Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Mol Cell ; 64(6): 1062-1073, 2016 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-27916660

RESUMO

The methylcytosine oxidase TET proteins play important roles in DNA demethylation and development. However, it remains elusive how exactly they target substrates and execute oxidation. Interestingly, we found that, in mice, the full-length TET1 isoform (TET1e) is restricted to early embryos, embryonic stem cells (ESCs), and primordial germ cells (PGCs). By contrast, a short isoform (TET1s) is preferentially expressed in somatic cells, which lacks the N terminus including the CXXC domain, a DNA-binding module that often recognizes CpG islands (CGIs) where TET1 predominantly occupies. Unexpectedly, TET1s can still bind CGIs despite the fact that its global chromatin binding is significantly reduced. Interestingly, global chromatin binding, but not targeted binding at CGIs, is correlated with TET1-mediated demethylation. Finally, mice with exclusive expression of Tet1s failed to erase imprints in PGCs and displayed developmental defects in progeny. These data show that isoform switch of TET1 regulates epigenetic memory erasure and mouse development.


Assuntos
Proteínas de Ligação a DNA/genética , Impressão Genômica , Células-Tronco Embrionárias Murinas/metabolismo , Óvulo/metabolismo , Proteínas Proto-Oncogênicas/genética , Espermatozoides/metabolismo , Animais , Sítios de Ligação , Cromatina/química , Cromatina/metabolismo , Ilhas de CpG , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/metabolismo , Embrião de Mamíferos , Regulação da Expressão Gênica no Desenvolvimento , Masculino , Camundongos , Células-Tronco Embrionárias Murinas/citologia , Óvulo/citologia , Regiões Promotoras Genéticas , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas Proto-Oncogênicas/química , Proteínas Proto-Oncogênicas/metabolismo , Espermatozoides/citologia
2.
J Biol Chem ; 291(2): 731-8, 2016 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-26620559

RESUMO

In mammals, active DNA demethylation involves oxidation of 5-methylcytosine (5mC) into 5-formylcytosine (5fC) and 5-carboxylcytosine (5caC) by Tet dioxygenases and excision of these two oxidized bases by thymine DNA glycosylase (TDG). Although TDG is essential for active demethylation in embryonic stem cells and induced pluripotent stem cells, it is hardly expressed in mouse zygotes and dispensable in pronuclear DNA demethylation. To search for other factors that might contribute to demethylation in mammalian cells, we performed a functional genomics screen based on a methylated luciferase reporter assay. UNG2, one of the glycosylases known to excise uracil residues from DNA, was found to reduce DNA methylation, thus activating transcription of a methylation-silenced reporter gene when co-transfected with Tet2 into HEK293T cells. Interestingly, UNG2 could decrease 5caC from the genomic DNA and a reporter plasmid in transfected cells, like TDG. Furthermore, deficiency in Ung partially impaired DNA demethylation in mouse zygotes. Our results suggest that UNG might be involved in Tet-mediated DNA demethylation.


Assuntos
Metilação de DNA , Proteínas de Ligação a DNA/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Uracila-DNA Glicosidase/metabolismo , Animais , Citosina/análogos & derivados , DNA/metabolismo , Dioxigenases , Genes Reporter , Loci Gênicos , Genoma Humano , Células HEK293 , Humanos , Camundongos , Plasmídeos/metabolismo , Transfecção , Uracila/metabolismo , Uracila-DNA Glicosidase/deficiência , Zigoto/metabolismo
3.
Nature ; 477(7366): 606-10, 2011 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-21892189

RESUMO

Sperm and eggs carry distinctive epigenetic modifications that are adjusted by reprogramming after fertilization. The paternal genome in a zygote undergoes active DNA demethylation before the first mitosis. The biological significance and mechanisms of this paternal epigenome remodelling have remained unclear. Here we report that, within mouse zygotes, oxidation of 5-methylcytosine (5mC) occurs on the paternal genome, changing 5mC into 5-hydroxymethylcytosine (5hmC). Furthermore, we demonstrate that the dioxygenase Tet3 (ref. 5) is enriched specifically in the male pronucleus. In Tet3-deficient zygotes from conditional knockout mice, paternal-genome conversion of 5mC into 5hmC fails to occur and the level of 5mC remains constant. Deficiency of Tet3 also impedes the demethylation process of the paternal Oct4 and Nanog genes and delays the subsequent activation of a paternally derived Oct4 transgene in early embryos. Female mice depleted of Tet3 in the germ line show severely reduced fecundity and their heterozygous mutant offspring lacking maternal Tet3 suffer an increased incidence of developmental failure. Oocytes lacking Tet3 also seem to have a reduced ability to reprogram the injected nuclei from somatic cells. Therefore, Tet3-mediated DNA hydroxylation is involved in epigenetic reprogramming of the zygotic paternal DNA following natural fertilization and may also contribute to somatic cell nuclear reprogramming during animal cloning.


Assuntos
Reprogramação Celular , Proteínas de Ligação a DNA/metabolismo , Dioxigenases/metabolismo , Epigênese Genética , Oócitos/enzimologia , Oócitos/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , 5-Metilcitosina/metabolismo , Alelos , Animais , Citosina/análogos & derivados , Citosina/metabolismo , DNA/química , DNA/genética , DNA/metabolismo , Metilação de DNA/genética , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/genética , Dioxigenases/genética , Embrião de Mamíferos/embriologia , Embrião de Mamíferos/metabolismo , Desenvolvimento Embrionário , Feminino , Fertilidade/genética , Regulação da Expressão Gênica no Desenvolvimento , Células Germinativas/metabolismo , Masculino , Camundongos , Fator 3 de Transcrição de Octâmero/genética , Oócitos/citologia , Oxirredução , Proteínas Proto-Oncogênicas/deficiência , Proteínas Proto-Oncogênicas/genética , Zigoto/citologia , Zigoto/metabolismo
4.
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
5.
Nat Cell Biol ; 25(4): 528-539, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-37024683

RESUMO

Upon stimulation by extrinsic stimuli, stem cells initiate a programme that enables differentiation or self-renewal. Disruption of the stem state exit has catastrophic consequences for embryogenesis and can lead to cancer. While some elements of this stem state switch are known, major regulatory mechanisms remain unclear. Here we show that this switch involves a global increase in splicing efficiency coordinated by DNA methyltransferase 3α (DNMT3A), an enzyme typically involved in DNA methylation. Proper activation of murine and human embryonic and haematopoietic stem cells depends on messenger RNA processing, influenced by DNMT3A in response to stimuli. DNMT3A coordinates splicing through recruitment of the core spliceosome protein SF3B1 to RNA polymerase and mRNA. Importantly, the DNA methylation function of DNMT3A is not required and loss of DNMT3A leads to impaired splicing during stem cell turnover. Finally, we identify the spliceosome as a potential therapeutic target in DNMT3A-mutated leukaemias. Together, our results reveal a modality through which DNMT3A and the spliceosome govern exit from the stem state towards differentiation.


Assuntos
DNA (Citosina-5-)-Metiltransferases , DNA Metiltransferase 3A , Animais , Humanos , Camundongos , Diferenciação Celular/genética , DNA (Citosina-5-)-Metiltransferases/genética , DNA (Citosina-5-)-Metiltransferases/metabolismo , Metilação de DNA , Células-Tronco Hematopoéticas/metabolismo
6.
Cell Stem Cell ; 30(11): 1503-1519.e8, 2023 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-37863054

RESUMO

Somatic mutations accumulate in all cells with age and can confer a selective advantage, leading to clonal expansion over time. In hematopoietic cells, mutations in a subset of genes regulating DNA repair or epigenetics frequently lead to clonal hematopoiesis (CH). Here, we describe the context and mechanisms that lead to enrichment of hematopoietic stem cells (HSCs) with mutations in SRCAP, which encodes a chromatin remodeler that also influences DNA repair. We show that SRCAP mutations confer a selective advantage in human cells and in mice upon treatment with the anthracycline-class chemotherapeutic doxorubicin and bone marrow transplantation. Furthermore, Srcap mutations lead to a lymphoid-biased expansion, driven by loss of SRCAP-regulated H2A.Z deposition and increased DNA repair. Altogether, we demonstrate that SRCAP operates at the intersection of multiple pathways in stem and progenitor cells, offering a new perspective on the functional impact of genetic variants that promote stem cell competition in the hematopoietic system.


Assuntos
Hematopoiese Clonal , Hematopoese , Animais , Humanos , Camundongos , Adenosina Trifosfatases/genética , Adenosina Trifosfatases/metabolismo , Reparo do DNA/genética , Epigênese Genética , Hematopoese/genética , Mutação/genética
7.
Nat Genet ; 54(5): 625-636, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35534561

RESUMO

DNA methyltransferase 3a (DNMT3A) plays a crucial role during mammalian development. Two isoforms of DNMT3A are differentially expressed from stem cells to somatic tissues, but their individual functions remain largely uncharacterized. Here we report that the long isoform DNMT3A1, but not the short DNMT3A2, is essential for mouse postnatal development. DNMT3A1 binds to and regulates bivalent neurodevelopmental genes in the brain. Strikingly, Dnmt3a1 knockout perinatal lethality could be partially rescued by DNMT3A1 restoration in the nervous system. We further show that the intrinsically disordered N terminus of DNMT3A1 is required for normal development and DNA methylation at DNMT3A1-enriched regions. Mechanistically, a ubiquitin-interacting motif embedded in a putative α-helix within the N terminus binds to mono-ubiquitinated histone H2AK119, probably mediating recruitment of DNMT3A1 to Polycomb-regulated regions. These data demonstrate an isoform-specific role for DNMT3A1 in mouse postnatal development and reveal the N terminus as a necessary regulatory domain for DNMT3A1 chromatin occupancy and functions in the nervous system.


Assuntos
Metilases de Modificação do DNA , Histonas , Animais , Metilação de DNA , Metilases de Modificação do DNA/metabolismo , Histonas/metabolismo , Camundongos , Isoformas de Proteínas
8.
Elife ; 112022 05 30.
Artigo em Inglês | MEDLINE | ID: mdl-35635747

RESUMO

DNA Methyltransferase 3 A (DNMT3A) is an important facilitator of differentiation of both embryonic and hematopoietic stem cells. Heterozygous germline mutations in DNMT3A lead to Tatton-Brown-Rahman Syndrome (TBRS), characterized by obesity and excessive height. While DNMT3A is known to impact feeding behavior via the hypothalamus, here we investigated a role in adipocyte progenitors utilizing heterozygous knockout mice that recapitulate cardinal TBRS phenotypes. These mice become morbidly obese due to adipocyte enlargement and tissue expansion. Adipose tissue in these mice exhibited defects in preadipocyte maturation and precocious activation of inflammatory gene networks, including interleukin-6 signaling. Adipocyte progenitor cell lines lacking DNMT3A exhibited aberrant differentiation. Furthermore, mice in which Dnmt3a was specifically ablated in adipocyte progenitors showed enlarged fat depots and increased progenitor numbers, partly recapitulating the TBRS obesity phenotypes. Loss of DNMT3A led to constitutive DNA hypomethylation, such that the DNA methylation landscape of young adipocyte progenitors resemble that of older wild-type mice. Together, our results demonstrate that DNMT3A coordinates both the central and local control of energy storage required to maintain normal weight and prevent inflammatory obesity.


Assuntos
Deficiência Intelectual , Erros Inatos do Metabolismo , Obesidade Mórbida , Adipogenia , Animais , DNA , DNA (Citosina-5-)-Metiltransferases/genética , DNA (Citosina-5-)-Metiltransferases/metabolismo , DNA Metiltransferase 3A , Deficiência Intelectual/genética , Camundongos
9.
Eukaryot Cell ; 9(3): 379-86, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20081062

RESUMO

Spliceosomal small nuclear ribonucleoproteins (snRNPs) in trypanosomes contain either the canonical heptameric Sm ring or variant Sm cores with snRNA-specific Sm subunits. Here we show biochemically by a combination of RNase H cleavage and tandem affinity purification that the U4 snRNP contains a variant Sm heteroheptamer core in which only SmD3 is replaced by SSm4. This U4-specific, nuclear-localized Sm core protein is essential for growth and splicing. As shown by RNA interference (RNAi) knockdown, SSm4 is specifically required for the integrity of the U4 snRNA and the U4/U6 di-snRNP in trypanosomes. In addition, we demonstrate by in vitro reconstitution of Sm cores that under stringent conditions, the SSm4 protein suffices to specify the assembly of U4 Sm cores. Together, these data indicate that the assembly of the U4-specific Sm core provides an essential step in U4/U6 di-snRNP biogenesis and splicing in trypanosomes.


Assuntos
Multimerização Proteica/fisiologia , Splicing de RNA , RNA Nuclear Pequeno/metabolismo , Ribonucleoproteína Nuclear Pequena U4-U6/biossíntese , Trypanosoma brucei brucei/metabolismo , Proteínas Centrais de snRNP/metabolismo , Proliferação de Células , Centrifugação com Gradiente de Concentração , Expressão Gênica/genética , Espaço Intranuclear/metabolismo , Espectrometria de Massas , Ligação Proteica/genética , Interferência de RNA , RNA de Cadeia Dupla/genética , RNA Nuclear Pequeno/genética , Ribonuclease H/metabolismo , Ribonucleoproteína Nuclear Pequena U4-U6/química , Ribonucleoproteína Nuclear Pequena U4-U6/genética , Trypanosoma brucei brucei/genética , Proteínas Centrais de snRNP/genética
10.
Nat Cell Biol ; 22(10): 1162-1169, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32958856

RESUMO

Stem cells need to be protected from genotoxic and proteotoxic stress to maintain a healthy pool throughout life1-3. Little is known about the proteostasis mechanism that safeguards stem cells. Here we report endoplasmic reticulum-associated degradation (ERAD) as a protein quality checkpoint that controls the haematopoietic stem cell (HSC)-niche interaction and determines the fate of HSCs. The SEL1L-HRD1 complex, the most conserved branch of ERAD4, is highly expressed in HSCs. Deletion of Sel1l led to niche displacement of HSCs and a complete loss of HSC identity, and allowed highly efficient donor-HSC engraftment without irradiation. Mechanistic studies identified MPL, the master regulator of HSC identity5, as a bona fide ERAD substrate that became aggregated in the endoplasmic reticulum following ERAD deficiency. Restoration of MPL signalling with an agonist partially rescued the number and reconstitution capacity of Sel1l-deficient HSCs. Our study defines ERAD as an essential proteostasis mechanism to safeguard a healthy stem cell pool by regulating the stem cell-niche interaction.


Assuntos
Degradação Associada com o Retículo Endoplasmático , Retículo Endoplasmático/metabolismo , Células-Tronco Hematopoéticas/citologia , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Receptores de Trombopoetina/metabolismo , Nicho de Células-Tronco , Ubiquitina-Proteína Ligases/metabolismo , Animais , Feminino , Células-Tronco Hematopoéticas/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de Trombopoetina/genética , Ubiquitina-Proteína Ligases/genética
12.
Genome Biol ; 19(1): 88, 2018 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-30001199

RESUMO

BACKGROUND: DNA methylation is a heritable epigenetic mark, enabling stable but reversible gene repression. In mammalian cells, DNA methyltransferases (DNMTs) are responsible for modifying cytosine to 5-methylcytosine (5mC), which can be further oxidized by the TET dioxygenases to ultimately cause DNA demethylation. However, the genome-wide cooperation and functions of these two families of proteins, especially at large under-methylated regions, called canyons, remain largely unknown. RESULTS: Here we demonstrate that DNMT3A and TET1 function in a complementary and competitive manner in mouse embryonic stem cells to mediate proper epigenetic landscapes and gene expression. The longer isoform of DNMT3A, DNMT3A1, exhibits significant enrichment at distal promoters and canyon edges, but is excluded from proximal promoters and canyons where TET1 shows prominent binding. Deletion of Tet1 increases DNMT3A1 binding capacity at and around genes with wild-type TET1 binding. However, deletion of Dnmt3a has a minor effect on TET1 binding on chromatin, indicating that TET1 may limit DNA methylation partially by protecting its targets from DNMT3A and establishing boundaries for DNA methylation. Local CpG density may determine their complementary binding patterns and therefore that the methylation landscape is encoded in the DNA sequence. Furthermore, DNMT3A and TET1 impact histone modifications which in turn regulate gene expression. In particular, they regulate Polycomb Repressive Complex 2 (PRC2)-mediated H3K27me3 enrichment to constrain gene expression from bivalent promoters. CONCLUSIONS: We conclude that DNMT3A and TET1 regulate the epigenome and gene expression at specific targets via their functional interplay.


Assuntos
DNA (Citosina-5-)-Metiltransferases/genética , Proteínas de Ligação a DNA/genética , Epigênese Genética/genética , Células-Tronco Embrionárias Murinas/metabolismo , Regiões Promotoras Genéticas/genética , Proteínas Proto-Oncogênicas/genética , 5-Metilcitosina/metabolismo , Animais , Linhagem Celular , Cromatina/genética , Metilação de DNA/genética , DNA Metiltransferase 3A , Dioxigenases/genética , Epigenômica/métodos , Camundongos
13.
Science ; 372(6538): 128-129, 2021 04 09.
Artigo em Inglês | MEDLINE | ID: mdl-33833110
14.
Cell Stem Cell ; 15(4): 447-459, 2014 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-25220291

RESUMO

The epigenomes of mammalian sperm and oocytes, characterized by gamete-specific 5-methylcytosine (5mC) patterns, are reprogrammed during early embryogenesis to establish full developmental potential. Previous studies have suggested that the paternal genome is actively demethylated in the zygote while the maternal genome undergoes subsequent passive demethylation via DNA replication during cleavage. Active demethylation is known to depend on 5mC oxidation by Tet dioxygenases and excision of oxidized bases by thymine DNA glycosylase (TDG). Here we show that both maternal and paternal genomes undergo widespread active and passive demethylation in zygotes before the first mitotic division. Passive demethylation was blocked by the replication inhibitor aphidicolin, and active demethylation was abrogated by deletion of Tet3 in both pronuclei. At actively demethylated loci, 5mCs were processed to unmodified cytosines. Surprisingly, the demethylation process was unaffected by the deletion of TDG from the zygote, suggesting the existence of other demethylation mechanisms downstream of Tet3-mediated oxidation.


Assuntos
Núcleo Celular/metabolismo , Metilação de DNA/genética , Mamíferos/genética , Zigoto/metabolismo , 5-Metilcitosina/metabolismo , Animais , Proteínas de Ligação a DNA/metabolismo , Dioxigenases , Feminino , Deleção de Genes , Loci Gênicos , Genoma/genética , Masculino , Dados de Sequência Molecular , Oxirredução , Proteínas Proto-Oncogênicas/metabolismo , Reprodutibilidade dos Testes , Reprodução/genética , Análise de Sequência de DNA , Timina DNA Glicosilase/metabolismo
15.
Nat Genet ; 45(12): 1504-9, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24162740

RESUMO

Vitamin C, a micronutrient known for its anti-scurvy activity in humans, promotes the generation of induced pluripotent stem cells (iPSCs) through the activity of histone demethylating dioxygenases. TET hydroxylases are also dioxygenases implicated in active DNA demethylation. Here we report that TET1 either positively or negatively regulates somatic cell reprogramming depending on the absence or presence of vitamin C. TET1 deficiency enhances reprogramming, and its overexpression impairs reprogramming in the context of vitamin C by modulating the obligatory mesenchymal-to-epithelial transition (MET). In the absence of vitamin C, TET1 promotes somatic cell reprogramming independent of MET. Consistently, TET1 regulates 5-hydroxymethylcytosine (5hmC) formation at loci critical for MET in a vitamin C-dependent fashion. Our findings suggest that vitamin C has a vital role in determining the biological outcome of TET1 function at the cellular level. Given its benefit to human health, vitamin C should be investigated further for its role in epigenetic regulation.


Assuntos
Ácido Ascórbico/farmacologia , Reprogramação Celular/efeitos dos fármacos , Proteínas de Ligação a DNA/fisiologia , Proteínas Proto-Oncogênicas/fisiologia , Animais , Células Cultivadas , Embrião de Mamíferos , Epigênese Genética/efeitos dos fármacos , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Células-Tronco Pluripotentes Induzidas/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Camundongos Knockout
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA