Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Mol Cell ; 66(4): 568-576.e4, 2017 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-28483418

RESUMO

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


Assuntos
Células-Tronco Embrionárias/enzimologia , Elementos Facilitadores Genéticos , Histona-Lisina N-Metiltransferase/metabolismo , Histonas/metabolismo , Regiões Promotoras Genéticas , RNA/biossíntese , Transcrição Gênica , Animais , Sistemas CRISPR-Cas , Linhagem Celular , Edição de Genes , Regulação da Expressão Gênica no Desenvolvimento , Histona-Lisina N-Metiltransferase/genética , Masculino , Metilação , Camundongos , Mutação , RNA/genética , Fatores de Tempo , Ativação Transcricional , Transfecção
2.
Trends Biochem Sci ; 33(7): 305-13, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18538573

RESUMO

Post-translational modifications of histones serve as docking sites and signals for effector proteins and chromatin-remodeling enzymes, thereby influencing many fundamental cellular processes. Nevertheless, there are huge gaps in the knowledge of which proteins read and write the 'histone code'. Several techniques have been used to decipher complex histone-modification patterns. However, none is entirely satisfactory owing to the inherent limitations of in vitro studies of histones, such as deficits in the knowledge of the proteins involved, and the associated difficulties in the consistent and quantitative generation of histone marks. An alternative technique that could prove to be a useful tool in the study of the histone code is the use of synthetic peptide arrays (SPOT blot analysis) as a screening approach to characterize macromolecules that interact with specific covalent modifications of histone tails.


Assuntos
Cromatina/metabolismo , Epigênese Genética , Histonas/metabolismo , Animais , Humanos , Modelos Biológicos , Análise Serial de Proteínas/métodos
3.
Cell Chem Biol ; 30(10): 1191-1210.e20, 2023 10 19.
Artigo em Inglês | MEDLINE | ID: mdl-37557181

RESUMO

KAT6A, and its paralog KAT6B, are histone lysine acetyltransferases (HAT) that acetylate histone H3K23 and exert an oncogenic role in several tumor types including breast cancer where KAT6A is frequently amplified/overexpressed. However, pharmacologic targeting of KAT6A to achieve therapeutic benefit has been a challenge. Here we describe identification of a highly potent, selective, and orally bioavailable KAT6A/KAT6B inhibitor CTx-648 (PF-9363), derived from a benzisoxazole series, which demonstrates anti-tumor activity in correlation with H3K23Ac inhibition in KAT6A over-expressing breast cancer. Transcriptional and epigenetic profiling studies show reduced RNA Pol II binding and downregulation of genes involved in estrogen signaling, cell cycle, Myc and stem cell pathways associated with CTx-648 anti-tumor activity in ER-positive (ER+) breast cancer. CTx-648 treatment leads to potent tumor growth inhibition in ER+ breast cancer in vivo models, including models refractory to endocrine therapy, highlighting the potential for targeting KAT6A in ER+ breast cancer.


Assuntos
Neoplasias da Mama , Humanos , Feminino , Neoplasias da Mama/genética , Histonas/metabolismo , Histona Acetiltransferases/genética , Histona Acetiltransferases/metabolismo , Transdução de Sinais , Linhagem Celular Tumoral
4.
J Biol Chem ; 286(1): 521-9, 2011 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-21047797

RESUMO

The eight mammalian Cbx proteins are chromodomain-containing proteins involved in regulation of heterochromatin, gene expression, and developmental programs. They are evolutionarily related to the Drosophila HP1 (dHP1) and Pc (dPc) proteins that are key components of chromatin-associated complexes capable of recognizing repressive marks such as trimethylated Lys-9 and Lys-27, respectively, on histone H3. However, the binding specificity and function of the human homologs, Cbx1-8, remain unclear. To this end we employed structural, biophysical, and mutagenic approaches to characterize the molecular determinants of sequence contextual methyllysine binding to human Cbx1-8 proteins. Although all three human HP1 homologs (Cbx1, -3, -5) replicate the structural and binding features of their dHP counterparts, the five Pc homologs (Cbx2, -4, -6, -7, -8) bind with lower affinity to H3K9me3 or H3K27me3 peptides and are unable to distinguish between these two marks. Additionally, peptide permutation arrays revealed a greater sequence tolerance within the Pc family and suggest alternative nonhistone sequences as potential binding targets for this class of chromodomains. Our structures explain the divergence of peptide binding selectivity in the Pc subfamily and highlight previously unrecognized features of the chromodomain that influence binding and specificity.


Assuntos
Proteínas Cromossômicas não Histona/química , Proteínas Cromossômicas não Histona/metabolismo , Sequência de Aminoácidos , Animais , Homólogo 5 da Proteína Cromobox , Sequência Conservada , Proteínas de Drosophila/química , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Histonas/metabolismo , Humanos , Interações Hidrofóbicas e Hidrofílicas , Metilação , Modelos Moleculares , Dados de Sequência Molecular , Complexo Repressor Polycomb 1 , Proteínas do Grupo Polycomb , Estrutura Terciária de Proteína , Proteínas Repressoras/metabolismo , Homologia de Sequência de Aminoácidos , Eletricidade Estática , Especificidade por Substrato
5.
J Biol Chem ; 286(27): 24300-11, 2011 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-21489993

RESUMO

Histone modifications and DNA methylation represent two layers of heritable epigenetic information that regulate eukaryotic chromatin structure and gene activity. UHRF1 is a unique factor that bridges these two layers; it is required for maintenance DNA methylation at hemimethylated CpG sites, which are specifically recognized through its SRA domain and also interacts with histone H3 trimethylated on lysine 9 (H3K9me3) in an unspecified manner. Here we show that UHRF1 contains a tandem Tudor domain (TTD) that recognizes H3 tail peptides with the heterochromatin-associated modification state of trimethylated lysine 9 and unmodified lysine 4 (H3K4me0/K9me3). Solution NMR and crystallographic data reveal the TTD simultaneously recognizes H3K9me3 through a conserved aromatic cage in the first Tudor subdomain and unmodified H3K4 within a groove between the tandem subdomains. The subdomains undergo a conformational adjustment upon peptide binding, distinct from previously reported mechanisms for dual histone mark recognition. Mutant UHRF1 protein deficient for H3K4me0/K9me3 binding shows altered localization to heterochromatic chromocenters and fails to reduce expression of a target gene, p16(INK4A), when overexpressed. Our results demonstrate a novel recognition mechanism for the combinatorial readout of histone modification states associated with gene silencing and add to the growing evidence for coordination of, and cross-talk between, the modification states of H3K4 and H3K9 in regulation of gene expression.


Assuntos
Proteínas Estimuladoras de Ligação a CCAAT/química , Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Heterocromatina/química , Heterocromatina/metabolismo , Histonas/química , Histonas/metabolismo , Animais , Sítios de Ligação , Proteínas Estimuladoras de Ligação a CCAAT/genética , Ilhas de CpG/fisiologia , Cristalografia por Raios X , Inibidor p16 de Quinase Dependente de Ciclina/química , Inibidor p16 de Quinase Dependente de Ciclina/genética , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Metilação de DNA/fisiologia , Regulação da Expressão Gênica/fisiologia , Heterocromatina/genética , Histonas/genética , Humanos , Camundongos , Camundongos Knockout , Ressonância Magnética Nuclear Biomolecular , Processamento de Proteína Pós-Traducional/fisiologia , Estrutura Terciária de Proteína , Ubiquitina-Proteína Ligases
6.
Nucleic Acids Res ; 37(7): 2204-10, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19233876

RESUMO

The MBT repeat has been recently identified as a key domain capable of methyl-lysine histone recognition. Functional work has pointed to a role for MBT domain-containing proteins in transcriptional repression of developmental control genes such as Hox genes. In this study, L3MBTL2, a human homolog of Drosophila Sfmbt critical for Hox gene silencing, is demonstrated to preferentially recognize lower methylation states of several histone-derived peptides through its fourth MBT repeat. High-resolution crystallographic analysis of the four MBT repeats of this protein reveals its unique asymmetric rhomboid architecture, as well as binding mechanism, which preclude the interaction of the first three MBT repeats with methylated peptides. Structural elucidation of an L3MBTL2-H4K20me1 complex and comparison with other MBT-histone peptide complexes also suggests that an absence of distinct surface contours surrounding the methyl-lysine-binding pocket may underlie the lack of sequence specificity observed for members of this protein family.


Assuntos
Histonas/metabolismo , Proteínas Nucleares/química , Proteínas Repressoras/química , Fatores de Transcrição/química , Histonas/química , Humanos , Lisina/metabolismo , Metilação , Modelos Moleculares , Proteínas Nucleares/metabolismo , Peptídeos/química , Peptídeos/metabolismo , Ligação Proteica , Sequências Repetitivas de Aminoácidos , Proteínas Repressoras/metabolismo , Fatores de Transcrição/metabolismo
7.
Sci Adv ; 6(12): eaaz9115, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32219172

RESUMO

Zinc finger protein Zscan4 is selectively expressed in mouse two-cell (2C) embryos undergoing zygotic genome activation (ZGA) and in a rare subpopulation of embryonic stem cells with 2C-like features. Here, we show that Zscan4 specifically recognizes a subset of (CA)n microsatellites, repeat sequences prone to genomic instability. Zscan4-associated microsatellite regions are characterized by low nuclease sensitivity and high histone occupancy. In vitro, Zscan4 binds nucleosomes and protects them from disassembly upon torsional strain. Furthermore, Zscan4 depletion leads to elevated DNA damage in 2C mouse embryos in a transcription-dependent manner. Together, our results identify Zscan4 as a DNA sequence-dependent microsatellite binding factor and suggest a developmentally regulated mechanism, which protects fragile genomic regions from DNA damage at a time of embryogenesis associated with high transcriptional burden and genomic stress.


Assuntos
Dano ao DNA , Células-Tronco Embrionárias/metabolismo , Repetições de Microssatélites , Fatores de Transcrição/metabolismo , Dedos de Zinco , Animais , Sítios de Ligação , Células-Tronco Embrionárias/citologia , Regulação da Expressão Gênica no Desenvolvimento , Genes Reporter , Camundongos , Modelos Biológicos , Nucleossomos/metabolismo , Motivos de Nucleotídeos , Ligação Proteica , Sequências Repetitivas de Ácido Nucleico
8.
Elife ; 4: e10150, 2015 Nov 02.
Artigo em Inglês | MEDLINE | ID: mdl-26523391

RESUMO

Prdm14 is a sequence-specific transcriptional regulator of embryonic stem cell (ESC) pluripotency and primordial germ cell (PGC) formation. It exerts its function, at least in part, through repressing genes associated with epigenetic modification and cell differentiation. Here, we show that this repressive function is mediated through an ETO-family co-repressor Mtgr1, which tightly binds to the pre-SET/SET domains of Prdm14 and co-occupies its genomic targets in mouse ESCs. We generated two monobodies, synthetic binding proteins, targeting the Prdm14 SET domain and demonstrate their utility, respectively, in facilitating crystallization and structure determination of the Prdm14-Mtgr1 complex, or as genetically encoded inhibitor of the Prdm14-Mtgr1 interaction. Structure-guided point mutants and the monobody abrogated the Prdm14-Mtgr1 association and disrupted Prdm14's function in mESC gene expression and PGC formation in vitro. Altogether, our work uncovers the molecular mechanism underlying Prdm14-mediated repression and provides renewable reagents for studying and controlling Prdm14 functions.


Assuntos
Células-Tronco Embrionárias/fisiologia , Gametogênese , Regulação da Expressão Gênica , Células Germinativas/fisiologia , Proteínas Repressoras/metabolismo , Fatores de Transcrição/metabolismo , Animais , Cristalografia por Raios X , Análise Mutacional de DNA , Proteínas de Ligação a DNA , Camundongos , Ligação Proteica , Conformação Proteica , Proteínas de Ligação a RNA , Proteínas Repressoras/química , Fatores de Transcrição/química
9.
J Mol Biol ; 423(5): 702-18, 2012 Nov 09.
Artigo em Inglês | MEDLINE | ID: mdl-22954662

RESUMO

Histone methylation has emerged as an important covalent modification involved in a variety of biological processes, especially regulation of transcription and chromatin dynamics. Lysine methylation is found in three distinct states (monomethylation, dimethylation and trimethylation), which are recognized by specific protein domains. The malignant brain tumor (MBT) domain is one such module found in several chromatin regulatory complexes including Polycomb repressive complex 1. Here, we present a comprehensive characterization of the human MBT family with emphasis on histone binding specificity. SPOT-blot peptide arrays were used to screen for the methyllysine-containing histone peptides that bind to MBT domains found in nine human proteins. Selected interactions were quantified using fluorescence polarization assays. We show that all MBT proteins recognize only monomethyllysine and/or dimethyllysine marks and provide evidence that some MBT domains recognize a defined consensus sequence while others bind in a promiscuous, non-sequence-specific manner. Furthermore, using structure-based mutants, we identify a triad of residues in the methyllysine binding pocket that imparts discrimination between monomethyllysine and dimethyllysine. This study represents a comprehensive analysis of MBT substrate specificity, establishing a foundation for the rational design of selective MBT domain inhibitors that may enable elucidation of their role in human biology and disease.


Assuntos
Neoplasias Encefálicas/metabolismo , Histonas/metabolismo , Sequência de Aminoácidos , Sequência de Bases , Sítios de Ligação , Biologia Computacional , Cristalografia por Raios X , Metilação de DNA , Primers do DNA , Polarização de Fluorescência , Humanos , Lisina/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Reação em Cadeia da Polimerase , Ligação Proteica
10.
Nat Struct Mol Biol ; 19(11): 1155-60, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23022729

RESUMO

A fundamental challenge in mammalian biology has been the elucidation of mechanisms linking DNA methylation and histone post-translational modifications. Human UHRF1 (ubiquitin-like PHD and RING finger domain-containing 1) has multiple domains that bind chromatin, and it is implicated genetically in the maintenance of DNA methylation. However, molecular mechanisms underlying DNA methylation regulation by UHRF1 are poorly defined. Here we show that UHRF1 association with methylated histone H3 Lys9 (H3K9) is required for DNA methylation maintenance. We further show that UHRF1 association with H3K9 methylation is insensitive to adjacent H3 S10 phosphorylation--a known mitotic 'phospho-methyl switch'. Notably, we demonstrate that UHRF1 mitotic chromatin association is necessary for DNA methylation maintenance through regulation of the stability of DNA methyltransferase-1. Collectively, our results define a previously unknown link between H3K9 methylation and the faithful epigenetic inheritance of DNA methylation, establishing a notable mitotic role for UHRF1 in this process.


Assuntos
Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Cromatina/metabolismo , Metilação de DNA/fisiologia , Epigênese Genética/fisiologia , Histonas/metabolismo , Mitose/fisiologia , Sequência de Aminoácidos , Proteínas Estimuladoras de Ligação a CCAAT/genética , Clonagem Molecular , Metilação de DNA/genética , Primers do DNA/genética , Escherichia coli , Polarização de Fluorescência , Células HeLa , Humanos , Espectroscopia de Ressonância Magnética , Análise em Microsséries , Dados de Sequência Molecular , Fosforilação , Ubiquitina-Proteína Ligases
11.
Nat Struct Mol Biol ; 14(12): 1229-30, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18026117

RESUMO

Crystal structures of the L3MBTL1 MBT repeats in complex with histone H4 peptides dimethylated on Lys20 (H4K20me2) show that only the second of the three MBT repeats can bind mono- and dimethylated histone peptides. Its binding pocket has similarities to that of 53BP1 and is able to recognize the degree of histone lysine methylation. An unexpected mode of peptide-mediated dimerization suggests a possible mechanism for chromatin compaction by L3MBTL1.


Assuntos
Histonas/metabolismo , Proteínas de Neoplasias/metabolismo , Sequência de Aminoácidos , Calorimetria , Proteínas Cromossômicas não Histona , Lisina/metabolismo , Metilação , Modelos Moleculares , Dados de Sequência Molecular , Proteínas de Neoplasias/química , Ligação Proteica , Proteínas Repressoras , Homologia de Sequência de Aminoácidos , Proteínas Supressoras de Tumor
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA