Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Mol Cell ; 81(6): 1260-1275.e12, 2021 03 18.
Artigo em Inglês | MEDLINE | ID: mdl-33561390

RESUMO

DNA methylation is implicated in neuronal biology via the protein MeCP2, the mutation of which causes Rett syndrome. MeCP2 recruits the NCOR1/2 co-repressor complexes to methylated cytosine in the CG dinucleotide, but also to sites of non-CG methylation, which are abundant in neurons. To test the biological significance of the dual-binding specificity of MeCP2, we replaced its DNA binding domain with an orthologous domain from MBD2, which can only bind mCG motifs. Knockin mice expressing the domain-swap protein displayed severe Rett-syndrome-like phenotypes, indicating that normal brain function requires the interaction of MeCP2 with sites of non-CG methylation, specifically mCAC. The results support the notion that the delayed onset of Rett syndrome is due to the simultaneous post-natal accumulation of mCAC and its reader MeCP2. Intriguingly, genes dysregulated in both Mecp2 null and domain-swap mice are implicated in other neurological disorders, potentially highlighting targets of relevance to the Rett syndrome phenotype.


Assuntos
Metilação de DNA , Proteína 2 de Ligação a Metil-CpG/metabolismo , Neurônios/metabolismo , Animais , Ilhas de CpG , Técnicas de Introdução de Genes , Células HeLa , Humanos , Masculino , Proteína 2 de Ligação a Metil-CpG/genética , Camundongos , Camundongos Transgênicos , Mutação , Células NIH 3T3 , Neurônios/patologia , Domínios Proteicos , Síndrome de Rett/genética , Síndrome de Rett/metabolismo , Síndrome de Rett/patologia
2.
Mol Cell ; 77(6): 1159-1161, 2020 03 19.
Artigo em Inglês | MEDLINE | ID: mdl-32200796

RESUMO

Distal regulatory elements control gene expression during differentiation. In this issue of Molecular Cell, Barnett et al. (2020) develop a new technology, called ATAC-Me, and discover that removal of DNA methylation is not a pre-requisite for the creation of accessible chromatin at active gene regulatory elements during cellular differentiation.


Assuntos
Cromatina , Metilação de DNA , Diferenciação Celular , Sequências Reguladoras de Ácido Nucleico
3.
Cell ; 151(7): 1417-30, 2012 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-23260135

RESUMO

The high level of 5-hydroxymethylcytosine (5hmC) present in neuronal genomes suggests that mechanisms interpreting 5hmC in the CNS may differ from those present in embryonic stem cells. Here, we present quantitative, genome-wide analysis of 5hmC, 5-methylcytosine (5mC), and gene expression in differentiated CNS cell types in vivo. We report that 5hmC is enriched in active genes and that, surprisingly, strong depletion of 5mC is observed over these regions. The contribution of these epigenetic marks to gene expression depends critically on cell type. We identify methyl-CpG-binding protein 2 (MeCP2) as the major 5hmC-binding protein in the brain and demonstrate that MeCP2 binds 5hmC- and 5mC-containing DNA with similar high affinities. The Rett-syndrome-causing mutation R133C preferentially inhibits 5hmC binding. These findings support a model in which 5hmC and MeCP2 constitute a cell-specific epigenetic mechanism for regulation of chromatin structure and gene expression.


Assuntos
Cerebelo/metabolismo , Citosina/análogos & derivados , Epigênese Genética , Proteína 2 de Ligação a Metil-CpG/metabolismo , 5-Metilcitosina/análogos & derivados , Animais , Cerebelo/citologia , Cromatina/metabolismo , Citosina/metabolismo , Humanos , Camundongos , Camundongos Knockout , Neuroglia/metabolismo , Neurônios/metabolismo , Células de Purkinje/metabolismo , Síndrome de Rett/metabolismo
4.
J Am Chem Soc ; 145(13): 7095-7100, 2023 04 05.
Artigo em Inglês | MEDLINE | ID: mdl-36961225

RESUMO

Selective, efficient, and controllable oxidation of cytosine modifications is valuable for epigenetic analyses, yet only limited progress has been made. Here, we present two modular chemical oxidation reactions: conversion of 5-hydroxymethylcytosine (5hmC) into 5-formylcytosine (5fC) using 4-acetamido-2,2,6,6-tetramethylpiperidine-1-oxoammonium tetrafluoroborate (ACT+BF4-) and further transformation of 5fC into 5-carboxycytosine (5caC) through Pinnick oxidation. Both reactions are mild and efficient on double-stranded DNA. We integrated these two oxidations with borane reduction to develop chemical-assisted pyridine borane sequencing plus (CAPS+), for direct and quantitative mapping of 5hmC. Compared with CAPS, CAPS+ improved the conversion rate and false-positive rate. We applied CAPS+ to mouse embryonic stem cells, human normal brain, and glioblastoma DNA samples and demonstrated its superior sensitivity in analyzing the hydroxymethylome.


