Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Mol Cell ; 67(5): 853-866.e5, 2017 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-28803779

RESUMO

Lysine crotonylation (Kcr) is a newly identified histone modification that is associated with active transcription in mammalian cells. Here we report that the chromodomain Y-like transcription corepressor CDYL negatively regulates histone Kcr by acting as a crotonyl-CoA hydratase to convert crotonyl-CoA to ß-hydroxybutyryl-CoA. We showed that the negative regulation of histone Kcr by CDYL is intrinsically linked to its transcription repression activity and functionally implemented in the reactivation of sex chromosome-linked genes in round spermatids and genome-wide histone replacement in elongating spermatids. Significantly, Cdyl transgenic mice manifest dysregulation of histone Kcr and reduction of male fertility with a decreased epididymal sperm count and sperm cell motility. Our study uncovers a biochemical pathway in the regulation of histone Kcr and implicates CDYL-regulated histone Kcr in spermatogenesis, adding to the understanding of the physiology of male reproduction and the mechanism of the spermatogenic failure in AZFc (Azoospermia Factor c)-deleted infertile men.


Assuntos
Acil Coenzima A/metabolismo , Proteínas Correpressoras/metabolismo , Enoil-CoA Hidratase/metabolismo , Histona Acetiltransferases/metabolismo , Histonas/metabolismo , Infertilidade Masculina/enzimologia , Processamento de Proteína Pós-Traducional , Proteínas/metabolismo , Espermatogênese , Espermatozoides/enzimologia , Testículo/enzimologia , Animais , Proteínas Correpressoras/genética , Enoil-CoA Hidratase/genética , Fertilidade , Predisposição Genética para Doença , Células HeLa , Histona Acetiltransferases/genética , Humanos , Hidroliases , Infertilidade Masculina/genética , Infertilidade Masculina/patologia , Infertilidade Masculina/fisiopatologia , Cinética , Lisina , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fenótipo , Domínios Proteicos , Proteínas/genética , Interferência de RNA , Células Sf9 , Contagem de Espermatozoides , Motilidade dos Espermatozoides , Espermatozoides/patologia , Testículo/patologia , Testículo/fisiopatologia , Transfecção
2.
Development ; 147(21)2020 07 17.
Artigo em Inglês | MEDLINE | ID: mdl-32541010

RESUMO

Kabuki syndrome (KS) is a congenital craniofacial disorder resulting from mutations in the KMT2D histone methylase (KS1) or the UTX histone demethylase (KS2). With small cohorts of KS2 patients, it is not clear whether differences exist in clinical manifestations relative to KS1. We mutated KMT2D in neural crest cells (NCCs) to study cellular and molecular functions in craniofacial development with respect to UTX. Similar to UTX, KMT2D NCC knockout mice demonstrate hypoplasia with reductions in frontonasal bone lengths. We have traced the onset of KMT2D and UTX mutant NCC frontal dysfunction to a stage of altered osteochondral progenitor differentiation. KMT2D NCC loss-of-function does exhibit unique phenotypes distinct from UTX mutation, including fully penetrant cleft palate, mandible hypoplasia and deficits in cranial base ossification. KMT2D mutant NCCs lead to defective secondary palatal shelf elevation with reduced expression of extracellular matrix components. KMT2D mutant chondrocytes in the cranial base fail to properly differentiate, leading to defective endochondral ossification. We conclude that KMT2D is required for appropriate cranial NCC differentiation and KMT2D-specific phenotypes may underlie differences between Kabuki syndrome subtypes.


Assuntos
Anormalidades Múltiplas/enzimologia , Anormalidades Múltiplas/patologia , Diferenciação Celular , Face/anormalidades , Doenças Hematológicas/enzimologia , Doenças Hematológicas/patologia , Histona-Lisina N-Metiltransferase/metabolismo , Proteína de Leucina Linfoide-Mieloide/metabolismo , Crista Neural/enzimologia , Crista Neural/patologia , Doenças Vestibulares/enzimologia , Doenças Vestibulares/patologia , Alelos , Animais , Linhagem da Célula , Movimento Celular , Condrócitos/patologia , Face/patologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Morfogênese , Mutação/genética , Osteogênese , Palato/embriologia , Palato/metabolismo , Palato/patologia , Fenótipo , Crânio/patologia
3.
Genes Dev ; 29(6): 672-85, 2015 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-25792601

RESUMO

Loss of function/dysregulation of inhibitor of growth 4 (ING4) and hyperactivation of NF-κB are frequent events in many types of human malignancies. However, the molecular mechanisms underlying these remarkable aberrations are not understood. Here, we report that ING4 is physically associated with JFK. We demonstrated that JFK targets ING4 for ubiquitination and degradation through assembly of an Skp1-Cul1-F-box (SCF) complex. We showed that JFK-mediated ING4 destabilization leads to the hyperactivation of the canonical NF-κB pathway and promotes angiogenesis and metastasis of breast cancer. Significantly, the expression of JFK is markedly up-regulated in breast cancer, and the level of JFK is negatively correlated with that of ING4 and positively correlated with an aggressive clinical behavior of breast carcinomas. Our study identified SCF(JFK) as a bona fide E3 ligase for ING4 and unraveled the JFK-ING4-NF-κB axis as an important player in the development and progression of breast cancer, supporting the pursuit of JFK as a potential target for breast cancer intervention.


Assuntos
Neoplasias da Mama/enzimologia , Neoplasias da Mama/fisiopatologia , Proteínas de Ciclo Celular/metabolismo , Proteínas F-Box/metabolismo , Proteínas de Homeodomínio/metabolismo , Neovascularização Patológica/enzimologia , Proteínas Supressoras de Tumor/metabolismo , Neoplasias da Mama/irrigação sanguínea , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Células MCF-7 , Complexos Multiproteicos , NF-kappa B/metabolismo , Metástase Neoplásica , Neovascularização Patológica/genética , Proteólise , Transdução de Sinais , Ubiquitinação
4.
J Biol Chem ; 292(44): 18113-18128, 2017 11 03.
Artigo em Inglês | MEDLINE | ID: mdl-28878014

RESUMO

The de novo assembly and post-splicing reassembly of the U4/U6.U5 tri-snRNP remain to be investigated. We report here that ZIP, a protein containing a CCCH-type zinc finger and a G-patch domain, as characterized by us previously, regulates pre-mRNA splicing independent of RNA binding. We found that ZIP physically associates with the U4/U6.U5 tri-small nuclear ribonucleoprotein (tri-snRNP). Remarkably, the ZIP-containing tri-snRNP, which has a sedimentation coefficient of ∼35S, is a tri-snRNP that has not been described previously. We also found that the 35S tri-snRNP contains hPrp24, indicative of a state in which the U4/U6 di-snRNP is integrating with the U5 snRNP. We found that the 35S tri-snRNP is enriched in the Cajal body, indicating that it is an assembly intermediate during 25S tri-snRNP maturation. We showed that the 35S tri-snRNP also contains hPrp43, in which ATPase/RNA helicase activities are stimulated by ZIP. Our study identified, for the first time, a tri-snRNP intermediate, shedding new light on the de novo assembly and recycling of the U4/U6.U5 tri-snRNP.


Assuntos
Processamento Alternativo , Antígenos de Neoplasias/metabolismo , Biogênese de Organelas , RNA Helicases/metabolismo , Proteínas de Ligação a RNA/metabolismo , Proteínas Repressoras/metabolismo , Spliceossomos/metabolismo , Proteases Específicas de Ubiquitina/metabolismo , Antígenos de Neoplasias/química , Antígenos de Neoplasias/genética , Corpos Enovelados/química , Corpos Enovelados/enzimologia , Corpos Enovelados/metabolismo , Células HeLa , Humanos , Imunoprecipitação , Células MCF-7 , Microscopia Eletrônica de Transmissão , Microscopia de Fluorescência , Peso Molecular , Mutação , Coloração Negativa , Oligopeptídeos/genética , Oligopeptídeos/metabolismo , Multimerização Proteica , Estabilidade Proteica , RNA Helicases/química , RNA Helicases/genética , Proteínas de Ligação a RNA/química , Proteínas de Ligação a RNA/genética , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Repressoras/química , Proteínas Repressoras/genética , Ribonucleoproteína Nuclear Pequena U5/química , Ribonucleoproteína Nuclear Pequena U5/metabolismo , Spliceossomos/química , Spliceossomos/enzimologia , Proteases Específicas de Ubiquitina/química , Proteases Específicas de Ubiquitina/genética
5.
FASEB J ; 28(11): 4821-34, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25063848

RESUMO

ATP-binding cassette (ABC) transporters are implicated in a diverse range of physiological and pathophysiological processes, such as cholesterol and lipid transportation and multidrug resistance. Despite the considerable efforts made in understanding of the cellular function of ABC proteins, the regulation mechanism of this type of protein is still poorly defined. Here we report the identification and functional characterization of a novel ATPase protein, protein associated with ABC transporters (PAAT), in humans. PAAT contains a nucleotide-binding domain (NBD)-like domain and a signal for intramitochondrial sorting. We showed that PAAT is localized in both the cytoplasm and the mitochondria and has an intrinsic ATPase activity. PAAT physically interacts with the 3 known mitochondrial inner membrane ABC proteins, ABCB7, ABCB8, and ABCB10, but not ABCB1, ABCB6, or ABCG2, and functionally regulates the transport of ferric nutrients and heme biosynthesis. Significantly, PAAT deficiency promotes cell death, reduces mitochondrial potential, and sensitizes mitochondria to oxidative stress-induced DNA damages. Our experiments revealed that PAAT is a novel ATPase and a trans-regulator of mitochondrial ABC transporters that plays an important role in the maintenance of mitochondrial homeostasis and cell survival.


Assuntos
Sistema A de Transporte de Aminoácidos/metabolismo , Homeostase/fisiologia , Mitocôndrias/metabolismo , Células Cultivadas , Humanos , Ligação Proteica , Transporte Proteico/fisiologia
6.
J Mol Biol ; 436(7): 168453, 2024 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-38266981

RESUMO

Histone-lysine N-methyltransferase 2 (KMT2) methyltransferases are critical for gene regulation, cell differentiation, animal development, and human diseases. KMT2 biological roles are often attributed to their methyltransferase activities on lysine 4 of histone H3 (H3K4). However, recent data indicate that KMT2 proteins also possess non-enzymatic functions. In this review, we discuss the current understanding of KMT2 family, with a focus on their enzymatic activity-dependent and -independent functions. Six mammalian KMT2 proteins of three subgroups, KMT2A/B (MLL1/2), KMT2C/D (MLL3/4), and KMT2F/G (SETD1A/B or SET1A/B), have shared and distinct protein domains, catalytic substrates, genomic localizations, and associated complex subunits. Recent studies have revealed the importance of KMT2C/D in enhancer regulation, differentiation, development, tumor suppression and highlighted KMT2C/D enzymatic activity-dependent and -independent roles in mouse embryonic development and cell differentiation. Catalytic dependent and independent functions for KMT2A/B and KMT2F/G in gene regulation, differentiation, and development are less understood. Finally, we provide our perspectives and lay out future research directions that may help advance the investigation on enzymatic activity-dependent and -independent biological roles and working mechanisms of KMT2 methyltransferases.


Assuntos
Histona-Lisina N-Metiltransferase , Histonas , Animais , Humanos , Camundongos , Diferenciação Celular/genética , Desenvolvimento Embrionário/genética , Regulação da Expressão Gênica , Histonas/metabolismo , Domínios Proteicos , Histona-Lisina N-Metiltransferase/química , Histona-Lisina N-Metiltransferase/genética , Histona-Lisina N-Metiltransferase/metabolismo , Neoplasias/genética
7.
Nat Commun ; 15(1): 4883, 2024 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-38849395

RESUMO

The human methyltransferase and transcriptional coactivator MLL4 and its paralog MLL3 are frequently mutated in cancer. MLL4 and MLL3 monomethylate histone H3K4 and contain a set of uncharacterized PHD fingers. Here, we report a novel function of the PHD2 and PHD3 (PHD2/3) fingers of MLL4 and MLL3 that bind to ASXL2, a component of the Polycomb repressive H2AK119 deubiquitinase (PR-DUB) complex. The structure of MLL4 PHD2/3 in complex with the MLL-binding helix (MBH) of ASXL2 and mutational analyses reveal the molecular mechanism which is conserved in homologous ASXL1 and ASXL3. The native interaction of the Trithorax MLL3/4 complexes with the PR-DUB complex in vivo depends solely on MBH of ASXL1/2, coupling the two histone modifying activities. ChIP-seq analysis in embryonic stem cells demonstrates that MBH of ASXL1/2 is required for the deubiquitinase BAP1 recruitment to MLL4-bound active enhancers. Our findings suggest an ASXL1/2-dependent functional link between the MLL3/4 and PR-DUB complexes.


Assuntos
Proteínas de Ligação a DNA , Histona-Lisina N-Metiltransferase , Ligação Proteica , Proteínas Repressoras , Proteínas Supressoras de Tumor , Ubiquitina Tiolesterase , Proteínas Supressoras de Tumor/metabolismo , Proteínas Supressoras de Tumor/genética , Humanos , Histona-Lisina N-Metiltransferase/metabolismo , Histona-Lisina N-Metiltransferase/genética , Ubiquitina Tiolesterase/metabolismo , Ubiquitina Tiolesterase/genética , Proteínas Repressoras/metabolismo , Proteínas Repressoras/genética , Animais , Proteínas de Ligação a DNA/metabolismo , Proteínas de Ligação a DNA/genética , Camundongos , Elementos Facilitadores Genéticos , Células HEK293 , Dedos de Zinco PHD , Histonas/metabolismo
8.
J Biol Chem ; 287(25): 21045-57, 2012 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-22547059

RESUMO

Ccr4d is a new member of the Ccr4 (carbon catabolite repression 4) family of proteins that are implicated in the regulation of mRNA stability and translation through mRNA deadenylation. However, Ccr4d is not believed to be involved in mRNA deadenylation. Thus, its biological function and mechanistic activity remain to be determined. Here, we report that Ccr4d is broadly expressed in various normal tissues, and the expression of Ccr4d is markedly down-regulated during cell cycle progression. We showed that Ccr4d inhibits cell proliferation and induces cell cycle arrest at G(1) phase. Our experiments further revealed that Ccr4d regulates the expression of p21 in a p53-independent manner. Mechanistic studies indicated that Ccr4d strongly bound to the 3'-UTR of p21 mRNA, leading to the stabilization of p21 mRNA. Interestingly, we found that the expression of Ccr4d is down-regulated in various tumor tissues. Collectively, our data indicate that Ccr4d functions as an anti-proliferating protein through the induction of cell cycle arrest via a p21-dependent and p53-independent pathway and suggest that Ccr4d might have an important role in carcinogenesis.


Assuntos
Pontos de Checagem do Ciclo Celular/fisiologia , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Proteínas Nucleares/metabolismo , Processamento Pós-Transcricional do RNA/fisiologia , Estabilidade de RNA/fisiologia , Fatores de Transcrição/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Regiões 3' não Traduzidas/fisiologia , Linhagem Celular Tumoral , Inibidor de Quinase Dependente de Ciclina p21/genética , Regulação para Baixo/fisiologia , Humanos , Proteínas Nucleares/genética , Fatores de Transcrição/genética , Proteína Supressora de Tumor p53/genética
9.
Nat Genet ; 55(4): 693-705, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-37012455

RESUMO

H3K4me1 methyltransferases MLL3 (KMT2C) and MLL4 (KMT2D) are critical for enhancer activation, cell differentiation and development. However, roles of MLL3/4 enzymatic activities and MLL3/4-mediated enhancer H3K4me1 in these processes remain unclear. Here we report that constitutive elimination of both MLL3 and MLL4 enzymatic activities prevents initiation of gastrulation and leads to early embryonic lethality in mice. However, selective elimination of MLL3/4 enzymatic activities in embryonic, but not extraembryonic, lineages leaves gastrulation largely intact. Consistent with this, embryonic stem cells (ESCs) lacking MLL3/4 enzymatic activities can differentiate toward the three embryonic germ layers but show aberrant differentiation to extraembryonic endoderm (ExEn) and trophectoderm. The failure in ExEn differentiation can be attributed to markedly reduced enhancer-binding of the lineage-determining transcription factor GATA6. Furthermore, we show that MLL3/4-catalyzed H3K4me1 is largely dispensable for enhancer activation during ESC differentiation. Together, our findings suggest a lineage-selective, but enhancer activation-independent, role of MLL3/4 methyltransferase activities in early embryonic development and ESC differentiation.


Assuntos
Desenvolvimento Embrionário , Histona-Lisina N-Metiltransferase , Animais , Camundongos , Diferenciação Celular/genética , Desenvolvimento Embrionário/genética , Células-Tronco Embrionárias , Histona-Lisina N-Metiltransferase/genética
10.
J Biol Chem ; 286(49): 42414-42425, 2011 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-22009739

RESUMO

Polycomb group proteins play essential roles in transcriptional regulation of multiple gene families involved in various pathophysiological processes. It is believed that Polycomb Repressive Complex 2 (PRC2) is targeted to chromatin by the EED subunit to methylate histone H3 lysine 27 (H3K27), leading to a repressive chromatin state that inhibits gene expression. Here we report that the chromodomain-containing protein CDYL specifically recognizes di- and tri-methylated H3K27 (H3K27me2 and H3K27me3) and directly interacts with EZH2, the catalytic subunit of PRC2. We show that CDYL dramatically enhances the methyltransferase activity of PRC2 toward oligonucleosome substrates in vitro. Genome-wide analysis of CDYL targets by ChIP sequencing revealed that CDYL and PRC2 share a number of genomic targets. CDYL is required for chromatin targeting and maximal enzymatic activity of PRC2 at their common target sites. Our experiments indicate that CDYL functions as a molecular bridge between PRC2 and the repressive chromatin mark H3K27me3, forming a positive feedback loop to facilitate the establishment and propagation of H3K27me3 modifications along the chromatin.


Assuntos
Histonas/química , Proteínas/fisiologia , Proteínas Repressoras/química , Animais , Domínio Catalítico , Linhagem Celular Tumoral , Cromatina/química , Proteínas Correpressoras , Epigênese Genética , Regulação da Expressão Gênica , Humanos , Hidroliases , Lisina/química , Metilação , Metiltransferases/química , Nucleossomos/química , Proteínas do Grupo Polycomb , Proteínas/química , Xenopus
11.
Science ; 377(6606): 666-669, 2022 08 05.
Artigo em Inglês | MEDLINE | ID: mdl-35926054

RESUMO

Muscle stem cells (MuSCs) reside in a specialized niche that ensures their regenerative capacity. Although we know that innate immune cells infiltrate the niche in response to injury, it remains unclear how MuSCs adapt to this altered environment for initiating repair. Here, we demonstrate that inflammatory cytokine signaling from the regenerative niche impairs the ability of quiescent MuSCs to reenter the cell cycle. The histone H3 lysine 27 (H3K27) demethylase JMJD3, but not UTX, allowed MuSCs to overcome inhibitory inflammation signaling by removing trimethylated H3K27 (H3K27me3) marks at the Has2 locus to initiate production of hyaluronic acid, which in turn established an extracellular matrix competent for integrating signals that direct MuSCs to exit quiescence. Thus, JMJD3-driven hyaluronic acid synthesis plays a proregenerative role that allows MuSC adaptation to inflammation and the initiation of muscle repair.


Assuntos
Ácido Hialurônico , Inflamação , Histona Desmetilases com o Domínio Jumonji , Músculo Esquelético , Mioblastos Esqueléticos , Regeneração , Nicho de Células-Tronco , Animais , Ciclo Celular , Histonas , Humanos , Ácido Hialurônico/biossíntese , Inflamação/metabolismo , Interferon gama/metabolismo , Interleucina-6 , Histona Desmetilases com o Domínio Jumonji/genética , Histona Desmetilases com o Domínio Jumonji/metabolismo , Camundongos , Músculo Esquelético/lesões , Músculo Esquelético/fisiologia , Mioblastos Esqueléticos/metabolismo
12.
J Mol Cell Biol ; 9(3): 178-194, 2017 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-28402439

RESUMO

Faithful transmission or restoration of epigenetic information such as repressive histone modifications through generations is critical for the maintenance of cell identity. We report here that chromodomain Y-like protein (CDYL), a chromodomain-containing transcription corepressor, is physically associated with chromatin assembly factor 1 (CAF-1) and the replicative helicase MCM complex. We showed that CDYL bridges CAF-1 and MCM, facilitating histone transfer and deposition during DNA replication. We demonstrated that CDYL recruits histone-modifying enzymes G9a, SETDB1, and EZH2 to replication forks, leading to the addition of H3K9me2/3 and H3K27me2/3 on newly deposited histone H3. Significantly, depletion of CDYL impedes early S phase progression and sensitizes cells to DNA damage. Our data indicate that CDYL plays an important role in the transmission/restoration of repressive histone marks, thereby preserving the epigenetic landscape for the maintenance of cell identity.


Assuntos
Replicação do DNA , Histonas/metabolismo , Proteínas/metabolismo , Cromatina/metabolismo , Proteínas Correpressoras , Dano ao DNA , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Epigênese Genética , Histona-Lisina N-Metiltransferase , Histonas/genética , Humanos , Hidroliases , Lisina/metabolismo , Proteínas de Manutenção de Minicromossomo/metabolismo , Proteínas Metiltransferases/metabolismo , Proteínas/genética , Fase S/fisiologia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
13.
Nat Commun ; 8(1): 691, 2017 09 25.
Artigo em Inglês | MEDLINE | ID: mdl-28947780

RESUMO

EGFR is required for animal development, and dysregulation of EGFR is critically implicated in malignant transformation. However, the molecular mechanism underlying the regulation of EGFR expression remains poorly explored. Here we report that the zinc-finger protein ZNF516 is a transcription repressor. ZNF516 is physically associated with the CtBP/LSD1/CoREST complex and transcriptionally represses a cohort of genes including EGFR that are critically involved in cell proliferation and motility. We demonstrate that the ZNF516-CtBP/LSD1/CoREST complex inhibits the proliferation and invasion of breast cancer cells in vitro and suppresses breast cancer growth and metastasis in vivo. Significantly, low expression of ZNF516 is positively associated with advanced pathological staging and poor survival of breast carcinomas. Our data indicate that ZNF516 is a transcription repressor and a potential suppressor of EGFR, adding to the understanding of EGFR-related breast carcinogenesis and supporting the pursuit of ZNF516 as a potential therapeutic target for breast cancer. EGFR is a well-known oncogene; however, the mechanisms regulating its expression are still unclear. Here, analysing genome-wide chromatin associations, the authors show that in breast cancer cells ZNF516 represses EGFR transcription through the interaction with the CtBP/LSD1/CoREST complex.


Assuntos
Neoplasias da Mama/genética , Cromatina/metabolismo , Proteínas de Ligação a DNA/fisiologia , Receptores ErbB/genética , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Proliferação de Células/genética , Proteínas Correpressoras/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Receptores ErbB/metabolismo , Feminino , Células HEK293 , Histona Desmetilases/metabolismo , Humanos , Estimativa de Kaplan-Meier , Modelos Genéticos , Invasividade Neoplásica/genética , Proteínas do Tecido Nervoso/metabolismo , Transporte Proteico
14.
Nat Genet ; 48(9): 1003-13, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27500525

RESUMO

FOXA1 functions in epigenetic reprogramming and is described as a 'pioneer factor'. However, exactly how FOXA1 achieves these remarkable biological functions is not fully understood. Here we report that FOXA1 associates with DNA repair complexes and is required for genomic targeting of DNA polymerase ß (POLB) in human cells. Genome-wide DNA methylomes demonstrate that the FOXA1 DNA repair complex is functionally linked to DNA demethylation in a lineage-specific fashion. Depletion of FOXA1 results in localized reestablishment of methylation in a large portion of FOXA1-bound regions, and the regions with the most consistent hypermethylation exhibit the greatest loss of POLB and are represented by active promoters and enhancers. Consistently, overexpression of FOXA1 commits its binding sites to active DNA demethylation in a POLB-dependent manner. Finally, FOXA1-associated DNA demethylation is tightly coupled with estrogen receptor genomic targeting and estrogen responsiveness. Together, these results link FOXA1-associated DNA demethylation to transcriptional pioneering by FOXA1.


Assuntos
Metilação de DNA , DNA Polimerase beta/metabolismo , Enzimas Reparadoras do DNA/genética , Reparo do DNA/genética , Epigenômica , Fator 3-alfa Nuclear de Hepatócito/genética , Neoplasias/genética , Transcrição Gênica , Sítios de Ligação , Enzimas Reparadoras do DNA/metabolismo , Estrogênios/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Células HeLa , Fator 3-alfa Nuclear de Hepatócito/metabolismo , Humanos , Células MCF-7 , Neoplasias/metabolismo , Regiões Promotoras Genéticas/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA