Humanos , Masculino , Próstata/citologia , Próstata/efeitos dos fármacos , Regulação para Cima , DNA Metiltransferase 3B
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...