Assuntos
Cistina , Cistina/análise , Humanos , Animais , Camundongos , Metilação de DNA , DNA/genética , Oxirredução
5.
Genome Res ; 30(10): 1393-1406, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32963030

RESUMO

Epigenetic modifications on chromatin play important roles in regulating gene expression. Although chromatin states are often governed by multilayered structure, how individual pathways contribute to gene expression remains poorly understood. For example, DNA methylation is known to regulate transcription factor binding but also to recruit methyl-CpG binding proteins that affect chromatin structure through the activity of histone deacetylase complexes (HDACs). Both of these mechanisms can potentially affect gene expression, but the importance of each, and whether these activities are integrated to achieve appropriate gene regulation, remains largely unknown. To address this important question, we measured gene expression, chromatin accessibility, and transcription factor occupancy in wild-type or DNA methylation-deficient mouse embryonic stem cells following HDAC inhibition. We observe widespread increases in chromatin accessibility at retrotransposons when HDACs are inhibited, and this is magnified when cells also lack DNA methylation. A subset of these elements has elevated binding of the YY1 and GABPA transcription factors and increased expression. The pronounced additive effect of HDAC inhibition in DNA methylation-deficient cells demonstrates that DNA methylation and histone deacetylation act largely independently to suppress transcription factor binding and gene expression.


Assuntos
Metilação de DNA , Epigênese Genética , Histona Desacetilases/metabolismo , Histonas/metabolismo , Fatores de Transcrição/metabolismo , Acetilação , Cromatina/metabolismo , Células-Tronco Embrionárias/efeitos dos fármacos , Células-Tronco Embrionárias/enzimologia , Células-Tronco Embrionárias/metabolismo , Genoma , Inibidores de Histona Desacetilases , Histona Desacetilases/farmacologia , Retroelementos
6.
Nucleic Acids Res ; 49(13): e76, 2021 07 21.
Artigo em Inglês | MEDLINE | ID: mdl-33905495

RESUMO

Whole genome base-resolution methylome sequencing allows for the most comprehensive analysis of DNA methylation, however, the considerable sequencing cost often limits its applications. While reduced representation sequencing can be an affordable alternative, over 80% of CpGs in the genome are not covered. Building on our recently developed TET-assisted pyridine borane sequencing (TAPS) method, we here described endonuclease enrichment TAPS (eeTAPS), which utilizes dihydrouracil (DHU)-cleaving endonuclease digestion of TAPS-converted DNA to enrich methylated CpG sites (mCpGs). eeTAPS can accurately detect 87% of mCpGs in the mouse genome with a sequencing depth equivalent to 4× whole genome sequencing. In comparison, reduced representation TAPS (rrTAPS) detected less than 4% of mCpGs with 2.5× sequencing depth. Our results demonstrate eeTAPS to be a new strategy for cost-effective genome-wide methylation analysis at single-CpG resolution that can fill the gap between whole-genome and reduced representation sequencing.


Assuntos
Metilação de DNA , Análise de Sequência de DNA/métodos , Animais , Células Cultivadas , Análise Custo-Benefício , Ilhas de CpG , Desoxirribonuclease (Dímero de Pirimidina) , Células-Tronco Embrionárias/metabolismo , Genômica/métodos , Camundongos , Análise de Sequência de DNA/economia , Uracila-DNA Glicosidase
7.
Nat Methods ; 16(5): 429-436, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-31011185

RESUMO

Replication of eukaryotic genomes is highly stochastic, making it difficult to determine the replication dynamics of individual molecules with existing methods. We report a sequencing method for the measurement of replication fork movement on single molecules by detecting nucleotide analog signal currents on extremely long nanopore traces (D-NAscent). Using this method, we detect 5-bromodeoxyuridine (BrdU) incorporated by Saccharomyces cerevisiae to reveal, at a genomic scale and on single molecules, the DNA sequences replicated during a pulse-labeling period. Under conditions of limiting BrdU concentration, D-NAscent detects the differences in BrdU incorporation frequency across individual molecules to reveal the location of active replication origins, fork direction, termination sites, and fork pausing/stalling events. We used sequencing reads of 20-160 kilobases to generate a whole-genome single-molecule map of DNA replication dynamics and discover a class of low-frequency stochastic origins in budding yeast. The D-NAscent software is available at https://github.com/MBoemo/DNAscent.git .


Assuntos
Replicação do DNA , Genoma Fúngico , Genômica/métodos , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Nanoporos , Saccharomyces cerevisiae/genética , Bromodesoxiuridina/metabolismo , DNA Fúngico/genética , Genoma , Software
8.
Nature ; 524(7563): 114-8, 2015 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-26200337

RESUMO

Cells require nucleotides to support DNA replication and repair damaged DNA. In addition to de novo synthesis, cells recycle nucleotides from the DNA of dying cells or from cellular material ingested through the diet. Salvaged nucleosides come with the complication that they can contain epigenetic modifications. Because epigenetic inheritance of DNA methylation mainly relies on copying of the modification pattern from parental strands, random incorporation of pre-modified bases during replication could have profound implications for epigenome fidelity and yield adverse cellular phenotypes. Although the salvage mechanism of 5-methyl-2'deoxycytidine (5mdC) has been investigated before, it remains unknown how cells deal with the recently identified oxidized forms of 5mdC: 5-hydroxymethyl-2'deoxycytidine (5hmdC), 5-formy-2'deoxycytidine (5fdC) and 5-carboxyl-2'deoxycytidine (5cadC). Here we show that enzymes of the nucleotide salvage pathway display substrate selectivity, effectively protecting newly synthesized DNA from the incorporation of epigenetically modified forms of cytosine. Thus, cell lines and animals can tolerate high doses of these modified cytidines without any deleterious effects on physiology. Notably, by screening cancer cell lines for growth defects after exposure to 5hmdC, we unexpectedly identify a subset of cell lines in which 5hmdC or 5fdC administration leads to cell lethality. Using genomic approaches, we show that the susceptible cell lines overexpress cytidine deaminase (CDA). CDA converts 5hmdC and 5fdC into variants of uridine that are incorporated into DNA, resulting in accumulation of DNA damage, and ultimately, cell death. Our observations extend current knowledge of the nucleotide salvage pathway by revealing the metabolism of oxidized epigenetic bases, and suggest a new therapeutic option for cancers, such as pancreatic cancer, that have CDA overexpression and are resistant to treatment with other cytidine analogues.


Assuntos
Citidina Desaminase/metabolismo , Citidina/análogos & derivados , Citidina/metabolismo , Citosina/metabolismo , Citosina/farmacologia , Epigênese Genética , Neoplasias/tratamento farmacológico , 5-Metilcitosina/metabolismo , 5-Metilcitosina/farmacologia , Animais , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Citidina/química , Citidina/farmacologia , Citidina Desaminase/genética , Citosina/análogos & derivados , Citosina/química , DNA/biossíntese , DNA/química , Dano ao DNA/efeitos dos fármacos , DNA Polimerase Dirigida por DNA/metabolismo , Desoxicitidina/análogos & derivados , Desoxicitidina/metabolismo , Desoxicitidina/farmacologia , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Camundongos , Neoplasias/genética , Neoplasias/metabolismo , Nucleotídeos/química , Nucleotídeos/metabolismo , Nucleotídeos/farmacologia , Oxirredução , Fosfotransferases/metabolismo , Especificidade por Substrato , Regulação para Cima , Uridina/análogos & derivados , Uridina/química , Uridina/metabolismo
9.
Mol Cell ; 49(6): 1023-33, 2013 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-23453809

RESUMO

Genomic imprinting directs the allele-specific marking and expression of loci according to their parental origin. Differential DNA methylation at imprinted control regions (ICRs) is established in gametes and, although largely preserved through development, can be experimentally reset by fusing somatic cells with embryonic germ cell (EGC) lines. Here, we show that the Ten-Eleven Translocation proteins Tet1 and Tet2 participate in the efficient erasure of imprints in this model system. The fusion of B cells with EGCs initiates pluripotent reprogramming, in which rapid re-expression of Oct4 is accompanied by an accumulation of 5-hydroxymethylcytosine (5hmC) at several ICRs. Tet2 was required for the efficient reprogramming capacity of EGCs, whereas Tet1 was necessary to induce 5-methylcytosine oxidation specifically at ICRs. These data show that the Tet1 and Tet2 proteins have discrete roles in cell-fusion-mediated pluripotent reprogramming and imprint erasure in somatic cells.


Assuntos
Fusão Celular , Proteínas de Ligação a DNA/fisiologia , Impressão Genômica , Proteínas Proto-Oncogênicas/fisiologia , 5-Metilcitosina/análogos & derivados , Animais , Linfócitos B/citologia , Sequência de Bases , Linhagem Celular , Citosina/análogos & derivados , Citosina/metabolismo , Metilação de DNA , Dioxigenases , Células-Tronco Embrionárias/citologia , Expressão Gênica , Células Germinativas/citologia , Proteínas de Fluorescência Verde/biossíntese , Humanos , Fator de Crescimento Insulin-Like II/genética , Camundongos , Dados de Sequência Molecular , Fator 3 de Transcrição de Octâmero/genética , Fator 3 de Transcrição de Octâmero/metabolismo , Polimorfismo de Nucleotídeo Único , Proteínas/genética , Proteínas/metabolismo , RNA Longo não Codificante/genética , Análise de Sequência de DNA
10.
PLoS Genet ; 14(10): e1007643, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30335751

RESUMO

The Fanconi Anemia (FA) pathway is important for repairing interstrand crosslinks (ICLs) between the Watson-Crick strands of the DNA double helix. An initial and essential stage in the repair process is the detection of the ICL. Here, we report the identification of UHRF2, a paralogue of UHRF1, as an ICL sensor protein. UHRF2 is recruited to ICLs in the genome within seconds of their appearance. We show that UHRF2 cooperates with UHRF1, to ensure recruitment of FANCD2 to ICLs. A direct protein-protein interaction is formed between UHRF1 and UHRF2, and between either UHRF1 and UHRF2, and FANCD2. Importantly, we demonstrate that the essential monoubiquitination of FANCD2 is stimulated by UHRF1/UHRF2. The stimulation is mediating by a retention of FANCD2 on chromatin, allowing for its monoubiquitination by the FA core complex. Taken together, we uncover a mechanism of ICL sensing by UHRF2, leading to FANCD2 recruitment and retention at ICLs, in turn facilitating activation of FANCD2 by monoubiquitination.


Assuntos
Reparo do DNA/fisiologia , Proteína do Grupo de Complementação D2 da Anemia de Fanconi/fisiologia , Ubiquitina-Proteína Ligases/fisiologia , Sequência de Aminoácidos , Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Proteínas Estimuladoras de Ligação a CCAAT/fisiologia , Linhagem Celular , Núcleo Celular/metabolismo , Cromatina/metabolismo , DNA/metabolismo , Dano ao DNA/fisiologia , Anemia de Fanconi/genética , Proteína do Grupo de Complementação D2 da Anemia de Fanconi/metabolismo , Proteínas de Grupos de Complementação da Anemia de Fanconi/genética , Células HEK293 , Células HeLa , Humanos , Domínios e Motivos de Interação entre Proteínas , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação
11.
Br J Cancer ; 118(5): 727-732, 2018 03 06.
Artigo em Inglês | MEDLINE | ID: mdl-29438375

RESUMO

BACKGROUND: Colorectal cancer (CRC) screening might be improved by using a measure of prior risk to modulate screening intensity or the faecal immunochemical test threshold. Intermediate molecular biomarkers could aid risk prediction by capturing both known and unknown risk factors. METHODS: We sampled normal bowel mucosa from the proximal colon, distal colon and rectum of 317 individuals undergoing colonoscopy. We defined cases as having a personal history of colorectal polyp(s)/cancer, and controls as having no history of colorectal neoplasia. Molecular analyses were performed for: telomere length (TL); global methylation; and the expression of genes in molecular pathways associated with colorectal tumourigenesis. We also calculated a polygenic risk score (PRS) based on CRC susceptibility polymorphisms. RESULTS: Bowel TL was significantly longer in cases than controls, but was not associated with blood TL. PRS was significantly and independently higher in cases. Hypermethylation showed a suggestive association with case:control status. No gene or pathway was differentially expressed between cases and controls. Gene expression often varied considerably between bowel locations. CONCLUSIONS: PRS and bowel TL (but not blood TL) may be clinically-useful predictors of CRC risk. Sample collection to assess these biomarkers is feasible in clinical practice, especially where population screening uses flexible sigmoidoscopy or colonoscopy.


Assuntos
Biomarcadores Tumorais/genética , Colo/química , Neoplasias Colorretais/diagnóstico , Reto/química , Homeostase do Telômero , Adulto , Idoso , Idoso de 80 Anos ou mais , Estudos de Casos e Controles , Colonoscopia , Neoplasias Colorretais/genética , Metilação de DNA , Detecção Precoce de Câncer , Feminino , Redes Reguladoras de Genes , Humanos , Masculino , Pessoa de Meia-Idade , Polimorfismo de Nucleotídeo Único , Fatores de Risco , Adulto Jovem
12.
Br J Cancer ; 118(12): 1683, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29780161

RESUMO

Since the publication of this paper, the authors noticed that James E. East was assigned to the incorrect affiliation. The affiliation information is provided correctly, above.

13.
Nat Chem Biol ; 16(6): 604-605, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32300239
14.
Nucleic Acids Res ; 40(4): e32, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22156374

RESUMO

Across vertebrate genomes methylation of cytosine residues within the context of CpG dinucleotides is a pervasive epigenetic mark that can impact gene expression and has been implicated in various developmental and disease-associated processes. Several biochemical approaches exist to profile DNA methylation, but recently an alternative approach based on profiling non-methylated CpGs was developed. This technique, called CxxC affinity purification (CAP), uses a ZF-CxxC (CxxC) domain to specifically capture DNA containing clusters of non-methylated CpGs. Here we describe a new CAP approach, called biotinylated CAP (Bio-CAP), which eliminates the requirement for specialized equipment while dramatically improving and simplifying the CxxC-based DNA affinity purification. Importantly, this approach isolates non-methylated DNA in a manner that is directly proportional to the density of non-methylated CpGs, and discriminates non-methylated CpGs from both methylated and hydroxymethylated CpGs. Unlike conventional CAP, Bio-CAP can be applied to nanogram quantities of genomic DNA and in a magnetic format is amenable to efficient parallel processing of samples. Furthermore, Bio-CAP can be applied to genome-wide profiling of non-methylated DNA with relatively small amounts of input material. Therefore, Bio-CAP is a simple and streamlined approach for characterizing regions of the non-methylated DNA, whether at specific target regions or genome wide.


Assuntos
Cromatografia de Afinidade/métodos , Ilhas de CpG , Metilação de DNA , Biotinilação , DNA/isolamento & purificação , Sequenciamento de Nucleotídeos em Larga Escala , Análise de Sequência de DNA , Dedos de Zinco
15.
J Med Chem ; 67(6): 4525-4540, 2024 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-38294854

RESUMO

Ten-eleven translocation enzymes (TETs) are Fe(II)/2-oxoglutarate (2OG) oxygenases that catalyze the sequential oxidation of 5-methylcytosine to 5-hydroxymethylcytosine, 5-formylcytosine, and 5-carboxylcytosine in eukaryotic DNA. Despite their roles in epigenetic regulation, there is a lack of reported TET inhibitors. The extent to which 2OG oxygenase inhibitors, including clinically used inhibitors and oncometabolites, modulate DNA modifications via TETs has been unclear. Here, we report studies on human TET1-3 inhibition by a set of 2OG oxygenase-focused inhibitors, employing both enzyme-based and cellular assays. Most inhibitors manifested similar potencies for TET1-3 and caused increases in cellular 5hmC levels. (R)-2-Hydroxyglutarate, an oncometabolite elevated in isocitrate dehydrogenase mutant cancer cells, showed different degrees of inhibition, with TET1 being less potently inhibited than TET3 and TET2, potentially reflecting the proposed role of TET2 mutations in tumorigenesis. The results highlight the tractability of TETs as drug targets and provide starting points for selective inhibitor design.


Assuntos
Dioxigenases , Glutaratos , Oxigenases , Humanos , Epigênese Genética , Oxigenases de Função Mista , Dioxigenases/metabolismo , DNA , Metilação de DNA , Proteínas Proto-Oncogênicas/metabolismo
16.
Life Sci Alliance ; 5(12)2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36122935

RESUMO

The DNA-binding protein MeCP2 is reported to bind methylated cytosine in CG and CA motifs in genomic DNA, but it was recently proposed that arrays of tandemly repeated CA containing either methylated or hydroxymethylated cytosine are the primary targets for MeCP2 binding and function. Here we investigated the predictions of this hypothesis using a range of published datasets. We failed to detect enrichment of cytosine modification at genomic CA repeat arrays in mouse brain regions and found no evidence for preferential MeCP2 binding at CA repeats. Moreover, we did not observe a correlation between the CA repeat density near genes and their degree of transcriptional deregulation when MeCP2 was absent. Our results do not provide support for the hypothesis that CA repeats are key mediators of MeCP2 function. Instead, we found that CA repeats are subject to CAC methylation to a degree that is typical of the surrounding genome and contribute modestly to MeCP2-mediated modulation of gene expression in accordance with their content of this canonical target motif.


Assuntos
Proteína 2 de Ligação a Metil-CpG , Animais , Citosina/metabolismo , DNA/metabolismo , Metilação de DNA , Proteína 2 de Ligação a Metil-CpG/metabolismo , Camundongos , Neurônios/metabolismo
17.
Neuro Oncol ; 24(12): 2093-2106, 2022 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-35468205

RESUMO

BACKGROUND: Glioblastoma (GBM) is the most common and malignant primary brain tumor in adults. Despite maximal treatment, median survival remains dismal at 14-24 months. Immunotherapies, such as checkpoint inhibition, have revolutionized management of some cancers but have little benefit for GBM patients. This is, in part, due to the low mutational and neoantigen burden in this immunogenically "cold" tumor. METHODS: U87MG and patient-derived cell lines were treated with 5-aza-2'-deoxycytidine (DAC) and underwent whole-exome and transcriptome sequencing. Cell lines were then subjected to cellular assays with neoantigen and cancer testis antigen (CTA) specific T cells. RESULTS: We demonstrate that DAC increases neoantigen and CTA mRNA expression through DNA hypomethylation. This results in increased neoantigen presentation by MHC class I in tumor cells, leading to increased neoantigen- and CTA-specific T-cell activation and killing of DAC-treated cancer cells. In addition, we show that patients have endogenous cancer-specific T cells in both tumor and blood, which show increased tumor-specific activation in the presence of DAC-treated cells. CONCLUSIONS: Our work shows that DAC increases GBM immunogenicity and consequent susceptibility to T-cell responses in vitro. Our results support a potential use of DAC as a sensitizing agent for immunotherapy.


Assuntos
Glioblastoma , Adulto , Masculino , Humanos , Glioblastoma/tratamento farmacológico , Glioblastoma/genética , Decitabina/farmacologia , Antígenos de Neoplasias/genética , Linfócitos T , Testículo , Linhagem Celular Tumoral
18.
Mol Cell Biol ; 26(13): 5033-42, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16782889

RESUMO

Rett syndrome (RTT) is a severe neurological disorder caused by mutations in the X-linked MECP2 gene, which encodes a methyl-CpG binding transcriptional repressor. Using the Mecp2-null mouse (an animal model for RTT) and differential display, we found that mice with neurological symptoms overexpress the nuclear gene for ubiquinol-cytochrome c reductase core protein 1 (Uqcrc1). Chromatin immunoprecipitation demonstrated that MeCP2 interacts with the Uqcrc1 promoter. Uqcrc1 encodes a subunit of mitochondrial respiratory complex III, and isolated mitochondria from the Mecp2-null brain showed elevated respiration rates associated with respiratory complex III and an overall reduction in coupling. A causal link between Uqcrc1 gene overexpression and enhanced complex III activity was established in neuroblastoma cells. Our findings raise the possibility that mitochondrial dysfunction contributes to pathology of the Mecp2-null mouse and may contribute to the long-known resemblance between Rett syndrome and certain mitochondrial disorders.


Assuntos
Complexo III da Cadeia de Transporte de Elétrons/genética , Proteína 2 de Ligação a Metil-CpG/metabolismo , Mitocôndrias/ultraestrutura , Doenças Mitocondriais/genética , Síndrome de Rett/genética , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Imunoprecipitação da Cromatina , Modelos Animais de Doenças , Regulação da Expressão Gênica , Proteína 2 de Ligação a Metil-CpG/genética , Camundongos , Camundongos Mutantes , Mitocôndrias/metabolismo , Doenças Mitocondriais/metabolismo , Doenças Mitocondriais/patologia , Regiões Promotoras Genéticas , Síndrome de Rett/metabolismo , Síndrome de Rett/patologia , Ativação Transcricional
19.
Sci Rep ; 9(1): 13463, 2019 09 17.
Artigo em Inglês | MEDLINE | ID: mdl-31530880

RESUMO

Expression of the mismatch repair gene MutL homolog 1 (MLH1) is silenced in a clinically important subgroup of sporadic colorectal cancers. These cancers exhibit hypermutability with microsatellite instability (MSI) and differ from microsatellite-stable (MSS) colorectal cancers in both prognosis and response to therapies. Loss of MLH1 is usually due to epigenetic silencing with associated promoter methylation; coding somatic mutations rarely occur. Here we use the presence of a colorectal cancer (CRC) risk variant (rs1800734) within the MLH1 promoter to investigate the poorly understood mechanisms of MLH1 promoter methylation and loss of expression. We confirm the association of rs1800734 with MSI+ but not MSS cancer risk in our own data and by meta-analysis. Using sensitive allele-specific detection methods, we demonstrate that MLH1 is the target gene for rs1800734 mediated cancer risk. In normal colon tissue, small allele-specific differences exist only in MLH1 promoter methylation, but not gene expression. In contrast, allele-specific differences in both MLH1 methylation and expression are present in MSI+ cancers. We show that MLH1 transcriptional repression is dependent on DNA methylation and can be reversed by a methylation inhibitor. The rs1800734 allele influences the rate of methylation loss and amount of re-expression. The transcription factor TFAP4 binds to the rs1800734 region but with much weaker binding to the risk than the protective allele. TFAP4 binding is absent on both alleles when promoter methylation is present. Thus we propose that TFAP4 binding shields the protective rs1800734 allele of the MLH1 promoter from BRAF induced DNA methylation more effectively than the risk allele.


Assuntos
Neoplasias Colorretais/genética , Proteínas de Ligação a DNA/metabolismo , Proteína 1 Homóloga a MutL/genética , Polimorfismo de Nucleotídeo Único , Regiões Promotoras Genéticas , Fatores de Transcrição/metabolismo , Alelos , Estudos de Casos e Controles , Ilhas de CpG , Metilação de DNA , Proteínas de Ligação a DNA/genética , Bases de Dados Factuais , Regulação Neoplásica da Expressão Gênica , Predisposição Genética para Doença , Humanos , Instabilidade de Microssatélites , Proteína 1 Homóloga a MutL/metabolismo , RNA Mensageiro/genética , Fatores de Transcrição/genética
20.
Nat Commun ; 10(1): 30, 2019 01 03.
Artigo em Inglês | MEDLINE | ID: mdl-30604745

RESUMO

The inactive X chromosome (Xi) in female mammals adopts an atypical higher-order chromatin structure, manifested as a global loss of local topologically associated domains (TADs), A/B compartments and formation of two mega-domains. Here we demonstrate that the non-canonical SMC family protein, SmcHD1, which is important for gene silencing on Xi, contributes to this unique chromosome architecture. Specifically, allelic mapping of the transcriptome and epigenome in SmcHD1 mutant cells reveals the appearance of sub-megabase domains defined by gene activation, CpG hypermethylation and depletion of Polycomb-mediated H3K27me3. These domains, which correlate with sites of SmcHD1 enrichment on Xi in wild-type cells, additionally adopt features of active X chromosome higher-order chromosome architecture, including A/B compartments and partial restoration of TAD boundaries. Xi chromosome architecture changes also occurred following SmcHD1 knockout in a somatic cell model, but in this case, independent of Xi gene derepression. We conclude that SmcHD1 is a key factor in defining the unique chromosome architecture of Xi.


Assuntos
Proteínas Cromossômicas não Histona/genética , Metilação de DNA/genética , Ativação Transcricional/genética , Inativação do Cromossomo X , Alelos , Animais , Sistemas CRISPR-Cas , Linhagem Celular , Proteínas Cromossômicas não Histona/metabolismo , Ilhas de CpG , Éxons/genética , Feminino , Fibroblastos , Técnicas de Inativação de Genes , Histonas/genética , Histonas/metabolismo , Masculino , Camundongos , Mutação Puntual , Proteínas do Grupo Polycomb/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA