Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Epigenetics ; 17(4): 422-443, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-33960278

RESUMO

Ten-Eleven Translocation (TET) proteins convert 5-methylcytosine (5mC) to 5-hydroxymethylcytosine (5hmC) leading to a dynamic epigenetic state of DNA that can influence transcription and chromatin organization. While TET proteins interact with complexes involved in transcriptional repression and activation, the overall understanding of the molecular mechanisms involved in TET-mediated regulation of gene expression still remains limited. Here, we show that TET proteins interact with the chromatin remodelling protein lymphoid-specific helicase (LSH/HELLS) in vivo and in vitro. In mouse embryonic fibroblasts (MEFs) and embryonic stem cells (ESCs) knock out of Lsh leads to a significant reduction of 5-hydroxymethylation amount in the DNA. Whole genome sequencing of 5hmC in wild-type versus Lsh knock-out MEFs and ESCs showed that in absence of Lsh, some regions of the genome gain 5hmC while others lose it, with mild correlation with gene expression changes. We further show that differentially hydroxymethylated regions did not completely overlap with differentially methylated regions indicating that changes in 5hmC distribution upon Lsh knock-out are not a direct consequence of 5mC decrease. Altogether, our results suggest that LSH, which interacts with TET proteins, contributes to the regulation of 5hmC levels and distribution in MEFs and ESCs.


Assuntos
Montagem e Desmontagem da Cromatina , Metilação de DNA , 5-Metilcitosina/metabolismo , Animais , Citosina/metabolismo , DNA/metabolismo , DNA Helicases/metabolismo , Fibroblastos/metabolismo , Genoma , Camundongos
6.
Nucleic Acids Res ; 44(16): 7592-604, 2016 09 19.
Artigo em Inglês | MEDLINE | ID: mdl-27179028

RESUMO

Methylation of DNA at carbon 5 of cytosine is essential for mammalian development and implicated in transcriptional repression of genes and transposons. New patterns of DNA methylation characteristic of lineage-committed cells are established at the exit from pluripotency by de novo DNA methyltransferases enzymes, DNMT3A and DNMT3B, which are regulated by developmental signaling and require access to chromatin-organized DNA. Whether or not the capacity for de novo DNA methylation of developmentally regulated loci is preserved in differentiated somatic cells and can occur in the absence of exogenous signals is currently unknown. Here, we demonstrate that fibroblasts derived from chromatin remodeling ATPase LSH (HELLS)-null mouse embryos, which lack DNA methylation from centromeric repeats, transposons and a number of gene promoters, are capable of reestablishing DNA methylation and silencing of misregulated genes upon re-expression of LSH. We also show that the ability of LSH to bind ATP and the cellular concentration of DNMT3B are critical for cell-autonomous de novo DNA methylation in somatic cells. These data suggest the existence of cellular memory that persists in differentiated cells through many cell generations and changes in transcriptional state.


Assuntos
Adenosina Trifosfatases/metabolismo , DNA Helicases/metabolismo , Metilação de DNA , Fibroblastos/metabolismo , 5-Metilcitosina/metabolismo , Animais , Diferenciação Celular/genética , Linhagem Celular , DNA (Citosina-5-)-Metiltransferases/metabolismo , Embrião de Mamíferos/citologia , Fibroblastos/citologia , Regulação da Expressão Gênica no Desenvolvimento , Inativação Gênica , Camundongos , Mutação/genética , Células NIH 3T3 , Regiões Promotoras Genéticas/genética , Sequências Repetitivas de Ácido Nucleico/genética , Retroelementos/genética , DNA Metiltransferase 3B
7.
Cell Rep ; 15(1): 77-85, 2016 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-27052169

RESUMO

DNA methylation at imprinting control regions (ICRs) is established in gametes in a sex-specific manner and has to be stably maintained during development and in somatic cells to ensure the correct monoallelic expression of imprinted genes. In addition to DNA methylation, the ICRs are marked by allele-specific histone modifications. Whether these marks are essential for maintenance of genomic imprinting is largely unclear. Here, we show that the histone H3 lysine 9 methylases G9a and GLP are required for stable maintenance of imprinted DNA methylation in embryonic stem cells; however, their catalytic activity and the G9a/GLP-dependent H3K9me2 mark are completely dispensable for imprinting maintenance despite the genome-wide loss of non-imprinted DNA methylation in H3K9me2-depleted cells. We provide additional evidence that the G9a/GLP complex protects imprinted DNA methylation by recruitment of de novo DNA methyltransferases, which antagonize TET dioxygenass-dependent erosion of DNA methylation at ICRs.


Assuntos
Metilação de DNA , Células-Tronco Embrionárias/metabolismo , Impressão Genômica , Antígenos de Histocompatibilidade/metabolismo , Histona-Lisina N-Metiltransferase/metabolismo , Linhagem Celular , Antígenos de Histocompatibilidade/genética , Histona-Lisina N-Metiltransferase/genética , Humanos
8.
Adv Exp Med Biol ; 773: 209-44, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24563350

RESUMO

For many years, the nuclear envelope was viewed as a passive barrier that separates the genetic material in the nucleus from the cytoplasm of the cell and permits regulated trafficking of various molecules through the nuclear pores. Research in the past two decades has shown that the nuclear envelope is a complex cellular compartment, which harbors tissue-specific resident proteins, extensively interacts with chromatin and contributes to spatial genome organization and regulation of gene expression. Chromatin at the nuclear periphery is organized into active and silenced domains punctuated by insulator elements. The nuclear envelope transmembrane proteins and the nuclear lamina serve as anchoring sites for heterochromatin. They recruit chromatin that has been modified with specific epigenetic marks, provide silencing factors that add new epigenetic modifications to genes located at the nuclear periphery, and sequester transcription factors away from the nuclear interior. On the other hand, proteins of the nuclear pores anchor as well as help generate active chromatin, promote transcription, and coordinate gene expression with mRNA export. The importance of these functions is underscored by aberrant distribution of peripheral chromatin and changes in gene expression that occur in cancer and heritable human diseases linked to mutations in nuclear envelope proteins. Although many mechanistic questions addressing the role of the nuclear envelope in genome organization and function have been answered in recent years, a great deal remains to be discovered in this exciting and rapidly moving field.


Assuntos
Regulação da Expressão Gênica , Genoma , Membrana Nuclear/fisiologia , Humanos
9.
Nucleic Acids Res ; 42(6): 3529-41, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24371281

RESUMO

Tumourigenic transformation of normal cells into cancer typically involves several steps resulting in acquisition of unlimited growth potential, evasion of apoptosis and non-responsiveness to growth inhibitory signals. Both genetic and epigenetic changes can contribute to cancer development and progression. Given the vast genetic heterogeneity of human cancers and difficulty to monitor cancer-initiating events in vivo, the precise relationship between acquisition of genetic mutations and the temporal progression of epigenetic alterations in transformed cells is largely unclear. Here, we use an in vitro model system to investigate the contribution of cellular immortality and oncogenic transformation of primary human cells to epigenetic reprogramming of DNA methylation and gene expression. Our data demonstrate that extension of replicative life span of the cells is sufficient to induce accumulation of DNA methylation at gene promoters and large-scale changes in gene expression in a time-dependent manner. In contrast, continuous expression of cooperating oncogenes in immortalized cells, although essential for anchorage-independent growth and evasion of apoptosis, does not affect de novo DNA methylation at promoters and induces subtle expression changes. Taken together, these observations imply that cellular immortality promotes epigenetic adaptation to highly proliferative state, whereas transforming oncogenes confer additional properties to transformed human cells.


Assuntos
Transformação Celular Neoplásica , Metilação de DNA , Epigênese Genética , Oncogenes , Animais , Linhagem Celular , Linhagem Celular Transformada , Humanos , Masculino , Camundongos , Células NIH 3T3 , Regiões Promotoras Genéticas
10.
Proc Natl Acad Sci U S A ; 110(51): 20693-8, 2013 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-24297921

RESUMO

The H19 gene controls the expression of several genes within the Imprinted Gene Network (IGN), involved in growth control of the embryo. However, the underlying mechanisms of this control remain elusive. Here, we identified the methyl-CpG-binding domain protein 1 MBD1 as a physical and functional partner of the H19 long noncoding RNA (lncRNA). The H19 lncRNA-MBD1 complex is required for the control of five genes of the IGN. For three of these genes--Igf2 (insulin-like growth factor 2), Slc38a4 (solute carrier family 38 member 4), and Peg1 (paternally expressed gene 1)--both MBD1 and H3K9me3 binding were detected on their differentially methylated regions. The H19 lncRNA-MBD1 complex, through its interaction with histone lysine methyltransferases, therefore acts by bringing repressive histone marks on the differentially methylated regions of these three direct targets of the H19 gene. Our data suggest that, besides the differential DNA methylation found on the differentially methylated regions of imprinted genes, an additional fine tuning of the expressed allele is achieved by a modulation of the H3K9me3 marks, mediated by the association of the H19 lncRNA with chromatin-modifying complexes, such as MBD1. This results in a precise control of the level of expression of growth factors in the embryo.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Embrião de Mamíferos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Redes Reguladoras de Genes/fisiologia , Impressão Genômica/fisiologia , RNA Longo não Codificante/metabolismo , Ribonucleoproteínas/metabolismo , Alelos , Animais , Metilação de DNA/fisiologia , Proteínas de Ligação a DNA/genética , Camundongos , Camundongos Knockout , Ribonucleoproteínas/genética
11.
J Cell Sci ; 125(Pt 22): 5524-34, 2012 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-22946062

RESUMO

LSH, a protein related to the SNF2 family of chromatin-remodelling ATPases, is essential for the correct establishment of DNA methylation levels and patterns in plants and mammalian cells. However, some of the phenotypes resulting from LSH deficiency cannot be explained easily by defects in DNA methylation. Here we show that LSH-deficient mouse and human fibroblasts show reduced viability after exposure to ionizing radiation and repair DNA double-strand breaks less efficiently than wild-type cells. A more detailed characterisation of this phenotype revealed that, in the absence of LSH, the histone variant H2AX is not efficiently phosphorylated in response to DNA damage. This results in impaired recruitment of MDC1 and 53BP1 proteins to DNA double-strand breaks and compromises phosphorylation of checkpoint kinase CHK2. Furthermore, we demonstrate that the ability of LSH to hydrolyse ATP is necessary for efficient phosphorylation of H2AX at DNA double-strand breaks and successful repair of DNA damage. Taken together, our data reveal a previously unsuspected role of LSH ATPase in the maintenance of genome stability in mammalian somatic cells, which is independent of its function in de novo DNA methylation during development.


Assuntos
Adenosina Trifosfatases/metabolismo , Quebras de DNA de Cadeia Dupla , DNA Helicases/metabolismo , Reparo do DNA , Histonas/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Fatores de Transcrição/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Animais , Proteínas de Ciclo Celular , Quinase do Ponto de Checagem 2 , Quebras de DNA de Cadeia Dupla/efeitos da radiação , DNA Helicases/deficiência , Metilação de DNA/efeitos da radiação , Reparo do DNA/efeitos da radiação , Ativação Enzimática/efeitos da radiação , Fibroblastos/metabolismo , Fibroblastos/efeitos da radiação , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Cinética , Mamíferos/metabolismo , Camundongos , Proteínas Mutantes/metabolismo , Fosforilação/efeitos da radiação , Proteínas Serina-Treonina Quinases/metabolismo , Radiação Ionizante , Transdução de Sinais/efeitos da radiação
12.
Dev Cell ; 23(2): 265-79, 2012 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-22841499

RESUMO

X chromosome inactivation involves multiple levels of chromatin modification, established progressively and in a stepwise manner during early development. The chromosomal protein Smchd1 was recently shown to play an important role in DNA methylation of CpG islands (CGIs), a late step in the X inactivation pathway that is required for long-term maintenance of gene silencing. Here we show that inactive X chromosome (Xi) CGI methylation can occur via either Smchd1-dependent or -independent pathways. Smchd1-dependent CGI methylation, the primary pathway, is acquired gradually over an extended period, whereas Smchd1-independent CGI methylation occurs rapidly after the onset of X inactivation. The de novo methyltransferase Dnmt3b is required for methylation of both classes of CGI, whereas Dnmt3a and Dnmt3L are dispensable. Xi CGIs methylated by these distinct pathways differ with respect to their sequence characteristics and immediate chromosomal environment. We discuss the implications of these results for understanding CGI methylation during development.


Assuntos
Proteínas Cromossômicas não Histona/metabolismo , Ilhas de CpG , Metilação de DNA , Inativação do Cromossomo X , Alelos , Animais , Linhagem Celular , Proteínas Cromossômicas não Histona/genética , Camundongos , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo
13.
Biochem Biophys Res Commun ; 411(1): 19-24, 2011 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-21722626

RESUMO

Translation elongation isoform eEF1A1 has a pivotal role in protein synthesis and is almost ubiquitously expressed. In mice and rats that transcription of the gene encoding eEF1A1 is downregulated to undetectable levels in muscle after weaning; eEF1A1 is then replaced by a separately encoded but closely related isoform eEF1A2, which has only previously been described in mammals. We now show that not only is eEF1A2 conserved in non-mammalian vertebrate species, but the down-regulation of eEF1A1 protein in muscle is preserved in Xenopus, with the protein being undetectable by adulthood. Interestingly, though, this down-regulation is controlled post-transcriptionally, and levels of full-length eEF1A1 mRNA remain similar to those of eEF1A2. The switching off of eEF1A1 in muscle is therefore sufficiently important to have evolved through the use of repression operating at different levels in different species. The 3'UTR of eEF1A1 is highly conserved and contains predicted binding sites for several miRNAs, suggesting a possible method for controlling of expression. We suggest that isoform switching may have evolved because of a need for certain cell types to modify the well-established non-canonical functions of eEF1A1.


Assuntos
Músculo Esquelético/metabolismo , Elongação Traducional da Cadeia Peptídica , Fator 1 de Elongação de Peptídeos/metabolismo , Xenopus laevis/metabolismo , Regiões 3' não Traduzidas , Sequência de Aminoácidos , Animais , Sequência Conservada , Regulação para Baixo , Evolução Molecular , Feminino , Camundongos , Dados de Sequência Molecular , Desenvolvimento Muscular/genética , Músculo Esquelético/citologia , Músculo Esquelético/crescimento & desenvolvimento , Fator 1 de Elongação de Peptídeos/genética , Ratos , Xenopus laevis/genética , Xenopus laevis/crescimento & desenvolvimento
15.
Prog Mol Biol Transl Sci ; 101: 377-98, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21507359

RESUMO

DNA methylation is a stable epigenetic mark in plant and vertebrate genomes; it is implicated in regulation of higher order chromatin structure, maintenance of genome integrity, and stable patterns of gene expression. Biological effects of DNA methylation are, at least in part, mediated by proteins that preferentially bind to methylated DNA. It is now recognized that several structurally unrelated protein folds have the ability to recognize methylated CpGs in vitro and in vivo. In this chapter, we focus on the three major families of methyl-CpG-binding proteins: the MBD protein family, Kaiso and Kaiso-like proteins, and SRA domain proteins. We discuss the structural bases of methyl-CpG recognition, the function and specific properties of individual proteins, and their role in human disease such as Rett syndrome and cancer.


Assuntos
Ilhas de CpG , Proteínas de Ligação a DNA/fisiologia , Animais , Metilação de DNA , Humanos
16.
Genome Res ; 21(1): 83-94, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21149390

RESUMO

LSH, a member of the SNF2 family of chromatin remodeling ATPases encoded by the Hells gene, is essential for normal levels of DNA methylation in the mammalian genome. While the role of LSH in the methylation of repetitive DNA sequences is well characterized, its contribution to the regulation of DNA methylation and the expression of protein-coding genes has not been studied in detail. In this report we investigate genome-wide patterns of DNA methylation at gene promoters in Hells(-/-) mouse embryonic fibroblasts (MEFs). We find that in the absence of LSH, DNA methylation is lost or significantly reduced at ∼20% of all normally methylated promoter sequences. As a consequence, a large number of genes are misexpressed in Hells(-/-) MEFs. Comparison of Hells(-/-) MEFs with wild-type MEFs and embryonic stem (ES) cells suggests that LSH is important for de novo DNA methylation events that accompany the establishment and differentiation of embryonic lineage cells. We further show that the generation of normal DNA methylation patterns and stable gene silencing at specific promoters require cooperation between LSH and the G9a/GLP complex of histone methylases. At such loci, G9a recruitment is compromised when LSH is absent or greatly reduced. Taken together, our data suggest a mechanism whereby LSH promotes binding of DNA methyltransferases and the G9a/GLP complex to specific loci and facilitates developmentally programmed DNA methylation and stable gene silencing during lineage commitment and differentiation.


Assuntos
DNA Helicases/metabolismo , Metilação de DNA , Regulação da Expressão Gênica no Desenvolvimento , Histona-Lisina N-Metiltransferase/metabolismo , Animais , Diferenciação Celular , Células Cultivadas , DNA Helicases/genética , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Fibroblastos/metabolismo , Inativação Gênica , Histona-Lisina N-Metiltransferase/genética , Metiltransferases/genética , Metiltransferases/metabolismo , Camundongos
17.
Nucleic Acids Res ; 38(14): 4620-34, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20378711

RESUMO

MBD1, a member of the methyl-CpG-binding domain family of proteins, has been reported to repress transcription of methylated and unmethylated promoters. As some MBD1 isoforms contain two DNA-binding domains-an MBD, which recognizes methylated DNA; and a CXXC3 zinc finger, which binds unmethylated CpG-it is unclear whether these two domains function independently of each other or if they cooperate in facilitating recruitment of MBD1 to particular genomic loci. In this report we investigate DNA-binding specificity of MBD and CXXC3 domains in vitro and in vivo. We find that the methyl-CpG-binding domain of MBD1 binds more efficiently to methylated DNA within a specific sequence context. We identify genes that are targeted by MBD1 in human cells and demonstrate that a functional MBD domain is necessary and sufficient for recruitment of MBD1 to specific sites at these loci, while DNA binding by the CXXC3 motif is largely dispensable. In summary, the binding preferences of MBD1, although dependent upon the presence of methylated DNA, are clearly distinct from those of other methyl-CpG-binding proteins, MBD2 and MeCP2.


Assuntos
Metilação de DNA , Proteínas de Ligação a DNA/metabolismo , Fatores de Transcrição/metabolismo , Linhagem Celular , Ilhas de CpG , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/genética , Inativação Gênica , Células HeLa , Proteína Vmw65 do Vírus do Herpes Simples/metabolismo , Humanos , Mutação Puntual , Regiões Promotoras Genéticas , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Receptores de Fator de Crescimento Neural/genética , Fatores de Transcrição/química , Fatores de Transcrição/genética , Proteínas rho de Ligação ao GTP/genética
18.
PLoS One ; 5(4): e9937, 2010 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-20376339

RESUMO

Differentiation of embryonic stem (ES) cells is accompanied by silencing of the Oct-4 gene and de novo DNA methylation of its regulatory region. Previous studies have focused on the requirements for promoter region methylation. We therefore undertook to analyse the progression of DNA methylation of the approximately 2000 base pair regulatory region of Oct-4 in ES cells that are wildtype or deficient for key proteins. We find that de novo methylation is initially seeded at two discrete sites, the proximal enhancer and distal promoter, spreading later to neighboring regions, including the remainder of the promoter. De novo methyltransferases Dnmt3a and Dnmt3b cooperate in the initial targeted stage of de novo methylation. Efficient completion of the pattern requires Dnmt3a and Dnmt1, but not Dnmt3b. Methylation of the Oct-4 promoter depends on the histone H3 lysine 9 methyltransferase G9a, as shown previously, but CpG methylation throughout most of the regulatory region accumulates even in the absence of G9a. Analysis of the Oct-4 regulatory domain as a whole has allowed us to detect targeted de novo methylation and to refine our understanding the roles of key protein components in this process.


Assuntos
Diferenciação Celular/genética , Metilação de DNA , Células-Tronco Embrionárias/citologia , Fator 3 de Transcrição de Octâmero/fisiologia , Sequências Reguladoras de Ácido Nucleico/genética , Animais , Linhagem Celular , Ilhas de CpG , DNA (Citosina-5-)-Metiltransferases/metabolismo , Camundongos , Regiões Promotoras Genéticas
19.
Mol Cell Biol ; 28(1): 215-26, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17967891

RESUMO

LSH, a protein related to the SNF2 family of chromatin-remodeling ATPases, is required for efficient DNA methylation in mammals. How LSH functions to support DNA methylation and whether it associates with a large protein complex containing DNA methyltransferase (DNMT) enzymes is currently unclear. Here we show that, unlike many other chromatin-remodeling ATPases, native LSH is present mostly as a monomeric protein in nuclear extracts of mammalian cells and cannot be detected in a large multisubunit complex. However, when targeted to a promoter of a reporter gene, LSH acts as an efficient transcriptional repressor. Using this as an assay to identify proteins that are required for LSH-mediated repression we found that LSH cooperates with the DNMTs DNMT1 and DNMT3B and with the histone deacetylases (HDACs) HDAC1 and HDAC2 to silence transcription. We show that transcriptional repression by LSH and interactions with HDACs are lost in DNMT1 and DNMT3B knockout cells but that the enzymatic activities of DNMTs are not required for LSH-mediated silencing. Our data suggest that LSH serves as a recruiting factor for DNMTs and HDACs to establish transcriptionally repressive chromatin which is perhaps further stabilized by DNA methylation at targeted loci.


Assuntos
DNA Helicases/metabolismo , Metilases de Modificação do DNA/metabolismo , Transcrição Gênica/genética , Linhagem Celular , Núcleo Celular/metabolismo , DNA (Citosina-5-)-Metiltransferases/genética , DNA (Citosina-5-)-Metiltransferases/metabolismo , DNA Helicases/genética , Metilases de Modificação do DNA/genética , Histona Desacetilases/genética , Histona Desacetilases/metabolismo , Humanos , Ligação Proteica , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , DNA Metiltransferase 3B
20.
EMBO J ; 25(22): 5317-28, 2006 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-17066076

RESUMO

In mammalian cells, DNA methylation is associated with heritable and stable gene repression, mediated in part by methyl-CpG-binding domain (MBD) proteins that recruit corepressors to modify chromatin. MBD1 protein, a member of the MBD family, forms a complex with SETDB1 histone methylase to silence transcription at target promoters by methylation of lysine 9 of histone H3. How MBD1-mediated transcriptional repression is regulated is currently unknown. Here we show that MBD1 is a target for sumoylation by PIAS1 (Protein Inhibitors of Activated STAT 1) and PIAS3 E3 SUMO (small ubiquitin-like modifier)-ligases, at two conserved lysine residues within the C-terminus of MBD1. Although sumoylated MBD1 binds to methylated DNA, it does not incorporate into a complex with SETDB1 and does not efficiently repress transcription of a target gene, p53BP2, in HeLa cells. Our data suggest that transcriptional silencing by MBD1 is regulated by a PIAS-mediated conjugation of SUMO1, which antagonizes the formation of a repressive complex with SETDB1.


Assuntos
Proteínas de Ligação a DNA/fisiologia , Chaperonas Moleculares/fisiologia , Proteínas Inibidoras de STAT Ativados/fisiologia , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/fisiologia , Fatores de Transcrição/fisiologia , Transcrição Gênica , Sequência de Aminoácidos , Proteínas Reguladoras de Apoptose , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Proteínas de Transporte/fisiologia , Cromatina/genética , Cromatina/fisiologia , Sequência Conservada , Metilação de DNA , Proteínas de Ligação a DNA/metabolismo , Regulação para Baixo , Inativação Gênica , Células HeLa , Histona-Lisina N-Metiltransferase , Histonas/metabolismo , Humanos , Chaperonas Moleculares/genética , Dados de Sequência Molecular , Ligação Proteica , Proteínas Inibidoras de STAT Ativados/genética , Proteínas Metiltransferases/metabolismo , Proteína SUMO-1 , Homologia de Sequência de Aminoácidos , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/genética , Fatores de Transcrição/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...