Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 81
Filtrar
1.
Cell ; 179(6): 1342-1356.e23, 2019 11 27.
Artículo en Inglés | MEDLINE | ID: mdl-31759698

RESUMEN

Mammalian switch/sucrose non-fermentable (mSWI/SNF) complexes are multi-component machines that remodel chromatin architecture. Dissection of the subunit- and domain-specific contributions to complex activities is needed to advance mechanistic understanding. Here, we examine the molecular, structural, and genome-wide regulatory consequences of recurrent, single-residue mutations in the putative coiled-coil C-terminal domain (CTD) of the SMARCB1 (BAF47) subunit, which cause the intellectual disability disorder Coffin-Siris syndrome (CSS), and are recurrently found in cancers. We find that the SMARCB1 CTD contains a basic α helix that binds directly to the nucleosome acidic patch and that all CSS-associated mutations disrupt this binding. Furthermore, these mutations abrogate mSWI/SNF-mediated nucleosome remodeling activity and enhancer DNA accessibility without changes in genome-wide complex localization. Finally, heterozygous CSS-associated SMARCB1 mutations result in dominant gene regulatory and morphologic changes during iPSC-neuronal differentiation. These studies unmask an evolutionarily conserved structural role for the SMARCB1 CTD that is perturbed in human disease.


Asunto(s)
Ensamble y Desensamble de Cromatina/genética , Proteínas Cromosómicas no Histona/metabolismo , Mutación/genética , Nucleosomas/metabolismo , Proteína SMARCB1/genética , Factores de Transcripción/metabolismo , Secuencia de Aminoácidos , Elementos de Facilitación Genéticos/genética , Femenino , Genoma Humano , Células HEK293 , Células HeLa , Heterocigoto , Humanos , Masculino , Modelos Moleculares , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Unión Proteica , Dominios Proteicos , Proteína SMARCB1/química , Proteína SMARCB1/metabolismo
2.
Immunity ; 57(3): 462-477.e9, 2024 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-38430908

RESUMEN

Inducible nucleosome remodeling at hundreds of latent enhancers and several promoters shapes the transcriptional response to Toll-like receptor 4 (TLR4) signaling in macrophages. We aimed to define the identities of the transcription factors that promote TLR-induced remodeling. An analysis strategy based on ATAC-seq and single-cell ATAC-seq that enriched for genomic regions most likely to undergo remodeling revealed that the transcription factor nuclear factor κB (NF-κB) bound to all high-confidence peaks marking remodeling during the primary response to the TLR4 ligand, lipid A. Deletion of NF-κB subunits RelA and c-Rel resulted in the loss of remodeling at high-confidence ATAC-seq peaks, and CRISPR-Cas9 mutagenesis of NF-κB-binding motifs impaired remodeling. Remodeling selectivity at defined regions was conferred by collaboration with other inducible factors, including IRF3- and MAP-kinase-induced factors. Thus, NF-κB is unique among TLR4-activated transcription factors in its broad contribution to inducible nucleosome remodeling, alongside its ability to activate poised enhancers and promoters assembled into open chromatin.


Asunto(s)
FN-kappa B , Receptor Toll-Like 4 , FN-kappa B/metabolismo , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo , Nucleosomas , Transducción de Señal , Regulación de la Expresión Génica , Factor de Transcripción ReIA/metabolismo
3.
Mol Cell ; 84(3): 476-489.e10, 2024 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-38211589

RESUMEN

Pioneer transcription factors (TFs) regulate cell fate by establishing transcriptionally primed and active states. However, cell fate control requires the coordination of both lineage-specific gene activation and repression of alternative-lineage programs, a process that is poorly understood. Here, we demonstrate that the pioneer TF FOXA coordinates with PRDM1 TF to recruit nucleosome remodeling and deacetylation (NuRD) complexes and Polycomb repressive complexes (PRCs), which establish highly occupied, accessible nucleosome conformation with bivalent epigenetic states, thereby preventing precocious and alternative-lineage gene expression during human endoderm differentiation. Similarly, the pioneer TF OCT4 coordinates with PRDM14 to form bivalent enhancers and repress cell differentiation programs in human pluripotent stem cells, suggesting that this may be a common and critical function of pioneer TFs. We propose that pioneer and PRDM TFs coordinate to safeguard cell fate through epigenetic repression mechanisms.


Asunto(s)
Nucleosomas , Factores de Transcripción , Humanos , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Nucleosomas/genética , Diferenciación Celular/genética , Proteínas del Grupo Polycomb/metabolismo , Epigénesis Genética
4.
Genes Dev ; 33(9-10): 565-577, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-30808655

RESUMEN

Heterochromatin protein 1 (HP1) proteins are key factors of eukaryotic heterochromatin that coordinate chromatin compaction and transcriptional gene silencing. Through their multivalency they act as adaptors between histone H3 Lys9 di/trimethyl marks in chromatin and effector complexes that bind to the HP1 chromoshadow domain. Most organisms encode for multiple HP1 isoforms and the molecular mechanisms that underpin their diverse functions in genome regulation remain poorly understood. In fission yeast, the two HP1 proteins Chp2 and Swi6 assume distinct roles and Chp2 is tightly associated with the nucleosome remodeling and deacetylation complex SHREC. Here we show that Chp2 directly engages the SHREC nucleosome remodeler subunit Mit1. The crystal structure of the interaction interface reveals an extraordinarily extensive and specific interaction between the chromoshadow domain of Chp2 and the N terminus of Mit1. The integrity of this interface is critical for high affinity binding and for heterochromatin formation. Comparison with Swi6 shows that the Chp2-Mit1 interface is highly selective and thereby provides the molecular basis for the functional specialization of an HP1 isoform.


Asunto(s)
Regulación Fúngica de la Expresión Génica , Silenciador del Gen , Proteínas Represoras/metabolismo , Proteínas de Schizosaccharomyces pombe/metabolismo , Schizosaccharomyces/genética , Cristalización , Heterocromatina/metabolismo , Unión Proteica , Isoformas de Proteínas , Proteínas Represoras/química , Proteínas Represoras/genética , Proteínas de Schizosaccharomyces pombe/química , Proteínas de Schizosaccharomyces pombe/genética
5.
Mol Cell ; 72(4): 661-672.e4, 2018 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-30392927

RESUMEN

Regular successions of positioned nucleosomes, or phased nucleosome arrays (PNAs), are predominantly known from transcriptional start sites (TSSs). It is unclear whether PNAs occur elsewhere in the genome. To generate a comprehensive inventory of PNAs for Drosophila, we applied spectral analysis to nucleosome maps and identified thousands of PNAs throughout the genome. About half of them are not near TSSs and are strongly enriched for an uncharacterized sequence motif. Through genome-wide reconstitution of physiological chromatin in Drosophila embryo extracts, we uncovered the molecular basis of PNA formation. We identified Phaser, an unstudied zinc finger protein that positions nucleosomes flanking the motif. It also revealed how the global activity of the chromatin remodelers CHRAC/ACF, together with local barrier elements, generates islands of regular phasing throughout the genome. Our work demonstrates the potential of chromatin assembly by embryo extracts as a powerful tool to reconstitute chromatin features on a global scale in vitro.


Asunto(s)
Ensamble y Desensamble de Cromatina/genética , Drosophila melanogaster/genética , Nucleosomas/genética , Animales , Cromatina/fisiología , Ensamble y Desensamble de Cromatina/fisiología , Mapeo Cromosómico/métodos , Drosophila/genética , Histonas , Ratones , Nucleosomas/fisiología , Sitio de Iniciación de la Transcripción/fisiología
6.
Proc Natl Acad Sci U S A ; 119(42): e2211568119, 2022 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-36215487

RESUMEN

Eukaryotic DNA replication is initiated at multiple chromosomal sites known as origins of replication that are specifically recognized by the origin recognition complex (ORC) containing multiple ATPase sites. In budding yeast, ORC binds to specific DNA sequences known as autonomously replicating sequences (ARSs) that are mostly nucleosome depleted. However, nucleosomes may still inhibit the licensing of some origins by occluding ORC binding and subsequent MCM helicase loading. Using purified proteins and single-molecule visualization, we find here that the ORC can eject histones from a nucleosome in an ATP-dependent manner. The ORC selectively evicts H2A-H2B dimers but leaves the (H3-H4)2 tetramer on DNA. It also discriminates canonical H2A from the H2A.Z variant, evicting the former while retaining the latter. Finally, the bromo-adjacent homology (BAH) domain of the Orc1 subunit is essential for ORC-mediated histone eviction. These findings suggest that the ORC is a bona fide nucleosome remodeler that functions to create a local chromatin environment optimal for origin activity.


Asunto(s)
Nucleosomas , Complejo de Reconocimiento del Origen , Adenosina Trifosfatasas/metabolismo , Adenosina Trifosfato , Cromatina , ADN/metabolismo , Replicación del ADN , Histonas/metabolismo , Nucleosomas/genética , Complejo de Reconocimiento del Origen/metabolismo , Origen de Réplica
7.
J Med Virol ; 96(2): e29436, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38380509

RESUMEN

Kaposi sarcoma (KS), caused by Herpesvirus-8 (HHV-8; KSHV), shows sporadic, endemic, and epidemic forms. While familial clustering of KS was previously recorded, the molecular basis of hereditary predilection to KS remains largely unknown. We demonstrate through genetic studies that a dominantly inherited missense mutation in BPTF segregates with a phenotype of classical KS in multiple immunocompetent individuals in two families. Using an rKSHV.219-infected CRISPR/cas9-model, we show that BPTFI2012T mutant cells exhibit higher latent-to-lytic ratio, decreased virion production, increased LANA staining, and latent phenotype in viral transcriptomics. RNA-sequencing demonstrated that KSHV infection dysregulated oncogenic-like response and P53 pathways, MAPK cascade, and blood vessel development pathways, consistent with KS. BPTFI2012T also enriched pathways of viral genome regulation and replication, immune response, and chemotaxis, including downregulation of IFI16, SHFL HLAs, TGFB1, and HSPA5, all previously associated with KSHV infection and tumorigenesis. Many of the differentially expressed genes are regulated by Rel-NF-κB, which regulates immune processes, cell survival, and proliferation and is pivotal to oncogenesis. We thus demonstrate BPTF mutation-mediated monogenic hereditary predilection of KSHV virus-induced oncogenesis, and suggest BPTF as a drug target.


Asunto(s)
Herpesvirus Humano 8 , Sarcoma de Kaposi , Humanos , Carcinogénesis , Herpesvirus Humano 8/fisiología , FN-kappa B/metabolismo , Sarcoma de Kaposi/genética , Latencia del Virus/genética , Replicación Viral
8.
J Exp Bot ; 75(14): 4332-4345, 2024 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-38436409

RESUMEN

Chromatin regulation in eukaryotes plays pivotal roles in controlling the developmental regulatory gene network. This review explores the intricate interplay between chromatin regulators and environmental signals, elucidating their roles in shaping plant development. As sessile organisms, plants have evolved sophisticated mechanisms to perceive and respond to environmental cues, orchestrating developmental programs that ensure adaptability and survival. A central aspect of this dynamic response lies in the modulation of versatile gene regulatory networks, mediated in part by various chromatin regulators. Here, we summarized current understanding of the molecular mechanisms through which chromatin regulators integrate environmental signals, influencing key aspects of plant development.


Asunto(s)
Cromatina , Desarrollo de la Planta , Cromatina/metabolismo , Desarrollo de la Planta/genética , Regulación de la Expresión Génica de las Plantas , Plantas/metabolismo , Plantas/genética , Transducción de Señal , Redes Reguladoras de Genes , Ambiente
9.
Biol Res ; 57(1): 22, 2024 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-38704609

RESUMEN

BACKGROUND: Chromatin dynamics is deeply involved in processes that require access to DNA, such as transcriptional regulation. Among the factors involved in chromatin dynamics at gene regulatory regions are general regulatory factors (GRFs). These factors contribute to establishment and maintenance of nucleosome-depleted regions (NDRs). These regions are populated by nucleosomes through histone deposition and nucleosome sliding, the latter catalyzed by a number of ATP-dependent chromatin remodeling complexes, including ISW1a. It has been observed that GRFs can act as barriers against nucleosome sliding towards NDRs. However, the relative ability of the different GRFs to hinder sliding activity is currently unknown. RESULTS: Considering this, we performed a comparative analysis for the main GRFs, with focus in their ability to modulate nucleosome sliding mediated by ISW1a. Among the GRFs tested in nucleosome remodeling assays, Rap1 was the only factor displaying the ability to hinder the activity of ISW1a. This effect requires location of the Rap1 cognate sequence on linker that becomes entry DNA in the nucleosome remodeling process. In addition, Rap1 was able to hinder nucleosome assembly in octamer transfer assays. Concurrently, Rap1 displayed the highest affinity for and longest dwell time from its target sequence, compared to the other GRFs tested. Consistently, through bioinformatics analyses of publicly available genome-wide data, we found that nucleosome occupancy and histone deposition in vivo are inversely correlated with the affinity of Rap1 for its target sequences in the genome. CONCLUSIONS: Our findings point to DNA binding affinity, residence time and location at particular translational positions relative to the nucleosome core as the key features of GRFs underlying their roles played in nucleosome sliding and assembly.


Asunto(s)
Ensamble y Desensamble de Cromatina , Proteínas de Unión al ADN , Nucleosomas , Nucleosomas/metabolismo , Nucleosomas/genética , Ensamble y Desensamble de Cromatina/fisiología , Adenosina Trifosfatasas/metabolismo , Adenosina Trifosfatasas/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Histonas/metabolismo
10.
Proc Natl Acad Sci U S A ; 118(36)2021 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-34465625

RESUMEN

The SNF2 family ATPase Amplified in Liver Cancer 1 (ALC1) is the only chromatin remodeling enzyme with a poly(ADP-ribose) (PAR) binding macrodomain. ALC1 functions together with poly(ADP-ribose) polymerase PARP1 to remodel nucleosomes. Activation of ALC1 cryptic ATPase activity and the subsequent nucleosome remodeling requires binding of its macrodomain to PAR chains synthesized by PARP1 and NAD+ A key question is whether PARP1 has a role(s) in ALC1-dependent nucleosome remodeling beyond simply synthesizing the PAR chains needed to activate the ALC1 ATPase. Here, we identify PARP1 separation-of-function mutants that activate ALC1 ATPase but do not support nucleosome remodeling by ALC1. Investigation of these mutants has revealed multiple functions for PARP1 in ALC1-dependent nucleosome remodeling and provides insights into its multifaceted role in chromatin remodeling.


Asunto(s)
ADN Helicasas/metabolismo , Proteínas de Unión al ADN/metabolismo , Nucleosomas/metabolismo , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Línea Celular Tumoral , Ensamble y Desensamble de Cromatina , Reparación del ADN , Humanos
11.
Proc Natl Acad Sci U S A ; 117(46): 28847-28858, 2020 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-33127760

RESUMEN

CHD7 encodes an ATP-dependent chromatin remodeling factor. Mutation of this gene causes multiple developmental disorders, including CHARGE (Coloboma of the eye, Heart defects, Atresia of the choanae, Retardation of growth/development, Genital abnormalities, and Ear anomalies) syndrome, in which conotruncal anomalies are the most prevalent form of heart defects. How CHD7 regulates conotruncal development remains unclear. In this study, we establish that deletion of Chd7 in neural crest cells (NCCs) causes severe conotruncal defects and perinatal lethality, thus providing mouse genetic evidence demonstrating that CHD7 cell-autonomously regulates cardiac NCC development, thereby clarifying a long-standing controversy in the literature. Using transcriptomic analyses, we show that CHD7 fine-tunes the expression of a gene network that is critical for cardiac NCC development. To gain further molecular insights into gene regulation by CHD7, we performed a protein-protein interaction screen by incubating recombinant CHD7 on a protein array. We find that CHD7 directly interacts with several developmental disorder-mutated proteins including WDR5, a core component of H3K4 methyltransferase complexes. This direct interaction suggested that CHD7 may recruit histone-modifying enzymes to target loci independently of its remodeling functions. We therefore generated a mouse model that harbors an ATPase-deficient allele and demonstrates that mutant CHD7 retains the ability to recruit H3K4 methyltransferase activity to its targets. Thus, our data uncover that CHD7 regulates cardiovascular development through ATP-dependent and -independent activities, shedding light on the etiology of CHD7-related congenital disorders. Importantly, our data also imply that patients carrying a premature stop codon versus missense mutations will likely display different molecular alterations; these patients might therefore require personalized therapeutic interventions.


Asunto(s)
Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Corazón/embriología , Adenosina Trifosfato/metabolismo , Alelos , Animales , Síndrome CHARGE/genética , Ensamble y Desensamble de Cromatina/genética , ADN Helicasas/metabolismo , Modelos Animales de Enfermedad , Embrión de Mamíferos/metabolismo , Regulación del Desarrollo de la Expresión Génica/genética , Cardiopatías Congénitas/genética , Ratones , Ratones Noqueados , Mutación , Cresta Neural/embriología , Cresta Neural/metabolismo , Organogénesis/fisiología
12.
Int J Mol Sci ; 24(5)2023 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-36902382

RESUMEN

Chromatin remodeling by ATP-dependent remodeling enzymes is crucial for all genomic processes, like transcription or replication. Eukaryotes harbor many remodeler types, and it is unclear why a given chromatin transition requires more or less stringently one or several remodelers. As a classical example, removal of budding yeast PHO8 and PHO84 promoter nucleosomes upon physiological gene induction by phosphate starvation essentially requires the SWI/SNF remodeling complex. This dependency on SWI/SNF may indicate specificity in remodeler recruitment, in recognition of nucleosomes as remodeling substrate or in remodeling outcome. By in vivo chromatin analyses of wild type and mutant yeast under various PHO regulon induction conditions, we found that overexpression of the remodeler-recruiting transactivator Pho4 allowed removal of PHO8 promoter nucleosomes without SWI/SNF. For PHO84 promoter nucleosome removal in the absence of SWI/SNF, an intranucleosomal Pho4 site, which likely altered the remodeling outcome via factor binding competition, was required in addition to such overexpression. Therefore, an essential remodeler requirement under physiological conditions need not reflect substrate specificity, but may reflect specific recruitment and/or remodeling outcomes.


Asunto(s)
Nucleosomas , Proteínas de Saccharomyces cerevisiae , Cromatina/metabolismo , Ensamble y Desensamble de Cromatina , Nucleosomas/metabolismo , Regiones Promotoras Genéticas , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo
13.
Biochem Soc Trans ; 50(2): 907-919, 2022 04 29.
Artículo en Inglés | MEDLINE | ID: mdl-35356970

RESUMEN

Structural and biochemical studies have identified a histone surface on each side of the nucleosome disk termed 'the nucleosome acidic patch' that acts as a regulatory hub for the function of numerous nuclear proteins, including ATP-dependent chromatin complexes (remodelers). Four major remodeler subfamilies, SWI/SNF, ISWI, CHD, and INO80, have distinct modes of interaction with one or both nucleosome acidic patches, contributing to their specific remodeling outcomes. Genome-wide sequencing analyses of various human cancers have uncovered high-frequency mutations in histone coding genes, including some that map to the acidic patch. How cancer-related acidic patch histone mutations affect nucleosome remodeling is mainly unknown. Recent advances in in vitro chromatin reconstitution have enabled access to physiologically relevant nucleosomes, including asymmetric nucleosomes that possess both wild-type and acidic patch mutant histone copies. Biochemical investigation of these substrates revealed unexpected remodeling outcomes with far-reaching implications for alteration of chromatin structure. This review summarizes recent findings of how different remodeler families interpret wild-type and mutant acidic patches for their remodeling functions and discusses models for remodeler-mediated changes in chromatin landscapes as a consequence of acidic patch mutations.


Asunto(s)
Histonas , Nucleosomas , Adenosina Trifosfatasas/metabolismo , Cromatina , Ensamble y Desensamble de Cromatina , Histonas/metabolismo , Humanos , Mutación
14.
Arch Toxicol ; 96(6): 1739-1749, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35362730

RESUMEN

Epigenetic complex NuRD (nucleosome remodeling and deacetylase) engages in a range of basic cellular processes, including chromatin modification. Changes in the activity of NuRD complex can influence gastric cancer progression. Multivariate logistic regression analyses were used to estimate the association between single-nucleotide polymorphisms (SNPs) and gastric cancer risk. Expression quantitative trait loci (eQTL) analysis was used to analyze the relationship between the genotypes and gene expression levels using data from the genotype tissue expression project (GTEx). Gene expression was calculated using databases from The Cancer Genome Atlas (TCGA) and The Gene Expression Omnibus (GEO). Kaplan-Meier plotter was used to evaluate the association between gene expression and survival. SNP rs11064275 T allele in CHD4, rs892022 A allele and rs2033481 A allele in GATAD2A were found to contribute to the decreased risk of gastric cancer. The increase in the number of favorable alleles of these three SNPs was associated with a lower risk of gastric cancer. rs2033481 and rs892022 were substantially correlated with GATAD2A mRNA expression levels. Meanwhile, we detected that the CHD4 and GATAD2A mRNA expression was increased in gastric cancer tissues compared with the adjacent normal tissues. Furthermore, we found that patients with higher CHD4 or GATAD2A mRNA expression level had more advantageous overall survival. Our findings indicated that genetic variants in NuRD complex subunits encoding genes may be promising predictors of gastric cancer risk.


Asunto(s)
Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2 , Neoplasias Gástricas , Humanos , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2/genética , Nucleosomas/genética , ARN Mensajero , Neoplasias Gástricas/genética
15.
Genes Dev ; 28(7): 672-82, 2014 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-24696452

RESUMEN

Eukaryotic gene regulation involves a balance between packaging of the genome into nucleosomes and enabling access to regulatory proteins and RNA polymerase. Nucleosomes are integral components of gene regulation that restrict access to both regulatory sequences and the underlying template. Whereas canonical histones package the newly replicated genome, they can be replaced with histone variants that alter nucleosome structure, stability, dynamics, and, ultimately, DNA accessibility. Here we consider how histone variants and their interacting partners are involved in transcriptional regulation through the creation of unique chromatin states.


Asunto(s)
Regulación de la Expresión Génica , Histonas/genética , Transcripción Genética/genética , Animales , Cromatina/metabolismo , Histonas/metabolismo , Nucleosomas/metabolismo
16.
Bioessays ; 41(9): e1900091, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31379012

RESUMEN

Here, the emerging data on DNA G-quadruplexes (G4s) as epigenetic modulators are reviewed and integrated. This concept has appeared and evolved substantially in recent years. First, persistent G4s (e.g., those stabilized by exogenous ligands) were linked to the loss of the histone code. More recently, transient G4s (i.e., those formed upon replication or transcription and unfolded rapidly by helicases) were implicated in CpG island methylation maintenance and de novo CpG methylation control. The most recent data indicate that there are direct interactions between G4s and chromatin remodeling factors. Finally, multiple findings support the indirect participation of G4s in chromatin reshaping via interactions with remodeling-related transcription factors (TFs) or damage responders. Here, the links between the above processes are analyzed; also, how further elucidation of these processes may stimulate the progress of epigenetic therapy is discussed, and the remaining open questions are highlighted.


Asunto(s)
Ensamble y Desensamble de Cromatina/genética , Epigénesis Genética , G-Cuádruplex , Histonas/genética , Islas de CpG , Metilación de ADN , Código de Histonas , Histonas/metabolismo , Humanos , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
17.
Proc Natl Acad Sci U S A ; 115(5): E876-E885, 2018 01 30.
Artículo en Inglés | MEDLINE | ID: mdl-29339483

RESUMEN

Mutations in CDCA7, the SNF2 family protein HELLS (LSH), or the DNA methyltransferase DNMT3b cause immunodeficiency-centromeric instability-facial anomalies (ICF) syndrome. While it has been speculated that DNA methylation defects cause this disease, little is known about the molecular function of CDCA7 and its functional relationship to HELLS and DNMT3b. Systematic analysis of how the cell cycle, H3K9 methylation, and the mitotic kinase Aurora B affect proteomic profiles of chromatin in Xenopus egg extracts revealed that HELLS and CDCA7 form a stoichiometric complex on chromatin, in a manner sensitive to Aurora B. Although HELLS alone fails to remodel nucleosomes, we demonstrate that the HELLS-CDCA7 complex possesses nucleosome remodeling activity. Furthermore, CDCA7 is essential for loading HELLS onto chromatin, and CDCA7 harboring patient ICF mutations fails to recruit the complex to chromatin. Together, our study identifies a unique bipartite nucleosome remodeling complex where the functional remodeling activity is split between two proteins and thus delineates the defective pathway in ICF syndrome.


Asunto(s)
ADN Helicasas/metabolismo , Cara/anomalías , Síndromes de Inmunodeficiencia/genética , Mutación , Proteínas Nucleares/metabolismo , Animales , Aurora Quinasa B/metabolismo , Ciclo Celular , Cromatina/química , Análisis por Conglomerados , ADN (Citosina-5-)-Metiltransferasas/genética , ADN Helicasas/genética , Metilación de ADN , Células HeLa , Histonas/metabolismo , Humanos , Proteínas Nucleares/genética , Nucleosomas/química , Óvulo/metabolismo , Péptidos/química , Enfermedades de Inmunodeficiencia Primaria , Unión Proteica , Dominios Proteicos , Proteómica , Interferencia de ARN , Xenopus laevis , ADN Metiltransferasa 3B
18.
Proc Natl Acad Sci U S A ; 115(26): 6727-6732, 2018 06 26.
Artículo en Inglés | MEDLINE | ID: mdl-29891665

RESUMEN

Cardiac development relies on proper cardiomyocyte differentiation, including expression and assembly of cell-type-specific actomyosin subunits into a functional cardiac sarcomere. Control of this process involves not only promoting expression of cardiac sarcomere subunits but also repressing expression of noncardiac myofibril paralogs. This level of transcriptional control requires broadly expressed multiprotein machines that modify and remodel the chromatin landscape to restrict transcription machinery access. Prominent among these is the nucleosome remodeling and deacetylase (NuRD) complex, which includes the catalytic core subunit CHD4. Here, we demonstrate that direct CHD4-mediated repression of skeletal and smooth muscle myofibril isoforms is required for normal cardiac sarcomere formation, function, and embryonic survival early in gestation. Through transcriptomic and genome-wide analyses of CHD4 localization, we identified unique CHD4 binding sites in smooth muscle myosin heavy chain, fast skeletal α-actin, and the fast skeletal troponin complex genes. We further demonstrate that in the absence of CHD4, cardiomyocytes in the developing heart form a hybrid muscle cell that contains cardiac, skeletal, and smooth muscle myofibril components. These misexpressed paralogs intercalate into the nascent cardiac sarcomere to disrupt sarcomere formation and cause impaired cardiac function in utero. These results demonstrate the genomic and physiological requirements for CHD4 in mammalian cardiac development.


Asunto(s)
ADN Helicasas/fisiología , Regulación del Desarrollo de la Expresión Génica , Cardiopatías Congénitas/genética , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2/fisiología , Miocitos Cardíacos/fisiología , Sarcómeros/fisiología , Animales , ADN Helicasas/química , ADN Helicasas/deficiencia , Femenino , Técnicas de Silenciamiento del Gen , Genes Letales , Corazón/diagnóstico por imagen , Corazón/embriología , Cardiopatías Congénitas/diagnóstico por imagen , Cardiopatías Congénitas/embriología , Cardiopatías Congénitas/patología , Masculino , Ratones , Proteínas Musculares/biosíntesis , Proteínas Musculares/genética , Miofibrillas/metabolismo , Miofibrillas/patología , Nucleosomas/metabolismo , Nucleosomas/ultraestructura , Sarcómeros/ultraestructura , Transcripción Genética , Ultrasonografía Prenatal
19.
Int J Mol Sci ; 22(10)2021 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-34068393

RESUMEN

The highly complex life cycle of the human malaria parasite, Plasmodium falciparum, is based on an orchestrated and tightly regulated gene expression program. In general, eukaryotic transcription regulation is determined by a combination of sequence-specific transcription factors binding to regulatory DNA elements and the packaging of DNA into chromatin as an additional layer. The accessibility of regulatory DNA elements is controlled by the nucleosome occupancy and changes of their positions by an active process called nucleosome remodeling. These epigenetic mechanisms are poorly explored in P. falciparum. The parasite genome is characterized by an extraordinarily high AT-content and the distinct architecture of functional elements, and chromatin-related proteins also exhibit high sequence divergence compared to other eukaryotes. Together with the distinct biochemical properties of nucleosomes, these features suggest substantial differences in chromatin-dependent regulation. Here, we highlight the peculiarities of epigenetic mechanisms in P. falciparum, addressing chromatin structure and dynamics with respect to their impact on transcriptional control. We focus on the specialized chromatin remodeling enzymes and discuss their essential function in P. falciparum gene regulation.


Asunto(s)
Ensamble y Desensamble de Cromatina , Epigénesis Genética , Regulación de la Expresión Génica , Malaria Falciparum/parasitología , Plasmodium falciparum/genética , Transcripción Genética , Animales , Humanos , Estadios del Ciclo de Vida
20.
J Biol Chem ; 294(1): 195-209, 2019 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-30409903

RESUMEN

Lineage specification of the three germ layers occurs during early embryogenesis and is critical for normal development. The nucleosome remodeling and deacetylase (NuRD) complex is a repressive chromatin modifier that plays a role in lineage commitment. However, the role of chromodomain helicase DNA-binding protein 4 (CHD4), one of the core subunits of the NuRD complex, in neural lineage commitment is poorly understood. Here, we report that the CHD4/NuRD complex plays a critical role in neural differentiation of mouse embryonic stem cells (ESCs). We found that RNAi-mediated Chd4 knockdown suppresses neural differentiation, as did knockdown of methyl-CpG-binding domain protein Mbd3, another NuRD subunit. Chd4 and Mbd3 knockdowns similarly affected changes in global gene expression during neural differentiation and up-regulated several mesendodermal genes. However, inhibition of mesendodermal genes by knocking out the master regulators of mesendodermal lineages, Brachyury and Eomes, through a CRISPR/Cas9 approach could not restore the impaired neural differentiation caused by the Chd4 knockdown, suggesting that CHD4 controls neural differentiation by not repressing other lineage differentiation processes. Notably, Chd4 knockdown increased the acetylation levels of p53, resulting in increased protein levels of p53. Double knockdown of Chd4 and p53 restored the neural differentiation rate. Furthermore, overexpression of BCL2, a downstream factor of p53, partially rescued the impaired neural differentiation caused by the Chd4 knockdown. Our findings reveal that the CHD4/NuRD complex regulates neural differentiation of ESCs by down-regulating p53.


Asunto(s)
Diferenciación Celular , ADN Helicasas/metabolismo , Regulación hacia Abajo , Neuronas/metabolismo , Nucleosomas/metabolismo , Proteína p53 Supresora de Tumor/biosíntesis , Animales , Línea Celular , ADN Helicasas/genética , Técnicas de Silenciamiento del Gen , Ratones , Células Madre Embrionarias de Ratones , Neuronas/citología , Nucleosomas/genética , Proteínas Proto-Oncogénicas c-bcl-2/biosíntesis , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteína p53 Supresora de Tumor/genética
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda