Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Mol Cell ; 80(5): 779-795.e10, 2020 12 03.
Artigo em Inglês | MEDLINE | ID: mdl-33207181

RESUMO

Protein aggregates disrupt cellular homeostasis, causing toxicity linked to neurodegeneration. Selective autophagic elimination of aggregates is critical to protein quality control, but how aggregates are selectively targeted for degradation is unclear. We compared the requirements for autophagy receptor proteins: OPTN, NBR1, p62, NDP52, and TAX1BP1 in clearance of proteotoxic aggregates. Endogenous TAX1BP1 is recruited to and required for the clearance of stress-induced aggregates, whereas ectopic expression of TAX1BP1 increases clearance through autophagy, promoting viability of human induced pluripotent stem cell-derived neurons. In contrast, TAX1BP1 depletion sensitizes cells to several forms of aggregate-induced proteotoxicity. Furthermore, TAX1BP1 is more specifically expressed in the brain compared to other autophagy receptor proteins. In vivo, loss of TAX1BP1 results in accumulation of high molecular weight ubiquitin conjugates and premature lipofuscin accumulation in brains of young TAX1BP1 knockout mice. TAX1BP1 mediates clearance of a broad range of cytotoxic proteins indicating therapeutic potential in neurodegenerative diseases.


Assuntos
Proteínas Reguladoras de Apoptose/deficiência , Autofagia , Encéfalo/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/deficiência , Proteínas de Neoplasias/deficiência , Doenças Neurodegenerativas/metabolismo , Agregação Patológica de Proteínas/metabolismo , Animais , Proteínas Reguladoras de Apoptose/metabolismo , Encéfalo/patologia , Feminino , Células HEK293 , Células HeLa , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Lipofuscina/genética , Lipofuscina/metabolismo , Masculino , Camundongos , Camundongos Knockout , Proteínas de Neoplasias/metabolismo , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/patologia , Agregação Patológica de Proteínas/genética , Agregação Patológica de Proteínas/patologia , Ratos , Ratos Sprague-Dawley , Ubiquitina/genética , Ubiquitina/metabolismo
4.
Mol Cell ; 56(3): 360-375, 2014 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-25284222

RESUMO

Phosphorylation is often used to promote protein ubiquitylation, yet we rarely understand quantitatively how ligase activation and ubiquitin (UB) chain assembly are integrated with phosphoregulation. Here we employ quantitative proteomics and live-cell imaging to dissect individual steps in the PINK1 kinase-PARKIN UB ligase mitochondrial control pathway disrupted in Parkinson's disease. PINK1 plays a dual role by phosphorylating PARKIN on its UB-like domain and poly-UB chains on mitochondria. PARKIN activation by PINK1 produces canonical and noncanonical UB chains on mitochondria, and PARKIN-dependent chain assembly is required for accumulation of poly-phospho-UB (poly-p-UB) on mitochondria. In vitro, PINK1 directly activates PARKIN's ability to assemble canonical and noncanonical UB chains and promotes association of PARKIN with both p-UB and poly-p-UB. Our data reveal a feedforward mechanism that explains how PINK1 phosphorylation of both PARKIN and poly-UB chains synthesized by PARKIN drives a program of PARKIN recruitment and mitochondrial ubiquitylation in response to mitochondrial damage.


Assuntos
Mitocôndrias/enzimologia , Poliubiquitina/biossíntese , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação , Retroalimentação Fisiológica , Células HeLa , Humanos , Potencial da Membrana Mitocondrial , Mutação de Sentido Incorreto , Doença de Parkinson/enzimologia , Fosforilação , Proteínas Quinases/metabolismo , Multimerização Proteica , Transporte Proteico , Proteômica , Ubiquitina-Proteína Ligases/genética
5.
Nature ; 524(7565): 309-314, 2015 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-26266977

RESUMO

Protein aggregates and damaged organelles are tagged with ubiquitin chains to trigger selective autophagy. To initiate mitophagy, the ubiquitin kinase PINK1 phosphorylates ubiquitin to activate the ubiquitin ligase parkin, which builds ubiquitin chains on mitochondrial outer membrane proteins, where they act to recruit autophagy receptors. Using genome editing to knockout five autophagy receptors in HeLa cells, here we show that two receptors previously linked to xenophagy, NDP52 and optineurin, are the primary receptors for PINK1- and parkin-mediated mitophagy. PINK1 recruits NDP52 and optineurin, but not p62, to mitochondria to activate mitophagy directly, independently of parkin. Once recruited to mitochondria, NDP52 and optineurin recruit the autophagy factors ULK1, DFCP1 and WIPI1 to focal spots proximal to mitochondria, revealing a function for these autophagy receptors upstream of LC3. This supports a new model in which PINK1-generated phospho-ubiquitin serves as the autophagy signal on mitochondria, and parkin then acts to amplify this signal. This work also suggests direct and broader roles for ubiquitin phosphorylation in other autophagy pathways.


Assuntos
Autofagia/fisiologia , Mitofagia/fisiologia , Proteínas Nucleares/metabolismo , Proteínas Quinases/metabolismo , Fator de Transcrição TFIIIA/metabolismo , Proteína Homóloga à Proteína-1 Relacionada à Autofagia , Proteínas Relacionadas à Autofagia , Proteínas de Transporte/metabolismo , Proteínas de Ciclo Celular , Células HeLa , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas de Membrana/metabolismo , Proteínas de Membrana Transportadoras , Proteínas Associadas aos Microtúbulos/metabolismo , Mitocôndrias/metabolismo , Proteínas Mitocondriais/metabolismo , Modelos Biológicos , Fosforilação , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligases/metabolismo
6.
Nature ; 496(7445): 372-6, 2013 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-23503661

RESUMO

The PARKIN ubiquitin ligase (also known as PARK2) and its regulatory kinase PINK1 (also known as PARK6), often mutated in familial early-onset Parkinson's disease, have central roles in mitochondrial homeostasis and mitophagy. Whereas PARKIN is recruited to the mitochondrial outer membrane (MOM) upon depolarization via PINK1 action and can ubiquitylate porin, mitofusin and Miro proteins on the MOM, the full repertoire of PARKIN substrates--the PARKIN-dependent ubiquitylome--remains poorly defined. Here we use quantitative diGly capture proteomics (diGly) to elucidate the ubiquitylation site specificity and topology of PARKIN-dependent target modification in response to mitochondrial depolarization. Hundreds of dynamically regulated ubiquitylation sites in dozens of proteins were identified, with strong enrichment for MOM proteins, indicating that PARKIN dramatically alters the ubiquitylation status of the mitochondrial proteome. Using complementary interaction proteomics, we found depolarization-dependent PARKIN association with numerous MOM targets, autophagy receptors, and the proteasome. Mutation of the PARKIN active site residue C431, which has been found mutated in Parkinson's disease patients, largely disrupts these associations. Structural and topological analysis revealed extensive conservation of PARKIN-dependent ubiquitylation sites on cytoplasmic domains in vertebrate and Drosophila melanogaster MOM proteins. These studies provide a resource for understanding how the PINK1-PARKIN pathway re-sculpts the proteome to support mitochondrial homeostasis.


Assuntos
Potencial da Membrana Mitocondrial , Mitocôndrias/metabolismo , Membranas Mitocondriais/metabolismo , Proteínas Mitocondriais/metabolismo , Proteoma/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação , Animais , Linhagem Celular , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Humanos , Camundongos , Mitocôndrias/química , Proteínas Quinases/metabolismo , Proteômica
7.
J Cell Biol ; 223(2)2024 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-38059900

RESUMO

Subcellular location and activation of Tank Binding Kinase 1 (TBK1) govern precise progression through mitosis. Either loss of activated TBK1 or its sequestration from the centrosomes causes errors in mitosis and growth defects. Yet, what regulates its recruitment and activation on the centrosomes is unknown. We identified that NAK-associated protein 1 (NAP1) is essential for mitosis, binding to and activating TBK1, which both localize to centrosomes. Loss of NAP1 causes several mitotic and cytokinetic defects due to inactivation of TBK1. Our quantitative phosphoproteomics identified numerous TBK1 substrates that are not only confined to the centrosomes but are also associated with microtubules. Substrate motifs analysis indicates that TBK1 acts upstream of other essential cell cycle kinases like Aurora and PAK kinases. We also identified NAP1 as a TBK1 substrate phosphorylating NAP1 at S318 to promote its degradation by the ubiquitin proteasomal system. These data uncover an important distinct function for the NAP1-TBK1 complex during cell division.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Citocinese , Mitose , Proteínas Serina-Treonina Quinases , Humanos , Ciclo Celular , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo
8.
J Cell Biol ; 220(2)2021 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-33464298

RESUMO

Genome-wide CRISPR screens have transformed our ability to systematically interrogate human gene function, but are currently limited to a subset of cellular phenotypes. We report a novel pooled screening approach for a wider range of cellular and subtle subcellular phenotypes. Machine learning and convolutional neural network models are trained on the subcellular phenotype to be queried. Genome-wide screening then utilizes cells stably expressing dCas9-KRAB (CRISPRi), photoactivatable fluorescent protein (PA-mCherry), and a lentiviral guide RNA (gRNA) pool. Cells are screened by using microscopy and classified by artificial intelligence (AI) algorithms, which precisely identify the genetically altered phenotype. Cells with the phenotype of interest are photoactivated and isolated via flow cytometry, and the gRNAs are identified by sequencing. A proof-of-concept screen accurately identified PINK1 as essential for Parkin recruitment to mitochondria. A genome-wide screen identified factors mediating TFEB relocation from the nucleus to the cytosol upon prolonged starvation. Twenty-one of the 64 hits called by the neural network model were independently validated, revealing new effectors of TFEB subcellular localization. This approach, AI-photoswitchable screening (AI-PS), offers a novel screening platform capable of classifying a broad range of mammalian subcellular morphologies, an approach largely unattainable with current methodologies at genome-wide scale.


Assuntos
Sistemas CRISPR-Cas/genética , Testes Genéticos , Genoma , Imageamento Tridimensional , Inteligência Artificial , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Proteína A de Ligação a Elemento de Resposta do AMP Cíclico/metabolismo , Aprendizado Profundo , Proteínas de Fluorescência Verde , Células HEK293 , Humanos , Modelos Biológicos , Redes Neurais de Computação , Fenótipo , Reprodutibilidade dos Testes , Análise de Célula Única , Máquina de Vetores de Suporte , Ubiquitina-Proteína Ligases/metabolismo , RNA Guia de Sistemas CRISPR-Cas
9.
J Mol Biol ; 432(8): 2510-2524, 2020 04 03.
Artigo em Inglês | MEDLINE | ID: mdl-31689437

RESUMO

Selective autophagy of mitochondria, or mitophagy, refers to the specific removal and degradation of damaged or surplus mitochondria via targeting to the lysosome for destruction. Disruptions in this homeostatic process may contribute to disease. The identification of diverse mitophagic pathways and how selectivity for each of these pathways is conferred is just beginning to be understood. The removal of both damaged and healthy mitochondria under disease and physiological conditions is controlled by either ubiquitin-dependent or receptor-dependent mechanisms. In this review, we will discuss the known types of mitophagy observed in mammals, recent findings related to PINK1/Parkin-mediated mitophagy (which is the most well-studied form of mitophagy), the implications of defective mitophagy to neurodegenerative processes, and unanswered questions inspiring future research that would enhance our understanding of mitochondrial quality control.


Assuntos
Mitocôndrias/patologia , Mitofagia , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/patologia , Proteínas Quinases/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitina/metabolismo , Animais , Humanos , Mitocôndrias/metabolismo , Transdução de Sinais , Proteínas Ubiquitinadas/metabolismo , Ubiquitinação
10.
Cell Rep ; 29(1): 225-235.e5, 2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31577952

RESUMO

PINK1 and Parkin are established mediators of mitophagy, the selective removal of damaged mitochondria by autophagy. PINK1 and Parkin have been proposed to act as tumor suppressors, as loss-of-function mutations are correlated with enhanced tumorigenesis. However, it is unclear how PINK1 and Parkin act in coordination during mitophagy to influence the cell cycle. Here we show that PINK1 and Parkin genetically interact with proteins involved in cell cycle regulation, and loss of PINK1 and Parkin accelerates cell growth. PINK1- and Parkin-mediated activation of TBK1 at the mitochondria during mitophagy leads to a block in mitosis due to the sequestration of TBK1 from its physiological role at centrosomes during mitosis. Our study supports a diverse role for the far-reaching, regulatory effects of mitochondrial quality control in cellular homeostasis and demonstrates that the PINK1/Parkin pathway genetically interacts with the cell cycle, providing a framework for understanding the molecular basis linking PINK1 and Parkin to mitosis.


Assuntos
Ciclo Celular/genética , Mitocôndrias/genética , Mitose/genética , Proteínas Quinases/genética , Proteínas Serina-Treonina Quinases/genética , Ubiquitina-Proteína Ligases/genética , Autofagia/genética , Linhagem Celular Tumoral , Proliferação de Células/genética , Células HCT116 , Células HEK293 , Células HeLa , Homeostase/genética , Humanos , Mitofagia/genética
11.
Sci Signal ; 11(527)2018 04 24.
Artigo em Inglês | MEDLINE | ID: mdl-29692360

RESUMO

In this issue of Science Signaling, Lu et al reveal a role for Parkin-mediated mitophagy in beige-to-white adipocyte transition. In the absence of Parkin, mitochondria in thermogenic beige adipocytes are preserved even after the elimination of cold mimetic stimuli-in contrast to their typical elimination during the white transition.


Assuntos
Adipócitos Bege , Tecido Adiposo Branco , Mitofagia , Termogênese , Ubiquitina-Proteína Ligases
12.
Elife ; 72018 01 23.
Artigo em Inglês | MEDLINE | ID: mdl-29360040

RESUMO

Damaged mitochondria are selectively eliminated by mitophagy. Parkin and PINK1, gene products mutated in familial Parkinson's disease, play essential roles in mitophagy through ubiquitination of mitochondria. Cargo ubiquitination by E3 ubiquitin ligase Parkin is important to trigger selective autophagy. Although autophagy receptors recruit LC3-labeled autophagic membranes onto damaged mitochondria, how other essential autophagy units such as ATG9A-integrated vesicles are recruited remains unclear. Here, using mammalian cultured cells, we demonstrate that RABGEF1, the upstream factor of the endosomal Rab GTPase cascade, is recruited to damaged mitochondria via ubiquitin binding downstream of Parkin. RABGEF1 directs the downstream Rab proteins, RAB5 and RAB7A, to damaged mitochondria, whose associations are further regulated by mitochondrial Rab-GAPs. Furthermore, depletion of RAB7A inhibited ATG9A vesicle assembly and subsequent encapsulation of the mitochondria by autophagic membranes. These results strongly suggest that endosomal Rab cycles on damaged mitochondria are a crucial regulator of mitophagy through assembling ATG9A vesicles.


Assuntos
Endossomos/enzimologia , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Mitofagia , Ubiquitina-Proteína Ligases/metabolismo , Proteínas rab de Ligação ao GTP/metabolismo , Proteínas rab5 de Ligação ao GTP/metabolismo , Proteínas Relacionadas à Autofagia/metabolismo , Células HeLa , Humanos , Proteínas de Membrana/metabolismo , Mapas de Interação de Proteínas , Proteínas de Transporte Vesicular/metabolismo , proteínas de unión al GTP Rab7
13.
mBio ; 7(4)2016 07 19.
Artigo em Inglês | MEDLINE | ID: mdl-27435465

RESUMO

UNLABELLED: Coxiella burnetii replicates in an acidified lysosome-derived vacuole. Biogenesis of the Coxiella-containing vacuole (CCV) requires bacterial effector proteins delivered into host cells by the Dot/Icm secretion system. Genetic and cell biological analysis revealed that an effector protein called Cig2 promotes constitutive fusion of autophagosomes with the CCV to maintain this compartment in an autolysosomal stage of maturation. This distinguishes the CCV from other pathogen-containing vacuoles that are targeted by the host autophagy pathway, which typically confers host resistance to infection by delivering the pathogen to a toxic lysosomal environment. By maintaining the CCV in an autolysosomal stage of maturation, Cig2 enabled CCV homotypic fusion and enhanced bacterial virulence in the Galleria mellonella (wax moth) model of infection by a mechanism that decreases host tolerance. Thus, C. burnetii residence in an autolysosomal organelle alters host tolerance of infection, which indicates that Cig2-dependent manipulation of a lysosome-derived vacuole influences the host response to infection. IMPORTANCE: Coxiella burnetii is an obligate, intracellular bacterial pathogen that replicates inside a unique, lysosome-like compartment called the Coxiella-containing vacuole (CCV). Over 130 bacterial effector proteins are delivered into the host cell cytosol by the C. burnetii Dot/Icm type IV secretion system. Although the Dot/Icm system is essential for pathogenesis, the functions of most effectors remain unknown. Here we show that the effector protein Cig2 is essential for converting the CCV to an organelle that is similar to the autolysosome. Cig2 function promotes constitutive fusion between the CCV and autophagosomes generated by selective autophagy. Cig2-directed biogenesis of an autolysosomal vacuole is essential for the unique fusogenic properties of the CCV and for virulence in an animal model of disease. This work highlights how bacterial subversion of the host autophagy pathway can influence the cell biological properties of the CCV and influence the host response to infection.


Assuntos
Autofagossomos/metabolismo , Proteínas de Bactérias/metabolismo , Coxiella burnetii/patogenicidade , Interações Hospedeiro-Patógeno , Vacúolos/metabolismo , Vacúolos/microbiologia , Animais , Modelos Animais de Doenças , Resistência à Doença , Infecções por Bactérias Gram-Negativas/imunologia , Infecções por Bactérias Gram-Negativas/microbiologia , Lepidópteros
14.
J Cell Biol ; 205(2): 143-53, 2014 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-24751536

RESUMO

PINK1 kinase activates the E3 ubiquitin ligase Parkin to induce selective autophagy of damaged mitochondria. However, it has been unclear how PINK1 activates and recruits Parkin to mitochondria. Although PINK1 phosphorylates Parkin, other PINK1 substrates appear to activate Parkin, as the mutation of all serine and threonine residues conserved between Drosophila and human, including Parkin S65, did not wholly impair Parkin translocation to mitochondria. Using mass spectrometry, we discovered that endogenous PINK1 phosphorylated ubiquitin at serine 65, homologous to the site phosphorylated by PINK1 in Parkin's ubiquitin-like domain. Recombinant TcPINK1 directly phosphorylated ubiquitin and phospho-ubiquitin activated Parkin E3 ubiquitin ligase activity in cell-free assays. In cells, the phosphomimetic ubiquitin mutant S65D bound and activated Parkin. Furthermore, expression of ubiquitin S65A, a mutant that cannot be phosphorylated by PINK1, inhibited Parkin translocation to damaged mitochondria. These results explain a feed-forward mechanism of PINK1-mediated initiation of Parkin E3 ligase activity.


Assuntos
Proteínas Quinases/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitina/metabolismo , Substituição de Aminoácidos , Animais , Linhagem Celular , Drosophila melanogaster , Ativação Enzimática/fisiologia , Humanos , Mutação de Sentido Incorreto , Fosforilação/fisiologia , Proteínas Quinases/genética , Estrutura Terciária de Proteína , Ubiquitina/genética , Ubiquitina-Proteína Ligases/genética
16.
Dev Cell ; 18(2): 167-8, 2010 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-20159586

RESUMO

The TRF1 subunit of the shelterin complex controls telomere length by regulating telomerase access to chromosome ends. Work from Zeng et al. (2010) now reveals in atomic detail how a battle between the SCF(FBX4) ubiquitin ligase and the shelterin component TIN2 controls TRF1 abundance and telomere length homeostasis.


Assuntos
Telômero/genética , Telômero/metabolismo , Proteína 1 de Ligação a Repetições Teloméricas/metabolismo , Humanos , Modelos Biológicos , Proteínas Ligases SKP Culina F-Box/metabolismo , Complexo Shelterina , Proteínas de Ligação a Telômeros/metabolismo
17.
Mol Cell ; 19(5): 667-78, 2005 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-16137622

RESUMO

DNA methylation is interpreted by a family of methyl-CpG binding domain (MBD) proteins that repress transcription through recruitment of corepressors that modify chromatin. To compare in vivo binding of MeCP2 and MBD2, we analyzed immunoprecipitated chromatin from primary human cells. Genomic sites occupied by the two MBD proteins were mutually exclusive. As MeCP2 was unable to colonize sites vacated by depletion of MBD2, we tested the hypothesis that methyl-CpG alone is insufficient to direct MeCP2 binding. In vitro selection for MeCP2 bound DNA-enriched fragments containing A/T bases ([A/T] > or = 4) adjacent to methyl-CpG. [A/T] > or = 4 was found to be essential for high-affinity binding at selected sites and at known MeCP2 target regions in the Bdnf and Dlx6 genes. MBD2 binding, however, did not require an A/T run. The unexpected restriction of MeCP2 to a defined subset of methyl-CpG sites will facilitate identification of genomic targets that are relevant to Rett Syndrome.


Assuntos
Proteínas Cromossômicas não Histona/metabolismo , Proteínas de Ligação a DNA/metabolismo , DNA/metabolismo , Proteínas Repressoras/metabolismo , Adenina/química , Sequência de Bases , Fator Neurotrófico Derivado do Encéfalo/genética , Linhagem Celular , Linhagem Celular Tumoral , Proteínas Cromossômicas não Histona/genética , Ilhas de CpG/genética , Metilação de DNA , Proteínas de Ligação a DNA/genética , Proteínas de Homeodomínio/genética , Humanos , Proteína 2 de Ligação a Metil-CpG , Mutação , Ligação Proteica , Proteínas Repressoras/genética , Timina/química
18.
Mol Cell ; 15(4): 595-605, 2004 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-15327775

RESUMO

In mammals, heterochromatin is characterized by DNA methylation at CpG dinucleotides and methylation at lysine 9 of histone H3. It is currently unclear whether there is a coordinated transmission of these two epigenetic modifications through DNA replication. Here we show that the methyl-CpG binding protein MBD1 forms a stable complex with histone H3-K9 methylase SETDB1. Moreover, during DNA replication, MBD1 recruits SETDB1 to the large subunit of chromatin assembly factor CAF-1 to form an S phase-specific CAF-1/MBD1/SETDB1 complex that facilitates methylation of H3-K9 during replication-coupled chromatin assembly. In the absence of MBD1, H3-K9 methylation is lost at multiple genomic loci and results in activation of p53BP2 gene, normally repressed by MBD1 in HeLa cells. Our data suggest a model in which H3-K9 methylation by SETDB1 is dependent on MBD1 and is heritably maintained through DNA replication to support the formation of stable heterochromatin at methylated DNA.


Assuntos
Montagem e Desmontagem da Cromatina , Replicação do DNA , Proteínas de Ligação a DNA/metabolismo , Histonas/metabolismo , Lisina/metabolismo , Proteínas Metiltransferases/metabolismo , Proteínas Reguladoras de Apoptose , Proteínas de Transporte/genética , Ilhas de CpG , Proteínas de Ligação a DNA/genética , Inativação Gênica , Células HeLa , Histona-Lisina N-Metiltransferase , Humanos , Substâncias Macromoleculares , Metilação , Modelos Biológicos , Regiões Promotoras Genéticas , Proteínas/metabolismo , RNA Interferente Pequeno/metabolismo , Fase S , Fatores de Transcrição , Técnicas do Sistema de Duplo-Híbrido
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA