Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 434
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Mol Cell ; 77(4): 857-874.e9, 2020 02 20.
Artigo em Inglês | MEDLINE | ID: mdl-31883950

RESUMO

The Polycomb repressive system is an essential chromatin-based regulator of gene expression. Despite being extensively studied, how the Polycomb system selects its target genes is poorly understood, and whether its histone-modifying activities are required for transcriptional repression remains controversial. Here, we directly test the requirement for PRC1 catalytic activity in Polycomb system function. To achieve this, we develop a conditional mutation system in embryonic stem cells that completely removes PRC1 catalytic activity. Using this system, we demonstrate that catalysis by PRC1 drives Polycomb chromatin domain formation and long-range chromatin interactions. Furthermore, we show that variant PRC1 complexes with DNA-binding activities occupy target sites independently of PRC1 catalytic activity, providing a putative mechanism for Polycomb target site selection. Finally, we discover that Polycomb-mediated gene repression requires PRC1 catalytic activity. Together these discoveries provide compelling evidence that PRC1 catalysis is central to Polycomb system function and gene regulation.


Assuntos
Regulação da Expressão Gênica , Complexo Repressor Polycomb 1/metabolismo , Animais , Biocatálise , Linhagem Celular , Cromatina/metabolismo , Células-Tronco Embrionárias/enzimologia , Células-Tronco Embrionárias/metabolismo , Células HEK293 , Histonas/metabolismo , Humanos , Masculino , Camundongos , Mutação Puntual , Complexo Repressor Polycomb 1/genética , Complexo Repressor Polycomb 2/metabolismo , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo
2.
Mol Cell ; 66(4): 568-576.e4, 2017 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-28483418

RESUMO

Monomethylation of histone H3 at lysine 4 (H3K4me1) and acetylation of histone H3 at lysine 27 (H3K27ac) are correlated with transcriptionally engaged enhancer elements, but the functional impact of these modifications on enhancer activity is not well understood. Here we used CRISPR/Cas9 genome editing to separate catalytic activity-dependent and independent functions of Mll3 (Kmt2c) and Mll4 (Kmt2d, Mll2), the major enhancer H3K4 monomethyltransferases. Loss of H3K4me1 from enhancers in Mll3/4 catalytically deficient cells causes partial reduction of H3K27ac, but has surprisingly minor effects on transcription from either enhancers or promoters. In contrast, loss of Mll3/4 proteins leads to strong depletion of enhancer Pol II occupancy and eRNA synthesis, concomitant with downregulation of target genes. Interestingly, downregulated genes exhibit reduced polymerase levels in gene bodies, but not at promoters, suggestive of pause-release defects. Altogether, our results suggest that enhancer H3K4me1 provides only a minor contribution to the long-range coactivator function of Mll3/4.


Assuntos
Células-Tronco Embrionárias/enzimologia , Elementos Facilitadores Genéticos , Histona-Lisina N-Metiltransferase/metabolismo , Histonas/metabolismo , Regiões Promotoras Genéticas , RNA/biossíntese , Transcrição Gênica , Animais , Sistemas CRISPR-Cas , Linhagem Celular , Edição de Genes , Regulação da Expressão Gênica no Desenvolvimento , Histona-Lisina N-Metiltransferase/genética , Masculino , Metilação , Camundongos , Mutação , RNA/genética , Fatores de Tempo , Ativação Transcricional , Transfecção
3.
Mol Cell ; 65(1): 91-104, 2017 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-27939942

RESUMO

Ataxia-telangiectasia mutated (ATM) regulates the DNA damage response as well as DNA double-strand break repair through homologous recombination. Here we show that ATM is hyperactive when the catalytic subunit of DNA-dependent protein kinase (DNA-PKcs) is chemically inhibited or when the DNA-PKcs gene is deleted in human cells. Pre-incubation of ATM protein with active DNA-PKcs also significantly reduces ATM activity in vitro. We characterize several phosphorylation sites in ATM that are targets of DNA-PKcs and show that phospho-mimetic mutations at these residues significantly inhibit ATM activity and impair ATM signaling upon DNA damage. In contrast, phospho-blocking mutations at one cluster of sites increase the frequency of apoptosis during normal cell growth. DNA-PKcs, which is integral to the non-homologous end joining pathway, thus negatively regulates ATM activity through phosphorylation of ATM. These observations illuminate an important regulatory mechanism for ATM that also controls DNA repair pathway choice.


Assuntos
Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Quebras de DNA de Cadeia Dupla , Reparo do DNA , Proteína Quinase Ativada por DNA/metabolismo , Proteínas Nucleares/metabolismo , Apoptose , Proteínas Mutadas de Ataxia Telangiectasia/genética , Ciclo Celular , Linhagem Celular Tumoral , Proliferação de Células , Proteína Quinase Ativada por DNA/genética , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Células-Tronco Embrionárias/enzimologia , Genótipo , Células HEK293 , Humanos , Mutação , Proteínas Nucleares/genética , Estresse Oxidativo , Fenótipo , Fosforilação , Interferência de RNA , Transdução de Sinais , Fatores de Tempo , Transfecção
4.
Mol Cell ; 67(2): 308-321.e6, 2017 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-28732206

RESUMO

Enhancer activation is a critical step for gene activation. Here we report an epigenetic crosstalk at enhancers between the UTX (H3K27 demethylase)-MLL4 (H3K4 methyltransferase) complex and the histone acetyltransferase p300. We demonstrate that UTX, in a demethylase activity-independent manner, facilitates conversion of inactive enhancers in embryonic stem cells to an active (H3K4me1+/H3K27ac+) state by recruiting and coupling the enzymatic functions of MLL4 and p300. Loss of UTX leads to attenuated enhancer activity, characterized by reduced levels of H3K4me1 and H3K27ac as well as impaired transcription. The UTX-MLL4 complex enhances p300-dependent H3K27 acetylation through UTX-dependent stimulation of p300 recruitment, while MLL4-mediated H3K4 monomethylation, reciprocally, requires p300 function. Importantly, MLL4-generated H3K4me1 further enhances p300-dependent transcription. This work reveals a previously unrecognized cooperativity among enhancer-associated chromatin modulators, including a unique function for UTX, in establishing an "active enhancer landscape" and defines a detailed mechanism for the joint deposition of H3K4me1 and H3K27ac.


Assuntos
Cromatina/metabolismo , Proteína p300 Associada a E1A/metabolismo , Células-Tronco Embrionárias/enzimologia , Elementos Facilitadores Genéticos , Histona Desmetilases/metabolismo , Histona-Lisina N-Metiltransferase/metabolismo , Transcrição Gênica , Ativação Transcricional , Animais , Cromatina/genética , Montagem e Desmontagem da Cromatina , Proteína p300 Associada a E1A/genética , Retroalimentação Fisiológica , Redes Reguladoras de Genes , Células HEK293 , Histona Desmetilases/genética , Histona-Lisina N-Metiltransferase/genética , Histonas/metabolismo , Humanos , Masculino , Metilação , Camundongos , Interferência de RNA , Transfecção
5.
Mol Cell ; 65(4): 589-603.e9, 2017 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-28212747

RESUMO

Pioneer transcription factors (TFs) function as genomic first responders, binding to inaccessible regions of chromatin to promote enhancer formation. The mechanism by which pioneer TFs gain access to chromatin remains an important unanswered question. Here we show that PARP-1, a nucleosome-binding protein, cooperates with intrinsic properties of the pioneer TF Sox2 to facilitate its binding to intractable genomic loci in embryonic stem cells. These actions of PARP-1 occur independently of its poly(ADP-ribosyl) transferase activity. PARP-1-dependent Sox2-binding sites reside in euchromatic regions of the genome with relatively high nucleosome occupancy and low co-occupancy by other transcription factors. PARP-1 stabilizes Sox2 binding to nucleosomes at suboptimal sites through cooperative interactions on DNA. Our results define intrinsic and extrinsic features that determine Sox2 pioneer activity. The conditional pioneer activity observed with Sox2 at a subset of binding sites may be a key feature of other pioneer TFs operating at intractable genomic loci.


Assuntos
DNA/metabolismo , Células-Tronco Embrionárias/enzimologia , Eucromatina/enzimologia , Regulação da Expressão Gênica no Desenvolvimento , Loci Gênicos , Nucleossomos/enzimologia , Células-Tronco Pluripotentes/enzimologia , Poli(ADP-Ribose) Polimerase-1/metabolismo , Fatores de Transcrição SOXB1/metabolismo , Animais , Sítios de Ligação , Linhagem Celular , DNA/genética , Eucromatina/genética , Humanos , Camundongos , Nucleossomos/genética , Poli(ADP-Ribose) Polimerase-1/genética , Ligação Proteica , Fatores de Transcrição SOXB1/genética , Transdução de Sinais , Fatores de Tempo , Transfecção
6.
Mol Cell ; 67(4): 594-607.e4, 2017 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-28735899

RESUMO

Pervasive transcription initiates from cryptic promoters and is observed in eukaryotes ranging from yeast to mammals. The Set2-Rpd3 regulatory system prevents cryptic promoter function within expressed genes. However, conserved systems that control pervasive transcription within intergenic regions have not been well established. Here we show that Mot1, Ino80 chromatin remodeling complex (Ino80C), and NC2 co-localize on chromatin and coordinately suppress pervasive transcription in S. cerevisiae and murine embryonic stem cells (mESCs). In yeast, all three proteins bind subtelomeric heterochromatin through a Sir3-stimulated mechanism and to euchromatin via a TBP-stimulated mechanism. In mESCs, the proteins bind to active and poised TBP-bound promoters along with promoters of polycomb-silenced genes apparently lacking TBP. Depletion of Mot1, Ino80C, or NC2 by anchor away in yeast or RNAi in mESCs leads to near-identical transcriptome phenotypes, with new subtelomeric transcription in yeast, and greatly increased pervasive transcription in both yeast and mESCs.


Assuntos
Adenosina Trifosfatases/metabolismo , Células-Tronco Embrionárias/enzimologia , Fosfoproteínas/metabolismo , Proteínas Repressoras/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/enzimologia , Fatores Associados à Proteína de Ligação a TATA/metabolismo , Fatores de Transcrição/metabolismo , Transcrição Gênica , ATPases Associadas a Diversas Atividades Celulares , Adenosina Trifosfatases/genética , Sítios de Ligação , Linhagem Celular , Proteínas de Ligação a DNA , Eucromatina/genética , Eucromatina/metabolismo , Regulação Fúngica da Expressão Gênica , Inativação Gênica , Genótipo , Heterocromatina/genética , Heterocromatina/metabolismo , Fenótipo , Fosfoproteínas/genética , Regiões Promotoras Genéticas , Ligação Proteica , Interferência de RNA , Proteínas Repressoras/genética , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Proteínas Reguladoras de Informação Silenciosa de Saccharomyces cerevisiae/genética , Proteínas Reguladoras de Informação Silenciosa de Saccharomyces cerevisiae/metabolismo , Fatores Associados à Proteína de Ligação a TATA/genética , Proteína de Ligação a TATA-Box/genética , Proteína de Ligação a TATA-Box/metabolismo , Fator de Transcrição TFIID , Fatores de Transcrição/genética , Transfecção
7.
Mol Cell ; 67(4): 550-565.e5, 2017 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-28803780

RESUMO

DNA methylation is an essential epigenetic mark in mammals that has to be re-established after each round of DNA replication. The protein UHRF1 is essential for this process; it has been proposed that the protein targets newly replicated DNA by cooperatively binding hemi-methylated DNA and H3K9me2/3, but this model leaves a number of questions unanswered. Here, we present evidence for a direct recruitment of UHRF1 by the replication machinery via DNA ligase 1 (LIG1). A histone H3K9-like mimic within LIG1 is methylated by G9a and GLP and, compared with H3K9me2/3, more avidly binds UHRF1. Interaction with methylated LIG1 promotes the recruitment of UHRF1 to DNA replication sites and is required for DNA methylation maintenance. These results further elucidate the function of UHRF1, identify a non-histone target of G9a and GLP, and provide an example of a histone mimic that coordinates DNA replication and DNA methylation maintenance.


Assuntos
Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , DNA Ligase Dependente de ATP/metabolismo , Metilação de DNA , Replicação do DNA , DNA/biossíntese , Epigênese Genética , Antígenos de Histocompatibilidade/metabolismo , Histona-Lisina N-Metiltransferase/metabolismo , Processamento de Proteína Pós-Traducional , Animais , Proteínas Estimuladoras de Ligação a CCAAT/química , Proteínas Estimuladoras de Ligação a CCAAT/genética , DNA/genética , DNA Ligase Dependente de ATP/química , DNA Ligase Dependente de ATP/genética , Células-Tronco Embrionárias/enzimologia , Células HEK293 , Células HeLa , Antígenos de Histocompatibilidade/química , Antígenos de Histocompatibilidade/genética , Histona-Lisina N-Metiltransferase/química , Histona-Lisina N-Metiltransferase/genética , Histonas/metabolismo , Humanos , Lisina , Metilação , Camundongos , Modelos Moleculares , Mimetismo Molecular , Mutação , Ligação Proteica , Conformação Proteica , Relação Estrutura-Atividade , Transfecção , Domínio Tudor , Ubiquitina-Proteína Ligases
8.
Mol Cell ; 65(2): 260-271, 2017 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-28107648

RESUMO

Poly(ADP-ribosyl)ation (PARylation) is a post-translational modification of proteins mediated by PARP family members, such as PARP-1. Although PARylation has been studied extensively, few examples of definitive biological roles for site-specific PARylation have been reported. Here we show that C/EBPß, a key pro-adipogenic transcription factor, is PARylated by PARP-1 on three amino acids in a conserved regulatory domain. PARylation at these sites inhibits C/EBPß's DNA binding and transcriptional activities and attenuates adipogenesis in various genetic and cell-based models. Interestingly, PARP-1 catalytic activity drops precipitously during the first 48 hr of differentiation, corresponding to a release of C/EBPß from PARylation-mediated inhibition. This promotes the binding of C/EBPß at enhancers controlling the expression of adipogenic target genes and continued differentiation. Depletion or chemical inhibition of PARP-1, or mutation of the PARylation sites on C/EBPß, enhances these early adipogenic events. Collectively, our results provide a clear example of how site-specific PARylation drives biological outcomes.


Assuntos
Adipócitos/enzimologia , Adipogenia , Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Células-Tronco Embrionárias/enzimologia , Poli(ADP-Ribose) Polimerase-1/metabolismo , Poli Adenosina Difosfato Ribose/metabolismo , Processamento de Proteína Pós-Traducional , Transcrição Gênica , Células 3T3-L1 , Adipócitos/efeitos dos fármacos , Adipogenia/efeitos dos fármacos , Animais , Sítios de Ligação , Proteína beta Intensificadora de Ligação a CCAAT/genética , DNA/genética , DNA/metabolismo , Células-Tronco Embrionárias/efeitos dos fármacos , Genótipo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação , Células NIH 3T3 , Fenótipo , Poli(ADP-Ribose) Polimerase-1/antagonistas & inibidores , Poli(ADP-Ribose) Polimerase-1/deficiência , Poli(ADP-Ribose) Polimerase-1/genética , Inibidores de Poli(ADP-Ribose) Polimerases/farmacologia , Ligação Proteica , Domínios Proteicos , Interferência de RNA , Transdução de Sinais , Fatores de Tempo , Transcrição Gênica/efeitos dos fármacos , Ativação Transcricional , Transfecção
9.
Genes Dev ; 31(17): 1732-1737, 2017 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-28939616

RESUMO

Of the six members of the COMPASS (complex of proteins associated with Set1) family of histone H3 Lys4 (H3K4) methyltransferases identified in mammals, Set1A has been shown to be essential for early embryonic development and the maintenance of embryonic stem cell (ESC) self-renewal. Like its familial relatives, Set1A possesses a catalytic SET domain responsible for histone H3K4 methylation. Whether H3K4 methylation by Set1A/COMPASS is required for ESC maintenance and during differentiation has not yet been addressed. Here, we generated ESCs harboring the deletion of the SET domain of Set1A (Set1AΔSET); surprisingly, the Set1A SET domain is dispensable for ESC proliferation and self-renewal. The removal of the Set1A SET domain does not diminish bulk H3K4 methylation in ESCs; instead, only a subset of genomic loci exhibited reduction in H3K4me3 in Set1AΔSET cells, suggesting a role for Set1A independent of its catalytic domain in ESC self-renewal. However, Set1AΔSET ESCs are unable to undergo normal differentiation, indicating the importance of Set1A-dependent H3K4 methylation during differentiation. Our data also indicate that during differentiation, Set1A but not Mll2 functions as the H3K4 methylase on bivalent genes and is required for their expression, supporting a model for transcriptional switch between Mll2 and Set1A during the self-renewing-to-differentiation transition. Together, our study implicates a critical role for Set1A catalytic methyltransferase activity in regulating ESC differentiation but not self-renewal and suggests the existence of context-specific H3K4 methylation that regulates transcriptional outputs during ESC pluripotency.


Assuntos
Diferenciação Celular/genética , Células-Tronco Embrionárias/citologia , Histona-Lisina N-Metiltransferase/genética , Histona-Lisina N-Metiltransferase/metabolismo , Histonas/metabolismo , Animais , Proliferação de Células/genética , Células-Tronco Embrionárias/enzimologia , Deleção de Genes , Regulação da Expressão Gênica no Desenvolvimento/genética , Metilação , Camundongos , Proteína de Leucina Linfoide-Mieloide/metabolismo , Domínios PR-SET/genética
10.
Mol Cell ; 62(6): 958-966, 2016 06 16.
Artigo em Inglês | MEDLINE | ID: mdl-27237050

RESUMO

Ubiquitination typically occurs through the sequential action of three enzymes catalyzing ubiquitin activation (E1), conjugation (E2), and ligation (E3) and regulates diverse eukaryotic cellular processes. Although monoubiquitination commonly confers nondegradative activities, mechanisms underlying its temporal and spatial regulation and functional plasticity still remain largely unknown. Here we demonstrate that SETDB1, a major histone H3K9 methyltransferase is monoubiquitinated at the evolutionarily conserved lysine-867 in its SET-Insertion domain. This ubiquitination is directly catalyzed by UBE2E family of E2 enzymes in an E3-independent manner while the conjugated-ubiquitin (Ub) is protected from active deubiquitination. The resulting constitutive lysine-867 monoubiquitination is essential for SETDB1's enzymatic activity and endogenous retrovirus silencing in murine embryonic stem cells. Furthermore, the canonical hydrophobic patch on the conjugated-Ub is critical for Ub protection and function. Together, our findings highlight an E3-independent mechanism for monoubiquitination and reveal mechanistic details of SETDB1's enzymatic activity and the functional significance of its SET-Insertion.


Assuntos
Metilação de DNA , Células-Tronco Embrionárias/enzimologia , Histona-Lisina N-Metiltransferase/metabolismo , Histonas/metabolismo , Proteínas Metiltransferases/metabolismo , Enzimas de Conjugação de Ubiquitina/metabolismo , Ubiquitinação , Motivos de Aminoácidos , Animais , Sistemas CRISPR-Cas , Catálise , Retrovirus Endógenos/genética , Inativação Gênica , Células HEK293 , Células HeLa , Histona-Lisina N-Metiltransferase/química , Histona-Lisina N-Metiltransferase/genética , Humanos , Interações Hidrofóbicas e Hidrofílicas , Lisina , Células MCF-7 , Camundongos , Conformação Proteica , Proteínas Metiltransferases/química , Proteínas Metiltransferases/genética , Relação Estrutura-Atividade , Transfecção , Enzimas de Conjugação de Ubiquitina/química , Enzimas de Conjugação de Ubiquitina/genética
11.
Mol Cell ; 62(2): 307-313, 2016 04 21.
Artigo em Inglês | MEDLINE | ID: mdl-27067599

RESUMO

One recurring theme in drug development is to exploit synthetic lethal properties as means to preferentially damage the DNA of cancer cells. We and others have previously developed inhibitors of the ATR kinase, shown to be particularly genotoxic for cells expressing certain oncogenes. In contrast, the mechanisms of resistance to ATR inhibitors remain unexplored. We report here on a genome-wide CRISPR-Cas9 screen that identified CDC25A as a major determinant of sensitivity to ATR inhibition. CDC25A-deficient cells resist high doses of ATR inhibitors, which we show is due to their failure to prematurely enter mitosis in response to the drugs. Forcing mitotic entry with WEE1 inhibitors restores the toxicity of ATR inhibitors in CDC25A-deficient cells. With ATR inhibitors now entering the clinic, our work provides a better understanding of the mechanisms by which these compounds kill cells and reveals genetic interactions that could be used for their rational use.


Assuntos
Antineoplásicos/farmacologia , Sistemas CRISPR-Cas , Resistencia a Medicamentos Antineoplásicos/genética , Células-Tronco Embrionárias/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Fosfatases cdc25/metabolismo , Proteínas Mutadas de Ataxia Telangiectasia/antagonistas & inibidores , Proteínas Mutadas de Ataxia Telangiectasia/genética , Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Proteínas de Ciclo Celular/antagonistas & inibidores , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular , Relação Dose-Resposta a Droga , Células-Tronco Embrionárias/enzimologia , Células-Tronco Embrionárias/patologia , Estudo de Associação Genômica Ampla , Humanos , Mitose/efeitos dos fármacos , Terapia de Alvo Molecular , Proteínas Nucleares/antagonistas & inibidores , Proteínas Nucleares/metabolismo , Proteínas Tirosina Quinases/antagonistas & inibidores , Proteínas Tirosina Quinases/metabolismo , Interferência de RNA , Transdução de Sinais/efeitos dos fármacos , Transfecção , Fosfatases cdc25/genética
12.
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
13.
Nature ; 548(7666): 219-223, 2017 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-28746311

RESUMO

Concomitant activation of the Wnt pathway and suppression of Mapk signalling by two small molecule inhibitors (2i) in the presence of leukaemia inhibitory factor (LIF) (hereafter termed 2i/L) induces a naive state in mouse embryonic stem (ES) cells that resembles the inner cell mass (ICM) of the pre-implantation embryo. Since the ICM exists only transiently in vivo, it remains unclear how sustained propagation of naive ES cells in vitro affects their stability and functionality. Here we show that prolonged culture of male mouse ES cells in 2i/L results in irreversible epigenetic and genomic changes that impair their developmental potential. Furthermore, we find that female ES cells cultured in conventional serum plus LIF medium phenocopy male ES cells cultured in 2i/L. Mechanistically, we demonstrate that the inhibition of Mek1/2 is predominantly responsible for these effects, in part through the downregulation of DNA methyltransferases and their cofactors. Finally, we show that replacement of the Mek1/2 inhibitor with a Src inhibitor preserves the epigenetic and genomic integrity as well as the developmental potential of ES cells. Taken together, our data suggest that, although short-term suppression of Mek1/2 in ES cells helps to maintain an ICM-like epigenetic state, prolonged suppression results in irreversible changes that compromise their developmental potential.


Assuntos
Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/enzimologia , MAP Quinase Quinase 1/antagonistas & inibidores , MAP Quinase Quinase 2/antagonistas & inibidores , Animais , Blastocisto , Instabilidade Cromossômica , Metilação de DNA , Feminino , Impressão Genômica , Cariotipagem , Masculino , Camundongos
14.
Nature ; 549(7673): 538-542, 2017 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-28959962

RESUMO

Glycosylation, the covalent attachment of carbohydrate structures onto proteins, is the most abundant post-translational modification. Over 50% of human proteins are glycosylated, which alters their activities in diverse fundamental biological processes. Despite the importance of glycosylation in biology, the identification and functional validation of complex glycoproteins has remained largely unexplored. Here we develop a novel quantitative approach to identify intact glycopeptides from comparative proteomic data sets, allowing us not only to infer complex glycan structures but also to directly map them to sites within the associated proteins at the proteome scale. We apply this method to human and mouse embryonic stem cells to illuminate the stem cell glycoproteome. This analysis nearly doubles the number of experimentally confirmed glycoproteins, identifies previously unknown glycosylation sites and multiple glycosylated stemness factors, and uncovers evolutionarily conserved as well as species-specific glycoproteins in embryonic stem cells. The specificity of our method is confirmed using sister stem cells carrying repairable mutations in enzymes required for fucosylation, Fut9 and Slc35c1. Ablation of fucosylation confers resistance to the bioweapon ricin, and we discover proteins that carry a fucosylation-dependent sugar code for ricin toxicity. Mutations disrupting a subset of these proteins render cells ricin resistant, revealing new players that orchestrate ricin toxicity. Our comparative glycoproteomics platform, SugarQb, enables genome-wide insights into protein glycosylation and glycan modifications in complex biological systems.


Assuntos
Células-Tronco Embrionárias/química , Células-Tronco Embrionárias/efeitos dos fármacos , Glicopeptídeos/análise , Glicoproteínas/análise , Proteoma/análise , Proteômica , Ricina/toxicidade , Animais , Células-Tronco Embrionárias/enzimologia , Células-Tronco Embrionárias/metabolismo , Fucosiltransferases/genética , Glicopeptídeos/química , Glicopeptídeos/metabolismo , Glicoproteínas/química , Glicoproteínas/metabolismo , Glicosilação , Humanos , Proteínas de Membrana Transportadoras/genética , Camundongos , Proteínas de Transporte de Monossacarídeos , Processamento de Proteína Pós-Traducional/genética , Proteoma/química , Proteoma/genética , Proteoma/metabolismo
15.
Mol Cell ; 58(2): 311-322, 2015 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-25773599

RESUMO

The remarkable capacity for pluripotency and self-renewal in embryonic stem cells (ESCs) requires a finely tuned transcriptional circuitry wherein the pathways and genes that initiate differentiation are suppressed, but poised to respond rapidly to developmental signals. To elucidate transcriptional control in mouse ESCs in the naive, ground state, we defined the distribution of engaged RNA polymerase II (Pol II) at high resolution. We find that promoter-proximal pausing of Pol II is most enriched at genes regulating cell cycle and signal transduction and not, as expected, at developmental or bivalent genes. Accordingly, ablation of the primary pause-inducing factor NELF does not increase expression of lineage markers, but instead causes proliferation defects, embryonic lethality, and dysregulation of ESC signaling pathways. Indeed, ESCs lacking NELF have dramatically attenuated FGF/ERK activity, rendering them resistant to differentiation. This work thus uncovers a key role for NELF-mediated pausing in establishing the responsiveness of stem cells to developmental cues.


Assuntos
Células-Tronco Embrionárias/enzimologia , Mamíferos/crescimento & desenvolvimento , RNA Polimerase III/metabolismo , Transdução de Sinais , Fatores de Transcrição/metabolismo , Animais , Ciclo Celular , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Fatores de Crescimento de Fibroblastos/metabolismo , Regulação da Expressão Gênica , Mamíferos/metabolismo , Camundongos , Dados de Sequência Molecular , Regiões Promotoras Genéticas , Fatores de Transcrição/genética
16.
Proc Natl Acad Sci U S A ; 117(5): 2519-2525, 2020 02 04.
Artigo em Inglês | MEDLINE | ID: mdl-31964807

RESUMO

The highly conserved COP9 signalosome (CSN), composed of 8 subunits (Cops1 to Cops8), has been implicated in pluripotency maintenance of human embryonic stem cells (ESCs). Yet, the mechanism for the CSN to regulate pluripotency remains elusive. We previously showed that Cops2, independent of the CSN, is essential for the pluripotency maintenance of mouse ESCs. In this study, we set out to investigate how Cops5 and Cops8 regulate ESC differentiation and tried to establish Cops5 and Cops8 knockout (KO) ESC lines by CRISPR/Cas9. To our surprise, no Cops5 KO ESC clones were identified out of 127 clones, while three Cops8 KO ESC lines were established out of 70 clones. We then constructed an inducible Cops5 KO ESC line. Cops5 KO leads to decreased expression of the pluripotency marker Nanog, proliferation defect, G2/M cell-cycle arrest, and apoptosis of ESCs. Further analysis revealed dual roles of Cops5 in maintaining genomic stability of ESCs. On one hand, Cops5 suppresses the autophagic degradation of Mtch2 to direct cellular metabolism toward glycolysis and minimize reactive oxygen species (ROS) production, thereby reducing endogenous DNA damage. On the other hand, Cops5 is required for high DNA damage repair (DDR) activities in ESCs. Without Cops5, elevated ROS and reduced DDR activities lead to DNA damage accumulation in ESCs. Subsequently, p53 is activated to trigger G2/M arrest and apoptosis. Altogether, our studies reveal an essential role of Cops5 in maintaining genome integrity and self-renewal of ESCs by regulating cellular metabolism and DDR pathways.


Assuntos
Complexo do Signalossomo COP9/metabolismo , Reparo do DNA , Células-Tronco Embrionárias/enzimologia , Instabilidade Genômica , Peptídeo Hidrolases/metabolismo , Animais , Apoptose , Complexo do Signalossomo COP9/genética , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Diferenciação Celular , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Pontos de Checagem da Fase G2 do Ciclo Celular , Técnicas de Inativação de Genes , Camundongos , Proteínas de Transporte da Membrana Mitocondrial/genética , Proteínas de Transporte da Membrana Mitocondrial/metabolismo , Fosforilação Oxidativa , Peptídeo Hidrolases/genética , Espécies Reativas de Oxigênio/metabolismo
17.
Genes Dev ; 29(23): 2420-34, 2015 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-26584619

RESUMO

Telomerase inactivation causes loss of the male germline in worms, fish, and mice, indicating a conserved dependence on telomere maintenance in this cell lineage. Here, using telomerase reverse transcriptase (Tert) reporter mice, we found that very high telomerase expression is a hallmark of undifferentiated spermatogonia, the mitotic population where germline stem cells reside. We exploited these high telomerase levels as a basis for purifying undifferentiated spermatogonia using fluorescence-activated cell sorting. Telomerase levels in undifferentiated spermatogonia and embryonic stem cells are comparable and much greater than in somatic progenitor compartments. Within the germline, we uncovered an unanticipated gradient of telomerase activity that also enables isolation of more mature populations. Transcriptomic comparisons of Tert(High) undifferentiated spermatogonia and Tert(Low) differentiated spermatogonia by RNA sequencing reveals marked differences in cell cycle and key molecular features of each compartment. Transplantation studies show that germline stem cell activity is confined to the Tert(High) cKit(-) population. Telomere shortening in telomerase knockout strains causes depletion of undifferentiated spermatogonia and eventual loss of all germ cells after undifferentiated spermatogonia drop below a critical threshold. These data reveal that high telomerase expression is a fundamental characteristic of germline stem cells, thus explaining the broad dependence on telomerase for germline immortality in metazoans.


Assuntos
Células-Tronco Adultas/enzimologia , Regulação Enzimológica da Expressão Gênica , Espermatogônias/enzimologia , Telomerase/genética , Telomerase/metabolismo , Animais , Diferenciação Celular/genética , Células-Tronco Embrionárias/enzimologia , Citometria de Fluxo , Técnicas de Introdução de Genes , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Regiões Promotoras Genéticas/genética
18.
Mol Cell ; 53(1): 49-62, 2014 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-24289921

RESUMO

H3K27me3 is deposited at promoters by the preferential association of Polycomb repressive complex 2 (PRC2) with CpG-rich DNA elements regulating development by repressing gene transcription. H3K27 is also present in mono- and dimethylated states; however, the functional roles of H3K27me1 and H3K27me2 deposition remain poorly characterized. Here, we show that PRC2 activity is not only associated with H3K27me3 but also regulates all forms of H3K27 methylation in a spatially defined manner, contributing to different genomic functions in mouse embryonic stem cells. H3K27me1 accumulates within transcribed genes, promotes transcription, and is regulated by Setd2-dependent H3K36me3 deposition. Contrarily, H3K27me2 is present on approximately 70% of total histone H3 and is distributed in large chromatin domains, exerting protective functions by preventing firing of non-cell-type-specific enhancers. Considering that only 5%-10% of deregulated genes in PRC2-deficient cells are direct H3K27me3 targets, our data support an active role for all H3K27 methylated forms in regulating transcription and determining cell identity.


Assuntos
Células-Tronco Embrionárias/enzimologia , Células-Tronco Embrionárias/imunologia , Histona Desmetilases com o Domínio Jumonji/metabolismo , Transcrição Gênica/fisiologia , Animais , Linhagem Celular , Células-Tronco Embrionárias/citologia , Histonas/genética , Histonas/metabolismo , Histona Desmetilases com o Domínio Jumonji/genética , Metilação , Camundongos , Complexo Repressor Polycomb 2/genética , Complexo Repressor Polycomb 2/metabolismo
19.
Mol Cell ; 55(2): 277-90, 2014 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-24981170

RESUMO

Heterochromatin is required to restrict aberrant expression of retrotransposons, but it remains poorly defined due to the underlying repeat-rich sequences. We dissected Suv39h-dependent histone H3 lysine 9 trimethylation (H3K9me3) by genome-wide ChIP sequencing in mouse embryonic stem cells (ESCs). Refined bioinformatic analyses of repeat subfamilies indicated selective accumulation of Suv39h-dependent H3K9me3 at interspersed repetitive elements that cover ∼5% of the ESC epigenome. The majority of the ∼8,150 intact long interspersed nuclear elements (LINEs) and endogenous retroviruses (ERVs), but only a minor fraction of the >1.8 million degenerate and truncated LINEs/ERVs, are enriched for Suv39h-dependent H3K9me3. Transcriptional repression of intact LINEs and ERVs is differentially regulated by Suv39h and other chromatin modifiers in ESCs but governed by DNA methylation in committed cells. These data provide a function for Suv39h-dependent H3K9me3 chromatin to specifically repress intact LINE elements in the ESC epigenome.


Assuntos
Células-Tronco Embrionárias/enzimologia , Retrovirus Endógenos/genética , Inativação Gênica , Histona-Lisina N-Metiltransferase/fisiologia , Histonas/metabolismo , Elementos Nucleotídeos Longos e Dispersos , Metiltransferases/fisiologia , Proteínas Repressoras/fisiologia , Animais , Células Cultivadas , Metilação de DNA , Camundongos , Processamento de Proteína Pós-Traducional
20.
Mol Cell ; 56(4): 564-79, 2014 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-25457166

RESUMO

Primordial germ cells (PGCs) and preimplantation embryos undergo epigenetic reprogramming, which includes comprehensive DNA demethylation. We found that PRMT5, an arginine methyltransferase, translocates from the cytoplasm to the nucleus during this process. Here we show that conditional loss of PRMT5 in early PGCs causes complete male and female sterility, preceded by the upregulation of LINE1 and IAP transposons as well as activation of a DNA damage response. Similarly, loss of maternal-zygotic PRMT5 also leads to IAP upregulation. PRMT5 is necessary for the repressive H2A/H4R3me2s chromatin modification on LINE1 and IAP transposons in PGCs, directly implicating this modification in transposon silencing during DNA hypomethylation. PRMT5 translocates back to the cytoplasm subsequently, to participate in the previously described PIWI-interacting RNA (piRNA) pathway that promotes transposon silencing via de novo DNA remethylation. Thus, PRMT5 is directly involved in genome defense during preimplantation development and in PGCs at the time of global DNA demethylation.


Assuntos
Blastocisto/enzimologia , Metilação de DNA , Instabilidade Genômica , Óvulo/enzimologia , Proteínas Metiltransferases/fisiologia , Espermatozoides/enzimologia , Animais , Apoptose , Blastocisto/citologia , Células Cultivadas , Dano ao DNA , Elementos de DNA Transponíveis , Desenvolvimento Embrionário , Células-Tronco Embrionárias/enzimologia , Feminino , Histonas/metabolismo , Masculino , Camundongos Transgênicos , Processamento de Proteína Pós-Traducional , Proteína-Arginina N-Metiltransferases
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA