Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
J Natl Cancer Inst ; 116(6): 974-982, 2024 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-38273663

RESUMO

BACKGROUND: The phenomenon of field cancerization reflects the transition of normal cells into those predisposed to cancer. Assessing the scope and intensity of this process in the colon may support risk prediction and colorectal cancer prevention. METHODS: The Swiss Epigenetic Colorectal Cancer Study (SWEPIC) study, encompassing 1111 participants for DNA methylation analysis and a subset of 84 for RNA sequencing, was employed to detect field cancerization in individuals with adenomatous polyps (AP). Methylation variations were evaluated for their discriminative capability, including in external cohorts, genomic localization, clinical correlations, and associated RNA expression patterns. RESULTS: Normal cecal tissue of individuals harboring an AP in the proximal colon manifested dysregulated DNA methylation compared to tissue from healthy individuals at 558 unique loci. Leveraging these adenoma-related differentially variable and methylated CpGs (aDVMCs), our classifier discerned between healthy and AP-adjacent tissues across SWEPIC datasets (cross-validated area under the receiver operating characteristic curve [ROC AUC] = 0.63-0.81), including within age-stratified cohorts. This discriminative capacity was validated in 3 external sets, differentiating healthy from cancer-adjacent tissue (ROC AUC = 0.82-0.88). Notably, aDVMC dysregulation correlated with polyp multiplicity. More than 50% of aDVMCs were significantly associated with age. These aDVMCs were enriched in active regions of the genome (P < .001), and associated genes exhibited altered expression in AP-adjacent tissues. CONCLUSIONS: Our findings underscore the early onset of field cancerization in the right colon during the neoplastic transformation process. A more extensive validation of aDVMC dysregulation as a stratification tool could pave the way for enhanced surveillance approaches, especially given its linkage to adenoma emergence.


Assuntos
Pólipos Adenomatosos , Metilação de DNA , Humanos , Pólipos Adenomatosos/genética , Pólipos Adenomatosos/patologia , Feminino , Masculino , Pessoa de Meia-Idade , Idoso , Biomarcadores Tumorais/genética , Mucosa Intestinal/patologia , Mucosa Intestinal/metabolismo , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Regulação Neoplásica da Expressão Gênica , Transformação Celular Neoplásica/genética , Ilhas de CpG/genética , Epigênese Genética
2.
J Gastroenterol ; 58(9): 856-867, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37300599

RESUMO

BACKGROUND: Individual colorectal polyp risk factors are well characterized; however, insights into their pathway-specific interactions are scarce. We aimed to identify the impact of individual risk factors and their joint effects on adenomatous (AP) and serrated polyp (SP) risk. METHODS: We collected information on 363 lifestyle and metabolic parameters from 1597 colonoscopy participants, resulting in over 521,000 data points. We used multivariate statistics and machine-learning approaches to assess associations of single variables and their interactions with AP and SP risk. RESULTS: Individual factors and their interactions showed common and polyp subtype-specific effects. Abdominal obesity, high body mass index (BMI), metabolic syndrome, and red meat consumption globally increased polyp risk. Age, gender, and western diet associated with AP risk, while smoking was associated with SP risk. CRC family history was associated with advanced adenomas and diabetes with sessile serrated lesions. Regarding lifestyle factor interactions, no lifestyle or dietary adjustments mitigated the adverse smoking effect on SP risk, whereas its negative effect was exacerbated by alcohol in the conventional pathway. The adverse effect of red meat on SP risk was not ameliorated by any factor, but was further exacerbated by western diet along the conventional pathway. No modification of any factor reduced the negative impact of metabolic syndrome on AP risk, whereas increased fatless fish or meat substitutes' intake mitigated its effect on SP risk. CONCLUSIONS: Individual risk factors and their interactions for polyp formation along the adenomatous and serrated pathways are strongly heterogeneous. Our findings may facilitate tailored lifestyle recommendations and contribute to a better understanding of how risk factor combinations impact colorectal carcinogenesis.


Assuntos
Adenoma , Pólipos Adenomatosos , Pólipos do Colo , Neoplasias Colorretais , Síndrome Metabólica , Humanos , Pólipos do Colo/epidemiologia , Pólipos do Colo/etiologia , Síndrome Metabólica/etiologia , Síndrome Metabólica/complicações , Neoplasias Colorretais/epidemiologia , Neoplasias Colorretais/etiologia , Adenoma/epidemiologia , Adenoma/etiologia , Adenoma/patologia , Fatores de Risco , Colonoscopia , Pólipos Adenomatosos/epidemiologia , Pólipos Adenomatosos/etiologia
3.
Clin Epigenetics ; 12(1): 164, 2020 11 03.
Artigo em Inglês | MEDLINE | ID: mdl-33143725

RESUMO

BACKGROUND: Colon cancer (CC) is the third most common cancer worldwide, highlighting the importance of developing effective prevention strategies. Accumulating evidence supports that aspirin use reduces CC incidence. We reported previously that aspirin suppresses age-associated and CC-relevant DNA methylation (DNAm) in healthy colon. Here we addressed the aspirin's effectiveness in longitudinal cohort. METHODS: We measured genome-wide DNAm in 124 healthy normal mucosa samples taken at baseline (time point 1, t1) and after 10-years follow-up (time point 2, t2) from a longitudinal female screening cohort. We investigated the time-dependent methylation drift in aspirin users and nonusers using multivariable regression and related the modulatory effect of aspirin to colonic epigenome-aging and CC. RESULTS: Over time, compared to nonusers, long-term (≥ 2 years) aspirin users showed less hypermethylated CpGs (proximal: 17% vs. 87%; distal: 16% vs. 70%) and more hypomethylated CpGs (proximal: 83% vs. 13%; distal: 84% vs. 30%). Overall, users showed 2% (P = 0.02) less mean methylation levels than nonusers in proximal colon and displayed repressed methylation age (mAge). Methylation loss in users occurred at several CC-specific tumor suppressors that gained methylation in nonusers. Methylation loss in users effected genes involved in immune system and inflammation, while methylation gain in nonusers effected genes involved in metabolism. CONCLUSIONS: This is the first longitudinal study demonstrating effectiveness of aspirin-use in suppression of age-related and CC-relevant hypermethylation in the normal colon. These findings provide a rationale for future studies to evaluate loci that may serve as markers to identify individuals that will benefit most from aspirin and hence increase its efficiency in CC prevention and therapy.


Assuntos
Envelhecimento/genética , Aspirina/metabolismo , Neoplasias do Colo/genética , Inibidores de Ciclo-Oxigenase/metabolismo , Idoso , Idoso de 80 Anos ou mais , Envelhecimento/efeitos dos fármacos , Aspirina/química , Estudos de Casos e Controles , Colo/metabolismo , Neoplasias do Colo/epidemiologia , Neoplasias do Colo/patologia , Neoplasias do Colo/prevenção & controle , Ilhas de CpG , Inibidores de Ciclo-Oxigenase/química , Metilação de DNA , Detecção Precoce de Câncer/métodos , Epigênese Genética/efeitos dos fármacos , Epigenômica , Feminino , Seguimentos , Estudo de Associação Genômica Ampla , Voluntários Saudáveis , Humanos , Incidência , Estudos Longitudinais , Pessoa de Meia-Idade
4.
Genes (Basel) ; 11(4)2020 03 25.
Artigo em Inglês | MEDLINE | ID: mdl-32218170

RESUMO

Modulated electromagnetic fields (wEMFs), as generated by modern communication technologies, have raised concerns about adverse health effects. The International Agency for Research on Cancer (IARC) classifies them as "possibly carcinogenic to humans" (Group 2B), yet, the underlying molecular mechanisms initiating and promoting tumorigenesis remain elusive. Here, we comprehensively assess the impact of technologically relevant wEMF modulations on the genome integrity of cultured human cells, investigating cell type-specificities as well as time- and dose-dependencies. Classical and advanced methodologies of genetic toxicology and DNA repair were applied, and key experiments were performed in two separate laboratories. Overall, we found no conclusive evidence for an induction of DNA damage nor for alterations of the DNA repair capacity in cells exposed to several wEMF modulations (i.e., GSM, UMTS, WiFi, and RFID). Previously reported observations of increased DNA damage after exposure of cells to GSM-modulated signals could not be reproduced. Experimental variables, presumably underlying the discrepant observations, were investigated and are discussed. On the basis of our data, we conclude that the possible carcinogenicity of wEMF modulations cannot be explained by an effect on genome integrity through direct DNA damage. However, we cannot exclude non-genotoxic, indirect, or secondary effects of wEMF exposure that may promote tumorigenesis in other ways.


Assuntos
Dano ao DNA , Campos Eletromagnéticos/efeitos adversos , Fibroblastos/patologia , Pulmão/patologia , Tecnologia sem Fio/instrumentação , Telefone Celular , Células Cultivadas , Reparo do DNA , Fibroblastos/efeitos da radiação , Humanos , Pulmão/efeitos da radiação
5.
Clin Epigenetics ; 11(1): 196, 2019 12 16.
Artigo em Inglês | MEDLINE | ID: mdl-31842975

RESUMO

BACKGROUND: Aberrations in DNA methylation are widespread in colon cancer (CC). Understanding origin and progression of DNA methylation aberrations is essential to develop effective preventive and therapeutic strategies. Here, we aimed to dissect CC subtype-specific methylation instability to understand underlying mechanisms and functions. METHODS: We have assessed genome-wide DNA methylation in the healthy normal colon mucosa (HNM), precursor lesions and CCs in a first comprehensive study to delineate epigenetic change along the process of colon carcinogenesis. Mechanistically, we used stable cell lines, genetically engineered mouse model of mutant BRAFV600E and molecular biology analysis to establish the role of BRAFV600E-mediated-TET inhibition in CpG-island methylator phenotype (CIMP) inititation. RESULTS: We identified two distinct patterns of CpG methylation instability, determined either by age-lifestyle (CC-neutral CpGs) or genetically (CIMP-CpGs). CC-neutral-CpGs showed age-dependent hypermethylation in HNM, all precursors, and CCs, while CIMP-CpGs showed hypermethylation specifically in sessile serrated adenomas/polyps (SSA/Ps) and CIMP-CCs. BRAFV600E-mutated CCs and precursors showed a significant downregulation of TET1 and TET2 DNA demethylases. Stable expression of BRAFV600E in nonCIMP CC cells and in a genetic mouse model was sufficient to repress TET1/TET2 and initiate hypermethylation at CIMP-CpGs, reversible by BRAFV600E inhibition. BRAFV600E-driven CIMP-CpG hypermethylation occurred at genes associated with established CC pathways, effecting functional changes otherwise achieved by genetic mutation in carcinogenesis. CONCLUSIONS: Hence, while age-lifestyle-driven hypermethylation occurs generally in colon carcinogenesis, BRAFV600E-driven hypermethylation is specific for the "serrated" pathway. This knowledge will advance the use of epigenetic biomarkers to assess subgroup-specific CC risk and disease progression.


Assuntos
Neoplasias do Colo/genética , Metilação de DNA , Proteínas de Ligação a DNA/metabolismo , Oxigenases de Função Mista/metabolismo , Mutação , Proteínas Proto-Oncogênicas B-raf/genética , Proteínas Proto-Oncogênicas/metabolismo , Animais , Células CACO-2 , Linhagem Celular Tumoral , Neoplasias do Colo/metabolismo , Dioxigenases , Regulação para Baixo , Epigênese Genética , Feminino , Redes Reguladoras de Genes , Células HT29 , Humanos , Masculino , Camundongos , Neoplasias Experimentais
6.
Neoplasia ; 21(2): 185-196, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30591423

RESUMO

Cell lines are essential tools to standardize and compare experimental findings in basic and translational cancer research. The current dogma states that cancer stem cells feature an increased tumor initiation capacity and are also chemoresistant. Here, we identified and comprehensively characterized three morphologically distinct cellular subtypes in the non-small cell lung cancer cell line A549 and challenge the current cancer stem cell dogma. Subtype-specific cellular morphology is maintained during short-term culturing, resulting in the formation of holoclonal, meroclonal, and paraclonal colonies. A549 holoclone cells were characterized by an epithelial and stem-like phenotype, paraclone cells featured a mesenchymal phenotype, whereas meroclone cells were phenotypically intermediate. Cell-surface marker expression of subpopulations changed over time, indicating an active epithelial-to-mesenchymal transition (EMT), in vitro and in vivo. EMT has been associated with the overexpression of the immunomodulators PD-L1 and PD-L2, which were 37- and 235-fold overexpressed in para- versus holoclone cells, respectively. We found that DNA methylation is involved in epigenetic regulation of marker expression. Holoclone cells were extremely sensitive to cisplatin and radiotherapy in vitro, whereas paraclone cells were highly resistant. However, inhibition of the receptor tyrosine kinase AXL, whose expression is associated with an EMT, specifically targeted the otherwise highly resistant paraclone cells. Xenograft tumor formation capacity was 24- and 269-fold higher in holo- than mero- and paraclone cells, respectively. Our results show that A549 subpopulations might serve as a unique system to explore the network of stemness, cellular plasticity, tumor initiation capacity, invasive and metastatic potential, and chemo/radiotherapy resistance.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/patologia , Transformação Celular Neoplásica , Transição Epitelial-Mesenquimal , Neoplasias Pulmonares/patologia , Células-Tronco Neoplásicas/metabolismo , Células A549 , Animais , Biomarcadores , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Transformação Celular Neoplásica/genética , Dano ao DNA , Metilação de DNA , Modelos Animais de Doenças , Transição Epitelial-Mesenquimal/genética , Perfilação da Expressão Gênica , Humanos , Imunofenotipagem , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Camundongos , Células-Tronco Neoplásicas/patologia , Transcriptoma
7.
Sci Rep ; 7: 43345, 2017 03 07.
Artigo em Inglês | MEDLINE | ID: mdl-28266526

RESUMO

Extremely-low-frequency magnetic fields (ELF-MF) have been classified as "possibly carcinogenic" to humans on the grounds of an epidemiological association of ELF-MF exposure with an increased risk of childhood leukaemia. Yet, underlying mechanisms have remained obscure. Genome instability seems an unlikely reason as the energy transmitted by ELF-MF is too low to damage DNA and induce cancer-promoting mutations. ELF-MF, however, may perturb the epigenetic code of genomes, which is well-known to be sensitive to environmental conditions and generally deranged in cancers, including leukaemia. We examined the potential of ELF-MF to influence key epigenetic modifications in leukaemic Jurkat cells and in human CD34+ haematopoietic stem cells undergoing in vitro differentiation into the neutrophilic lineage. During granulopoiesis, sensitive genome-wide profiling of multiple replicate experiments did not reveal any statistically significant, ELF-MF-dependent alterations in the patterns of active (H3K4me2) and repressive (H3K27me3) histone marks nor in DNA methylation. However, ELF-MF exposure showed consistent effects on the reproducibility of these histone and DNA modification profiles (replicate variability), which appear to be of a stochastic nature but show preferences for the genomic context. The data indicate that ELF-MF exposure stabilizes active chromatin, particularly during the transition from a repressive to an active state during cell differentiation.


Assuntos
Epigênese Genética/efeitos da radiação , Células-Tronco Hematopoéticas/efeitos da radiação , Células Jurkat/efeitos da radiação , Campos Magnéticos , Diferenciação Celular/efeitos da radiação , DNA/metabolismo , Histonas/metabolismo , Humanos , Metilação
8.
DNA Repair (Amst) ; 44: 92-102, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27247237

RESUMO

Pathways that control and modulate DNA methylation patterning in mammalian cells were poorly understood for a long time, although their importance in establishing and maintaining cell type-specific gene expression was well recognized. The discovery of proteins capable of converting 5-methylcytosine (5mC) to putative substrates for DNA repair introduced a novel and exciting conceptual framework for the investigation and ultimate discovery of molecular mechanisms of DNA demethylation. Against the prevailing notion that DNA methylation is a static epigenetic mark, it turned out to be dynamic and distinct mechanisms appear to have evolved to effect global and locus-specific DNA demethylation. There is compelling evidence that DNA repair, in particular base excision repair, contributes significantly to the turnover of 5mC in cells. By actively demethylating DNA, DNA repair supports the developmental establishment as well as the maintenance of DNA methylation landscapes and gene expression patterns. Yet, while the biochemical pathways are relatively well-established and reviewed, the biological context, function and regulation of DNA repair-mediated active DNA demethylation remains uncertain. In this review, we will thus summarize and critically discuss the evidence that associates active DNA demethylation by DNA repair with specific functional contexts including the DNA methylation erasure in the early embryo, the control of pluripotency and cellular differentiation, the maintenance of cell identity, and the nuclear reprogramming.


Assuntos
DNA Glicosilases/genética , Reparo do DNA , DNA/genética , Epigênese Genética , Oxigenases de Função Mista/genética , Proteínas Proto-Oncogênicas/genética , 5-Metilcitosina/metabolismo , Animais , Reprogramação Celular , DNA/metabolismo , Dano ao DNA , DNA Glicosilases/metabolismo , Metilação de DNA , Embrião de Mamíferos , Humanos , Oxigenases de Função Mista/metabolismo , Família Multigênica , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo , Proteínas Proto-Oncogênicas/metabolismo
9.
Bioelectromagnetics ; 37(3): 183-189, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26991812

RESUMO

Exposure to extremely low-frequency magnetic fields (ELF-MF) was evaluated in an International Agency for Research on Cancer (IARC) Monographs as "possibly carcinogenic to humans" in 2001, based on increased childhood leukemia risk observed in epidemiological studies. We conducted a hazard assessment using available scientific evidence published before March 2015, with inclusion of new research findings from the Advanced Research on Interaction Mechanisms of electroMagnetic exposures with Organisms for Risk Assessment (ARIMMORA) project. The IARC Monograph evaluation scheme was applied to hazard identification. In ARIMMORA for the first time, a transgenic mouse model was used to mimic the most common childhood leukemia: new pathogenic mechanisms were indicated, but more data are needed to draw definitive conclusions. Although experiments in different animal strains showed exposure-related decreases of CD8+ T-cells, a role in carcinogenesis must be further established. No direct damage of DNA by exposure was observed. Overall in the literature, there is limited evidence of carcinogenicity in humans and inadequate evidence of carcinogenicity in experimental animals, with only weak supporting evidence from mechanistic studies. New exposure data from ARIMMORA confirmed that if the association is nevertheless causal, up to 2% of childhood leukemias in Europe, as previously estimated, may be attributable to ELF-MF. In summary, ARIMMORA concludes that the relationship between ELF-MF and childhood leukemia remains consistent with possible carcinogenicity in humans. While this scientific uncertainty is dissatisfactory for science and public health, new mechanistic insight from ARIMMORA experiments points to future research that could provide a step-change in future assessments. Bioelectromagnetics. 37:183-189, 2016. © 2016 Wiley Periodicals, Inc.

10.
Nat Commun ; 7: 10806, 2016 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-26932196

RESUMO

Cytosine methylation in CpG dinucleotides is an epigenetic DNA modification dynamically established and maintained by DNA methyltransferases and demethylases. Molecular mechanisms of active DNA demethylation began to surface only recently with the discovery of the 5-methylcytosine (5mC)-directed hydroxylase and base excision activities of ten-eleven translocation (TET) proteins and thymine DNA glycosylase (TDG). This implicated a pathway operating through oxidation of 5mC by TET proteins, which generates substrates for TDG-dependent base excision repair (BER) that then replaces 5mC with C. Yet, direct evidence for a productive coupling of TET with BER has never been presented. Here we show that TET1 and TDG physically interact to oxidize and excise 5mC, and proof by biochemical reconstitution that the TET-TDG-BER system is capable of productive DNA demethylation. We show that the mechanism assures a sequential demethylation of symmetrically methylated CpGs, thereby avoiding DNA double-strand break formation but contributing to the mutability of methylated CpGs.


Assuntos
Metilação de DNA , Reparo do DNA/fisiologia , Proteínas de Ligação a DNA/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Timina DNA Glicosilase/metabolismo , Ilhas de CpG , Citosina/metabolismo , Quebras de DNA de Cadeia Dupla , Proteínas de Ligação a DNA/genética , Epigênese Genética , Escherichia coli/metabolismo , Regulação da Expressão Gênica/fisiologia , Proteínas Proto-Oncogênicas/genética , Timina DNA Glicosilase/genética
11.
Nucleic Acids Res ; 43(8): 3986-97, 2015 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-25845601

RESUMO

Growth arrest and DNA-damage-inducible protein 45 (Gadd45) family members have been implicated in DNA demethylation in vertebrates. However, it remained unclear how they contribute to the demethylation process. Here, we demonstrate that Gadd45a promotes active DNA demethylation through thymine DNA glycosylase (TDG) which has recently been shown to excise 5-formylcytosine (5fC) and 5-carboxylcytosine (5caC) generated in Ten-eleven-translocation (Tet)-initiated oxidative demethylation. The connection of Gadd45a with oxidative demethylation is evidenced by the enhanced activation of a methylated reporter gene in HEK293T cells expressing Gadd45a in combination with catalytically active TDG and Tet. Gadd45a interacts with TDG physically and increases the removal of 5fC and 5caC from genomic and transfected plasmid DNA by TDG. Knockout of both Gadd45a and Gadd45b from mouse ES cells leads to hypermethylation of specific genomic loci most of which are also targets of TDG and show 5fC enrichment in TDG-deficient cells. These observations indicate that the demethylation effect of Gadd45a is mediated by TDG activity. This finding thus unites Gadd45a with the recently defined Tet-initiated demethylation pathway.


Assuntos
Proteínas de Ciclo Celular/fisiologia , Proteínas Nucleares/fisiologia , Timina DNA Glicosilase/metabolismo , Animais , Proteínas de Ciclo Celular/genética , Citosina/análogos & derivados , Citosina/metabolismo , Metilação de DNA , Proteínas de Ligação a DNA/metabolismo , Dioxigenases , Células-Tronco Embrionárias/metabolismo , Células HEK293 , Humanos , Camundongos Knockout , Proteínas Nucleares/genética , Proteínas Proto-Oncogênicas/metabolismo , Ativação Transcricional
12.
J Natl Cancer Inst ; 106(7)2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24973978

RESUMO

BACKGROUND: Aberrant DNA methylation in gene promoters is associated with aging and cancer, but the circumstances determining methylation change are unknown. We investigated the impact of lifestyle modulators of colorectal cancer (CRC) risk on the stability of gene promoter methylation in the colonic mucosa. METHODS: We measured genome-wide promoter CpG methylation in normal colon biopsies (n = 1092) from a female screening cohort, investigated the interaction of lifestyle factors with age-dependent increase in methylation with log-linear multivariable regression, and related their modifying effect to hypermethylation in CRC. All statistical tests were two-sided. RESULTS: Of 20025 promoter-associated CpGs analyzed, 1713 showed statistically significant age-dependent methylation gains. Fewer CpGs acquired methylation in users of aspirin (≥ 2 years) and hormonal replacement therapy (HRT age ≥ 50 years) compared with nonusers (43 vs 1355; 1 vs1377, respectively), whereas more CpGs were affected in smokers (≥ 20 years) and individuals with a body mass index (BMI) of 25 kg/m(2) and greater compared with control groups (180 vs 39; 554 vs 144, respectively). Fifty percent of the CpGs showing age-dependent methylation were found hypermethylated in CRC (odds ratio [OR] = 20; 95% confidence interval [CI] = 18 to 23; P < 2 × 10(-16)). These loci gained methylation with a higher median rate compared with age-only methylated sites (P = 2 × 10(-76)) and were enriched for polycomb regions (OR = 3.67). Importantly, aspirin (P < .001) and HRT use (P < .001) reduced the methylation rate at these cancer-related genes, whereas smoking (P < .001) and high BMI (P = .004) increased it. CONCLUSIONS: Lifestyle, including aspirin use, modulates age-associated DNA methylation change in the colonic epithelium and thereby impacts the evolution of cancer methylomes.


Assuntos
Anti-Inflamatórios não Esteroides/administração & dosagem , Aspirina/administração & dosagem , Colo/metabolismo , Neoplasias Colorretais/prevenção & controle , Metilação de DNA , Estilo de Vida , Fatores Etários , Idoso , Estudos de Coortes , Neoplasias Colorretais/metabolismo , Ilhas de CpG/genética , Detecção Precoce de Câncer/métodos , Feminino , Estudo de Associação Genômica Ampla , Humanos , Programas de Rastreamento/métodos , Pessoa de Meia-Idade , Regiões Promotoras Genéticas
13.
Nat Chem Biol ; 10(7): 574-81, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24838012

RESUMO

Ten eleven translocation (Tet) enzymes oxidize the epigenetically important DNA base 5-methylcytosine (mC) stepwise to 5-hydroxymethylcytosine (hmC), 5-formylcytosine and 5-carboxycytosine. It is currently unknown whether Tet-induced oxidation is limited to cytosine-derived nucleobases or whether other nucleobases are oxidized as well. We synthesized isotopologs of all major oxidized pyrimidine and purine bases and performed quantitative MS to show that Tet-induced oxidation is not limited to mC but that thymine is also a substrate that gives 5-hydroxymethyluracil (hmU) in mouse embryonic stem cells (mESCs). Using MS-based isotope tracing, we show that deamination of hmC does not contribute to the steady-state levels of hmU in mESCs. Protein pull-down experiments in combination with peptide tracing identifies hmU as a base that influences binding of chromatin remodeling proteins and transcription factors, suggesting that hmU has a specific function in stem cells besides triggering DNA repair.


Assuntos
Proteínas de Ligação a DNA/metabolismo , DNA/metabolismo , Células-Tronco Embrionárias/metabolismo , Pentoxil (Uracila)/análogos & derivados , Proteínas Proto-Oncogênicas/metabolismo , Timina/metabolismo , 5-Metilcitosina/análogos & derivados , Animais , Sequência de Bases , Isótopos de Carbono , Montagem e Desmontagem da Cromatina , Cromatografia Líquida , Citosina/análogos & derivados , Citosina/metabolismo , Proteínas de Ligação a DNA/genética , Dioxigenases , Células-Tronco Embrionárias/citologia , Expressão Gênica , Camundongos , Dados de Sequência Molecular , Oxirredução , Pentoxil (Uracila)/metabolismo , Ligação Proteica , Proteínas Proto-Oncogênicas/genética , Espectrometria de Massas por Ionização por Electrospray , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
14.
Prog Drug Res ; 67: 51-68, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21141724

RESUMO

The successful establishment and stable maintenance of cell identity are critical for organismal development and tissue homeostasis. Cell identity is provided by epigenetic mechanisms that facilitate a selective readout of the genome. Operating at the level of chromatin, they establish defined gene expression programs during cell differentiation. Among the epigenetic modifications in mammalian chromatin, the 5'-methylation of cytosine in CpG dinucleotides is unique in that it affects the DNA rather than histones and the biochemistry of the DNA methylating enzymes offers a mechanistic explanation for stable inheritance. Yet, DNA methylation states appear to be more dynamic and their maintenance more complex than existing models predict. Also, methylation patterns are by far not always faithfully inherited, as best exemplified by human cancers. Often, these show widespread hypo- or hypermethylation across their genomes, reflecting an underlying epigenetic instability that may have contributed to carcinogenesis. The phenotype of unstable methylation in cancer illustrates the importance of quality control in the DNA methylation system and implies the existence of proof-reading mechanisms that enforce fidelity to DNA methylation in healthy tissue. Fidelity seems particularly important in islands of unmethylated CpG-rich sequences where an accurate maintenance of un- or differentially methylated states is critical for stable expression of nearby genes. Methylation proof-reading in such sequences requires a system capable of recognition and active demethylation of erroneously methylated CpGs. Active demethylation of 5-methylcytosine has been known to occur for long, but the underlying mechanisms have remained enigmatic and controversial. However, recent progress in this direction substantiates a role of DNA repair in such processes. This review will address general aspects of cytosine methylation stability in mammalian DNA and explore a putative role of DNA repair in methylation control.


Assuntos
Metilação de DNA , Reparo do DNA , Animais , Citosina/metabolismo , Instabilidade Genômica , Humanos
15.
FASEB J ; 23(11): 3978-89, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19622798

RESUMO

Poly(ADP-ribose) polymerase 1 (PARP1) is a chromatin-associated nuclear protein and functions as a molecular stress sensor. At the cellular level, PARP1 has been implicated in a wide range of processes, such as maintenance of genome stability, cell death, and transcription. PARP1 functions as a transcriptional coactivator of nuclear factor kappaB (NF-kappaB) and hypoxia inducible factor 1 (HIF1). In proteomic studies, PARP1 was found to be modified by small ubiquitin-like modifiers (SUMOs). Here, we characterize PARP1 as a substrate for modification by SUMO1 and SUMO3, both in vitro and in vivo. PARP1 is sumoylated at the single lysine residue K486 within its automodification domain. Interestingly, modification of PARP1 with SUMO does not affect its ADP-ribosylation activity but completely abrogates p300-mediated acetylation of PARP1, revealing an intriguing crosstalk of sumoylation and acetylation on PARP1. Genetic complementation of PARP1-depleted cells with wild-type and sumoylation-deficient PARP1 revealed that SUMO modification of PARP1 restrains its transcriptional coactivator function and subsequently reduces gene expression of distinct PARP1-regulated target genes.


Assuntos
Poli(ADP-Ribose) Polimerases/metabolismo , Proteína SUMO-1/metabolismo , Acetilação , Acilação , Sequência de Aminoácidos , Cisteína Endopeptidases/metabolismo , Proteína p300 Associada a E1A/metabolismo , Endopeptidases/metabolismo , Humanos , Células K562 , Lisina/metabolismo , Poli(ADP-Ribose) Polimerases/genética , Ativação Transcricional/fisiologia , Ubiquitinas/metabolismo
16.
PLoS Biol ; 7(4): e91, 2009 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-19402749

RESUMO

5-Fluorouracil (5-FU), a chemotherapeutic drug commonly used in cancer treatment, imbalances nucleotide pools, thereby favoring misincorporation of uracil and 5-FU into genomic DNA. The processing of these bases by DNA repair activities was proposed to cause DNA-directed cytotoxicity, but the underlying mechanisms have not been resolved. In this study, we investigated a possible role of thymine DNA glycosylase (TDG), one of four mammalian uracil DNA glycosylases (UDGs), in the cellular response to 5-FU. Using genetic and biochemical tools, we found that inactivation of TDG significantly increases resistance of both mouse and human cancer cells towards 5-FU. We show that excision of DNA-incorporated 5-FU by TDG generates persistent DNA strand breaks, delays S-phase progression, and activates DNA damage signaling, and that the repair of 5-FU-induced DNA strand breaks is more efficient in the absence of TDG. Hence, excision of 5-FU by TDG, but not by other UDGs (UNG2 and SMUG1), prevents efficient downstream processing of the repair intermediate, thereby mediating DNA-directed cytotoxicity. The status of TDG expression in a cancer is therefore likely to determine its response to 5-FU-based chemotherapy.


Assuntos
Antimetabólitos Antineoplásicos/farmacologia , Morte Celular/efeitos dos fármacos , Dano ao DNA , Reparo do DNA/efeitos dos fármacos , Fluoruracila/farmacologia , Neoplasias/tratamento farmacológico , Timina DNA Glicosilase/metabolismo , Animais , Antimetabólitos Antineoplásicos/uso terapêutico , Ciclo Celular/genética , Linhagem Celular Tumoral , DNA Glicosilases/metabolismo , Fluoruracila/uso terapêutico , Camundongos , Neoplasias/genética , Transdução de Sinais , Uracila-DNA Glicosidase/metabolismo
17.
Oncol Rep ; 17(6): 1421-7, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17487400

RESUMO

Epigenetic alterations have been reported in colorectal neoplasia which can either complement or in some cases be predisposed to genetic alterations such as K-ras mutations. We examined the promoter methylation status of the CDKN2A and O6-methylguanine-DNA methyltransferase (MGMT) genes, after sodium bisulfite conversion and DNA amplification with methylation specific PCR. Moreover, we searched for G to A transitions in codons 12 and 13 of the K-ras oncogene in normal colorectal mucosae, aberrant crypt foci (ACF, early premalignant lesions) and carcinomas. CDKN2A hypermethylation was an infrequent event in ACF (2 of 26, 7.7%). On the contrary, MGMT hypermethylation was found in the normal mucosae (3 of the 12 samples, 25%), in 14 of the 26 ACF (53.8%) and in 7 of the 9 (77.8%) carcinomas examined. K-ras mutations were evident in 6 ACF (23%) and in 3 carcinomas (33.3%), mostly associated with MGMT promoter hypermethylation. These findings strongly support the hypothesis that epigenetic mechanisms play an important role in the early steps of colorectal carcinogenesis.


Assuntos
Transformação Celular Neoplásica/genética , Neoplasias Colorretais/genética , Epigênese Genética , Regulação Neoplásica da Expressão Gênica , O(6)-Metilguanina-DNA Metiltransferase/genética , Idoso , Idoso de 80 Anos ou mais , Carcinoma/genética , Inibidor p16 de Quinase Dependente de Ciclina/genética , Metilação de DNA , Feminino , Genes ras/genética , Humanos , Mucosa Intestinal/metabolismo , Masculino , Pessoa de Meia-Idade , Mutação , Regiões Promotoras Genéticas , Análise de Sequência de DNA , Sulfitos/química
18.
Nucleic Acids Res ; 35(7): 2321-32, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17389648

RESUMO

The ligation of DNA double-strand breaks in the process of non-homologous end-joining (NHEJ) is accomplished by a heterodimeric enzyme complex consisting of DNA ligase IV and an associated non-catalytic factor. This DNA ligase also accounts for the fatal joining of unprotected telomere ends. Hence, its activity must be tightly controlled. Here, we describe interactions of the DNA ligase IV-associated proteins Lif1p and XRCC4 of yeast and human with the putatively orthologous G-patch proteins Ntr1p/Spp382p and NTR1/TFIP11 that have recently been implicated in mRNA splicing. These conserved interactions occupy the DNA ligase IV-binding sites of Lif1p and XRCC4, thus preventing the formation of an active enzyme complex. Consistently, an excess of Ntr1p in yeast reduces NHEJ efficiency in a plasmid ligation assay as well as in a chromosomal double-strand break repair (DSBR) assay. Both yeast and human NTR1 also interact with PinX1, another G-patch protein that has dual functions in the regulation of telomerase activity and telomere stability, and in RNA processing. Like PinX1, NTR1 localizes to telomeres and associates with nucleoli in yeast and human cells, suggesting a function in localized control of DSBR.


Assuntos
Quebras de DNA de Cadeia Dupla , Reparo do DNA , Proteínas de Saccharomyces cerevisiae/metabolismo , Telômero/metabolismo , Proteínas de Ciclo Celular , Proteínas de Ligação a DNA/metabolismo , Células HeLa , Humanos , Proteínas Nucleares/análise , Proteínas Nucleares/metabolismo , Fatores de Processamento de RNA , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/fisiologia , Homologia de Sequência de Aminoácidos , Proteínas de Ligação a Telômeros/análise , Proteínas Supressoras de Tumor/metabolismo , Técnicas do Sistema de Duplo-Híbrido
19.
Mol Cell ; 22(1): 51-62, 2006 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-16600869

RESUMO

Alterations in DNA repair lead to genomic instability and higher risk of cancer. DNA base excision repair (BER) corrects damaged bases, apurinic sites, and single-strand DNA breaks. Here, a regulatory mechanism for DNA polymerase beta (Pol beta) is described. Pol beta was found to form a complex with the protein arginine methyltransferase 6 (PRMT6) and was specifically methylated in vitro and in vivo. Methylation of Pol beta by PRMT6 strongly stimulated DNA polymerase activity by enhancing DNA binding and processivity, while single nucleotide insertion and dRP-lyase activity were not affected. Two residues, R83 and R152, were identified in Pol beta as the sites of methylation by PRMT6. Genetic complementation of Pol beta knockout cells with R83/152K mutant revealed the importance of these residues for the cellular resistance to DNA alkylating agent. Based on our findings, we propose that PRMT6 plays a role as a regulator of BER.


Assuntos
Arginina/metabolismo , Metilação de DNA , DNA Polimerase beta/metabolismo , Regulação da Expressão Gênica , Proteínas Nucleares/fisiologia , Proteína-Arginina N-Metiltransferases/fisiologia , Animais , Arginina/química , Dano ao DNA , DNA Ligases/fisiologia , DNA Polimerase beta/genética , Embrião de Mamíferos/citologia , Embrião de Mamíferos/enzimologia , Fibroblastos/citologia , Fibroblastos/enzimologia , Teste de Complementação Genética , Humanos , Camundongos , Camundongos Knockout
20.
Nucleic Acids Res ; 33(19): 6393-404, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16282588

RESUMO

Gene activation involves protein complexes with diverse enzymatic activities, some of which are involved in chromatin modification. We have shown previously that the base excision repair enzyme thymine DNA glycosylase (TDG) acts as a potent coactivator for estrogen receptor-alpha. To further understand how TDG acts in this context, we studied its interaction with known coactivators of nuclear receptors. We find that TDG interacts in vitro and in vivo with the p160 coactivator SRC1, with the interaction being mediated by a previously undescribed motif encoding four equally spaced tyrosine residues in TDG, each tyrosine being separated by three amino acids. This is found to interact with two motifs in SRC1 also containing tyrosine residues separated by three amino acids. Site-directed mutagenesis shows that the tyrosines encoded in these motifs are critical for the interaction. The related p160 protein TIF2 does not interact with TDG and has the altered sequence, F-X-X-X-Y, at the equivalent positions relative to SRC1. Substitution of the phenylalanines to tyrosines is sufficient to bring about interaction of TIF2 with TDG. These findings highlight a new protein-protein interaction motif based on Y-X-X-X-Y and provide new insight into the interaction of diverse proteins in coactivator complexes.


Assuntos
Timina DNA Glicosilase/química , Timina DNA Glicosilase/metabolismo , Transativadores/metabolismo , Fatores de Transcrição/metabolismo , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Células COS , Chlorocebus aethiops , Histona Acetiltransferases , Humanos , Dados de Sequência Molecular , Coativador 1 de Receptor Nuclear , Sequências Repetitivas de Aminoácidos , Transativadores/química , Fatores de Transcrição/química , Tirosina/análise
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA