Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Methods Mol Biol ; 2529: 229-251, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35733018

RESUMO

Chromatin immunoprecipitation (ChIP) enables the study of DNA-protein interactions. When coupled with high-throughput sequencing (ChIP-seq), this method allows the generation of genome-wide profiles of the distribution of specific proteins in a given cellular context. Typical ChIP-seq experiments require millions of cells as input material and thus are not ideal to study many in vivo cell populations. Here, we describe an ultra-low-input native ChIP-seq method, ULI-NChIP-seq, to profile histone modification patterns in as low as 150 cells.


Assuntos
Sequenciamento de Nucleotídeos em Larga Escala , Histonas , Imunoprecipitação da Cromatina/métodos , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Histonas/genética , Histonas/metabolismo , Metilação , Processamento de Proteína Pós-Traducional , Análise de Sequência de DNA/métodos
2.
Nat Rev Genet ; 23(3): 137-153, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34608297

RESUMO

DNA methylation (DNAme) and histone post-translational modifications (PTMs) have important roles in transcriptional regulation. Although many reports have characterized the functions of such chromatin marks in isolation, recent genome-wide studies reveal surprisingly complex interactions between them. Here, we focus on the interplay between DNAme and methylation of specific lysine residues on the histone H3 tail. We describe the impact of genetic perturbation of the relevant methyltransferases in the mouse on the landscape of chromatin marks as well as the transcriptome. In addition, we discuss the specific neurodevelopmental growth syndromes and cancers resulting from pathogenic mutations in the human orthologues of these genes. Integrating these observations underscores the fundamental importance of crosstalk between DNA and histone H3 methylation in development and disease.


Assuntos
Cromatina/metabolismo , Metilação de DNA/genética , Doença/genética , Crescimento e Desenvolvimento/genética , Animais , Montagem e Desmontagem da Cromatina/fisiologia , Histonas/metabolismo , Humanos , Camundongos , Processamento de Proteína Pós-Traducional
3.
Nat Commun ; 12(1): 7020, 2021 12 02.
Artigo em Inglês | MEDLINE | ID: mdl-34857746

RESUMO

Silencing of a subset of germline genes is dependent upon DNA methylation (DNAme) post-implantation. However, these genes are generally hypomethylated in the blastocyst, implicating alternative repressive pathways before implantation. Indeed, in embryonic stem cells (ESCs), an overlapping set of genes, including germline "genome-defence" (GGD) genes, are upregulated following deletion of the H3K9 methyltransferase SETDB1 or subunits of the non-canonical PRC1 complex PRC1.6. Here, we show that in pre-implantation embryos and naïve ESCs (nESCs), hypomethylated promoters of germline genes bound by the PRC1.6 DNA-binding subunits MGA/MAX/E2F6 are enriched for RING1B-dependent H2AK119ub1 and H3K9me3. Accordingly, repression of these genes in nESCs shows a greater dependence on PRC1.6 than DNAme. In contrast, GGD genes are hypermethylated in epiblast-like cells (EpiLCs) and their silencing is dependent upon SETDB1, PRC1.6/RING1B and DNAme, with H3K9me3 and DNAme establishment dependent upon MGA binding. Thus, GGD genes are initially repressed by PRC1.6, with DNAme subsequently engaged in post-implantation embryos.


Assuntos
Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fator de Transcrição E2F6/genética , Regulação da Expressão Gênica no Desenvolvimento , Histona-Lisina N-Metiltransferase/genética , Histonas/genética , Proteínas do Grupo Polycomb/genética , Animais , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Metilação de DNA , Fator de Transcrição E2F6/metabolismo , Implantação do Embrião , Embrião de Mamíferos , Epigênese Genética , Feminino , Inativação Gênica , Genes Reporter , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Histona-Lisina N-Metiltransferase/metabolismo , Histonas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Células-Tronco Embrionárias Murinas/citologia , Células-Tronco Embrionárias Murinas/metabolismo , Complexo Repressor Polycomb 1/genética , Complexo Repressor Polycomb 1/metabolismo , Proteínas do Grupo Polycomb/metabolismo , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , Transdução de Sinais , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo
4.
Dev Cell ; 56(5): 671-686.e6, 2021 03 08.
Artigo em Inglês | MEDLINE | ID: mdl-33596408

RESUMO

A father's lifestyle impacts offspring health; yet, the underlying molecular mechanisms remain elusive. We hypothesized that a diet that changes methyl donor availability will alter the sperm and embryo epigenomes to impact embryonic gene expression and development. Here, we demonstrate that a folate-deficient (FD) diet alters histone H3 lysine 4 trimethylation (H3K4me3) in sperm at developmental genes and putative enhancers. A subset of H3K4me3 alterations in sperm are retained in the pre-implantation embryo and associated with deregulated embryonic gene expression. Using a genetic mouse model in which sires have pre-existing altered H3K4me2/3 in sperm, we show that a FD diet exacerbates alterations in sperm H3K4me3 and embryonic gene expression, leading to an increase in developmental defect severity. These findings imply that paternal H3K4me3 is transmitted to the embryo and influences gene expression and development. It further suggests that epigenetic errors can accumulate in sperm to worsen offspring developmental outcomes.


Assuntos
Anormalidades Congênitas/patologia , Metilação de DNA , Dieta , Embrião de Mamíferos/citologia , Regulação da Expressão Gênica no Desenvolvimento , Histonas/química , Espermatozoides/metabolismo , Animais , Animais Recém-Nascidos , Cromatina/química , Cromatina/genética , Anormalidades Congênitas/etiologia , Anormalidades Congênitas/metabolismo , Embrião de Mamíferos/metabolismo , Epigênese Genética , Feminino , Histonas/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fenótipo
5.
Nat Genet ; 52(10): 1088-1098, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32929285

RESUMO

De novo DNA methylation (DNAme) in mammalian germ cells is dependent on DNMT3A and DNMT3L. However, oocytes and spermatozoa show distinct patterns of DNAme. In mouse oocytes, de novo DNAme requires the lysine methyltransferase (KMTase) SETD2, which deposits H3K36me3. We show here that SETD2 is dispensable for de novo DNAme in the male germline. Instead, the lysine methyltransferase NSD1, which broadly deposits H3K36me2 in euchromatic regions, plays a critical role in de novo DNAme in prospermatogonia, including at imprinted genes. However, males deficient in germline NSD1 show a more severe defect in spermatogenesis than Dnmt3l-/- males. Notably, unlike DNMT3L, NSD1 safeguards a subset of genes against H3K27me3-associated transcriptional silencing. In contrast, H3K36me2 in oocytes is predominantly dependent on SETD2 and coincides with H3K36me3. Furthermore, females with NSD1-deficient oocytes are fertile. Thus, the sexually dimorphic pattern of DNAme in mature mouse gametes is orchestrated by distinct profiles of H3K36 methylation.


Assuntos
DNA (Citosina-5-)-Metiltransferases/genética , Histona-Lisina N-Metiltransferase/genética , Espermatogênese/genética , Animais , Metilação de DNA/genética , DNA Metiltransferase 3A , Feminino , Histona-Lisina N-Metiltransferase/deficiência , Histonas/genética , Humanos , Masculino , Camundongos , Camundongos Knockout , Proteínas Nucleares/genética , Oócitos/crescimento & desenvolvimento , Oócitos/metabolismo , Proteínas do Grupo Polycomb/genética , Espermatozoides/crescimento & desenvolvimento , Espermatozoides/metabolismo , Fatores de Transcrição/genética
6.
Viruses ; 12(7)2020 07 21.
Artigo em Inglês | MEDLINE | ID: mdl-32708087

RESUMO

Insertions of endogenous retroviruses cause a significant fraction of mutations in inbred mice but not all strains are equally susceptible. Notably, most new Intracisternal A particle (IAP) ERV mutagenic insertions have occurred in C3H mice. We show here that strain-specific insertional polymorphic IAPs accumulate faster in C3H/HeJ mice, relative to other sequenced strains, and that IAP transcript levels are higher in C3H/HeJ embryonic stem (ES) cells compared to other ES cells. To investigate the mechanism for high IAP activity in C3H mice, we identified 61 IAP copies in C3H/HeJ ES cells enriched with H3K4me3 (a mark of active promoters) and, among those tested, all are unmethylated in C3H/HeJ ES cells. Notably, 13 of the 61 are specific to C3H/HeJ and are members of the non-autonomous 1Δ1 IAP subfamily that is responsible for nearly all new insertions in C3H. One copy is full length with intact open reading frames and hence potentially capable of providing proteins in trans to other 1Δ1 elements. This potential "master copy" is present in other strains, including 129, but its 5' long terminal repeat (LTR) is methylated in 129 ES cells. Thus, the unusual IAP activity in C3H may be due to reduced epigenetic repression coupled with the presence of a master copy.


Assuntos
Epigenômica , Genes de Partícula A Intracisternal/genética , Genes de Partícula A Intracisternal/fisiologia , Camundongos Endogâmicos C3H/genética , Animais , Células Cultivadas , Células-Tronco Embrionárias , Metilação , Camundongos , Camundongos Endogâmicos C57BL/genética , Regiões Promotoras Genéticas , Especificidade da Espécie , Sequências Repetidas Terminais
7.
Nat Cell Biol ; 22(4): 355-357, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32231308
8.
Science ; 367(6480): 870-874, 2020 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-32079766

RESUMO

Diapause is a state of suspended development that helps organisms survive extreme environments. How diapause protects living organisms is largely unknown. Using the African turquoise killifish (Nothobranchius furzeri), we show that diapause preserves complex organisms for extremely long periods of time without trade-offs for subsequent adult growth, fertility, and life span. Transcriptome analyses indicate that diapause is an active state, with dynamic regulation of metabolism and organ development genes. The most up-regulated genes in diapause include Polycomb complex members. The chromatin mark regulated by Polycomb, H3K27me3, is maintained at key developmental genes in diapause, and the Polycomb member CBX7 mediates repression of metabolism and muscle genes in diapause. CBX7 is functionally required for muscle preservation and diapause maintenance. Thus, vertebrate diapause is a state of suspended life that is actively maintained by specific chromatin regulators, and this has implications for long-term organism preservation.


Assuntos
Diapausa/fisiologia , Peixes Listrados/crescimento & desenvolvimento , Músculo Esquelético/crescimento & desenvolvimento , Complexo Repressor Polycomb 1/metabolismo , Animais , Diapausa/genética , Regulação da Expressão Gênica no Desenvolvimento , Histonas/metabolismo , Mutação , Complexo Repressor Polycomb 1/genética
9.
Nat Commun ; 10(1): 5674, 2019 12 12.
Artigo em Inglês | MEDLINE | ID: mdl-31831741

RESUMO

Imprinted genes are expressed from a single parental allele, with the other allele often silenced by DNA methylation (DNAme) established in the germline. While species-specific imprinted orthologues have been documented, the molecular mechanisms underlying the evolutionary switch from biallelic to imprinted expression are unknown. During mouse oogenesis, gametic differentially methylated regions (gDMRs) acquire DNAme in a transcription-guided manner. Here we show that oocyte transcription initiating in lineage-specific endogenous retroviruses (ERVs) is likely responsible for DNAme establishment at 4/6 mouse-specific and 17/110 human-specific imprinted gDMRs. The latter are divided into Catarrhini- or Hominoidea-specific gDMRs embedded within transcripts initiating in ERVs specific to these primate lineages. Strikingly, imprinting of the maternally methylated genes Impact and Slc38a4 was lost in the offspring of female mice harboring deletions of the relevant murine-specific ERVs upstream of these genes. Our work reveals an evolutionary mechanism whereby maternally silenced genes arise from biallelically expressed progenitors.


Assuntos
Metilação de DNA , Evolução Molecular , Impressão Genômica , Regiões Promotoras Genéticas/genética , Retroviridae/genética , Animais , Epigenômica , Feminino , Células Germinativas , Haplorrinos , Humanos , Macaca , Masculino , Camundongos , Oócitos/metabolismo , Pan troglodytes , Primatas , Especificidade da Espécie , Sequências Repetidas Terminais
10.
Epigenetics Chromatin ; 12(1): 49, 2019 08 09.
Artigo em Inglês | MEDLINE | ID: mdl-31399135

RESUMO

BACKGROUND: KRAB zinc finger proteins (KZFPs) represent one of the largest families of DNA-binding proteins in vertebrate genomes and appear to have evolved to silence transposable elements (TEs) including endogenous retroviruses through sequence-specific targeting of repressive chromatin states. ZFP57 is required to maintain the post-fertilization DNA methylation memory of parental origin at genomic imprints. Here we conduct RNA-seq and ChIP-seq analyses in normal and ZFP57 mutant mouse ES cells to understand the relative importance of ZFP57 at imprints, unique and repetitive regions of the genome. RESULTS: Over 80% of ZFP57 targets are TEs, however, ZFP57 is not essential for their repression. The remaining targets lie within unique imprinted and non-imprinted sequences. Though the loss of ZFP57 influences imprinted genes as expected, the majority of unique gene targets lose H3K9me3 with little effect on DNA methylation and very few exhibit alterations in expression. Comparison of ZFP57 mutants with DNA methyltransferase-deleted ES cells (TKO) identifies a remarkably similar pattern of H3K9me3 loss across the genome. These data define regions where H3K9me3 is secondary to DNA methylation and we propose that ZFP57 is the principal if not sole methylation-sensitive KZFP in mouse ES cells. Finally, we examine dynamics of DNA and H3K9 methylation during pre-implantation development and show that sites bound by ZFP57 in ES cells maintain DNA methylation and H3K9me3 at imprints and at non-imprinted regions on the maternally inherited chromosome throughout preimplantation development. CONCLUSION: Our analyses suggest the evolution of a rare DNA methylation-sensitive KZFP that is not essential for repeat silencing, but whose primary function is to maintain DNA methylation and repressive histone marks at germline-derived imprinting control regions.


Assuntos
Elementos de DNA Transponíveis/genética , Impressão Genômica , Proteínas Repressoras/metabolismo , Animais , Sítios de Ligação , Linhagem Celular , Metilação de DNA , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Histonas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Ligação Proteica , Proteínas Repressoras/deficiência , Proteínas Repressoras/genética , Proteína 28 com Motivo Tripartido/metabolismo
11.
Nat Genet ; 51(5): 844-856, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-31040401

RESUMO

The oocyte epigenome plays critical roles in mammalian gametogenesis and embryogenesis. Yet, how it is established remains elusive. Here, we report that histone-lysine N-methyltransferase SETD2, an H3K36me3 methyltransferase, is a crucial regulator of the mouse oocyte epigenome. Deficiency in Setd2 leads to extensive alterations of the oocyte epigenome, including the loss of H3K36me3, failure in establishing the correct DNA methylome, invasion of H3K4me3 and H3K27me3 into former H3K36me3 territories and aberrant acquisition of H3K4me3 at imprinting control regions instead of DNA methylation. Importantly, maternal depletion of SETD2 results in oocyte maturation defects and subsequent one-cell arrest after fertilization. The preimplantation arrest is mainly due to a maternal cytosolic defect, since it can be largely rescued by normal oocyte cytosol. However, chromatin defects, including aberrant imprinting, persist in these embryos, leading to embryonic lethality after implantation. Thus, these data identify SETD2 as a crucial player in establishing the maternal epigenome that in turn controls embryonic development.


Assuntos
Desenvolvimento Embrionário/genética , Epigênese Genética , Impressão Genômica , Histona-Lisina N-Metiltransferase/genética , Histona-Lisina N-Metiltransferase/metabolismo , Animais , DNA (Citosina-5-)-Metiltransferases/deficiência , DNA (Citosina-5-)-Metiltransferases/genética , DNA (Citosina-5-)-Metiltransferases/metabolismo , Metilação de DNA , Feminino , Código das Histonas/genética , Histona-Lisina N-Metiltransferase/deficiência , Histonas/metabolismo , Masculino , Camundongos , Camundongos Knockout , Modelos Genéticos , Oócitos/metabolismo , Oogênese/genética , Gravidez
12.
Cell Rep ; 27(1): 282-293.e4, 2019 04 02.
Artigo em Inglês | MEDLINE | ID: mdl-30943408

RESUMO

Mammalian histone methyltransferase G9a (also called EHMT2) deposits H3K9me2 on chromatin and is essential for postimplantation development. However, its role in oogenesis and preimplantation development remains poorly understood. We show that H3K9me2-enriched chromatin domains in mouse oocytes are generally depleted of CG methylation, contrasting with their association in embryonic stem and somatic cells. Oocyte-specific disruption of G9a results in reduced H3K9me2 enrichment and impaired reorganization of heterochromatin in oocytes, but only a modest reduction in CG methylation is detected. Furthermore, in both oocytes and 2-cell embryos, G9a depletion has limited impact on the expression of genes and retrotransposons. Although their CG methylation is minimally affected, preimplantation embryos derived from such oocytes show abnormal chromosome segregation and frequent developmental arrest. Our findings illuminate the functional importance of G9a independent of CG methylation in preimplantation development and call into question the proposed role for H3K9me2 in CG methylation protection in zygotes.


Assuntos
Blastocisto/metabolismo , Metilação de DNA , Código das Histonas , Histona-Lisina N-Metiltransferase/metabolismo , Oócitos/metabolismo , Animais , Segregação de Cromossomos , Feminino , Histona-Lisina N-Metiltransferase/genética , Histonas/química , Histonas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL
13.
Nat Commun ; 9(1): 3331, 2018 08 20.
Artigo em Inglês | MEDLINE | ID: mdl-30127397

RESUMO

De novo DNA methylation (DNAme) during mouse oogenesis occurs within transcribed regions enriched for H3K36me3. As many oocyte transcripts originate in long terminal repeats (LTRs), which are heterogeneous even between closely related mammals, we examined whether species-specific LTR-initiated transcription units (LITs) shape the oocyte methylome. Here we identify thousands of syntenic regions in mouse, rat, and human that show divergent DNAme associated with private LITs, many of which initiate in lineage-specific LTR retrotransposons. Furthermore, CpG island (CGI) promoters methylated in mouse and/or rat, but not human oocytes, are embedded within rodent-specific LITs and vice versa. Notably, at a subset of such CGI promoters, DNAme persists on the maternal genome in fertilized and parthenogenetic mouse blastocysts or in human placenta, indicative of species-specific epigenetic inheritance. Polymorphic LITs are also responsible for disparate DNAme at promoter CGIs in distantly related mouse strains, revealing that LITs also promote intra-species divergence in CGI DNAme.


Assuntos
Metilação de DNA/genética , Padrões de Herança/genética , Oócitos/metabolismo , Retroelementos/genética , Sequências Repetidas Terminais/genética , Transcrição Gênica , Animais , Ilhas de CpG/genética , DNA Intergênico/genética , Fertilização/genética , Regulação da Expressão Gênica , Humanos , Mamíferos/metabolismo , Camundongos Endogâmicos C57BL , Polimorfismo Genético , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Especificidade da Espécie , Sintenia/genética
14.
BMC Genomics ; 19(1): 463, 2018 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-29907088

RESUMO

BACKGROUND: Allele-specific transcriptional regulation, including of imprinted genes, is essential for normal mammalian development. While the regulatory regions controlling imprinted genes are associated with DNA methylation (DNAme) and specific histone modifications, the interplay between transcription and these epigenetic marks at allelic resolution is typically not investigated genome-wide due to a lack of bioinformatic packages that can process and integrate multiple epigenomic datasets with allelic resolution. In addition, existing ad-hoc software only consider SNVs for allele-specific read discovery. This limitation omits potentially informative INDELs, which constitute about one fifth of the number of SNVs in mice, and introduces a systematic reference bias in allele-specific analyses. RESULTS: Here, we describe MEA, an INDEL-aware Methylomic and Epigenomic Allele-specific analysis pipeline which enables user-friendly data exploration, visualization and interpretation of allelic imbalance. Applying MEA to mouse embryonic datasets yields robust allele-specific DNAme maps and low reference bias. We validate allele-specific DNAme at known differentially methylated regions and show that automated integration of such methylation data with RNA- and ChIP-seq datasets yields an intuitive, multidimensional view of allelic gene regulation. MEA uncovers numerous novel dynamically methylated loci, highlighting the sensitivity of our pipeline. Furthermore, processing and visualization of epigenomic datasets from human brain reveals the expected allele-specific enrichment of H3K27ac and DNAme at imprinted as well as novel monoallelically expressed genes, highlighting MEA's utility for integrating human datasets of distinct provenance for genome-wide analysis of allelic phenomena. CONCLUSIONS: Our novel pipeline for standardized allele-specific processing and visualization of disparate epigenomic and methylomic datasets enables rapid analysis and navigation with allelic resolution. MEA is freely available as a Docker container at https://github.com/julienrichardalbert/MEA .


Assuntos
Alelos , Metilação de DNA , Epigênese Genética , Epigenômica/métodos , Software , Animais , Imunoprecipitação da Cromatina , Ilhas de CpG , Perfilação da Expressão Gênica , Células Germinativas/metabolismo , Humanos , Mutação INDEL , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Análise de Sequência de DNA , Análise de Sequência de RNA , Sítio de Iniciação de Transcrição
16.
Genome Res ; 28(1): 37-51, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29229671

RESUMO

Phosphorylation of histone H3 at serine 10 (H3S10ph) by Aurora kinases plays an important role in mitosis; however, H3S10ph also marks regulatory regions of inducible genes in interphase mammalian cells, implicating mitosis-independent functions. Using the fluorescent ubiquitin-mediated cell cycle indicator (FUCCI), we found that 30% of the genome in interphase mouse embryonic stem cells (ESCs) is marked with H3S10ph. H3S10ph broadly demarcates gene-rich regions in G1 and is positively correlated with domains of early DNA replication timing (RT) but negatively correlated with H3K9me2 and lamin-associated domains (LADs). Consistent with mitosis-independent kinase activity, this pattern was preserved in ESCs treated with Hesperadin, a potent inhibitor of Aurora B/C kinases. Disruption of H3S10ph by expression of nonphosphorylatable H3.3S10A results in ectopic spreading of H3K9me2 into adjacent euchromatic regions, mimicking the phenotype observed in Drosophila JIL-1 kinase mutants. Conversely, interphase H3S10ph domains expand in Ehmt1 (also known as Glp) null ESCs, revealing that H3S10ph deposition is restricted by H3K9me2. Strikingly, spreading of H3S10ph at RT transition regions (TTRs) is accompanied by aberrant transcription initiation of genes co-oriented with the replication fork in Ehmt1-/- and Ehmt2-/- ESCs, indicating that establishment of repressive chromatin on the leading strand following DNA synthesis may depend upon these lysine methyltransferases. H3S10ph is also anti-correlated with H3K9me2 in interphase murine embryonic fibroblasts (MEFs) and is restricted to intragenic regions of actively transcribing genes by EHMT2. Taken together, these observations reveal that H3S10ph may play a general role in restricting the spreading of repressive chromatin in interphase mammalian cells.


Assuntos
Cromatina/metabolismo , Replicação do DNA/fisiologia , Fibroblastos/metabolismo , Histonas/metabolismo , Interfase/fisiologia , Células-Tronco Embrionárias Murinas/metabolismo , Animais , Cromatina/genética , Drosophila melanogaster , Fibroblastos/citologia , Histonas/genética , Camundongos , Camundongos Knockout , Células-Tronco Embrionárias Murinas/citologia
17.
Dev Cell ; 43(3): 257-258, 2017 11 06.
Artigo em Inglês | MEDLINE | ID: mdl-29112847

RESUMO

CpG island promoters are generally devoid of DNA methylation in somatic cells but are frequently methylated during tumorigenesis. Reporting recently in Nature, Smith et al. (2017) show that the signaling-induced methylome in early extraembryonic tissues resembles that of many cancers, suggesting that placental nuclear programming might be co-opted in tumorigenesis.


Assuntos
Carcinogênese/genética , Ilhas de CpG/genética , Metilação de DNA/fisiologia , Neoplasias/genética , Placenta/metabolismo , Animais , Feminino , Humanos , Gravidez , Regiões Promotoras Genéticas/genética
19.
Nat Genet ; 49(7): 974-975, 2017 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-28656984

RESUMO

A study in this issue demonstrates that epigenome-modifying drugs used in cancer chemotherapy induce transcription from thousands of previously unannotated transcription start sites, most of which are derived from ancient endogenous retroviruses (ERVs). This work, coupled with previous related findings, suggests that induction of ERVs, rather than direct effects on specific genes, may have a central role in the cellular responses to such agents and, in turn, their therapeutic efficacy.


Assuntos
Retrovirus Endógenos , Epigenômica , Humanos
20.
PLoS Genet ; 12(10): e1006390, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27741228

RESUMO

[This corrects the article DOI: 10.1371/journal.pgen.1004933.].

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...