Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
EMBO J ; 42(12): e111272, 2023 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-37143403

RESUMO

Patients with chronic obstructive pulmonary disease (COPD) are still waiting for curative treatments. Considering its environmental cause, we hypothesized that COPD will be associated with altered epigenetic signaling in lung cells. We generated genome-wide DNA methylation maps at single CpG resolution of primary human lung fibroblasts (HLFs) across COPD stages. We show that the epigenetic landscape is changed early in COPD, with DNA methylation changes occurring predominantly in regulatory regions. RNA sequencing of matched fibroblasts demonstrated dysregulation of genes involved in proliferation, DNA repair, and extracellular matrix organization. Data integration identified 110 candidate regulators of disease phenotypes that were linked to fibroblast repair processes using phenotypic screens. Our study provides high-resolution multi-omic maps of HLFs across COPD stages. We reveal novel transcriptomic and epigenetic signatures associated with COPD onset and progression and identify new candidate regulators involved in the pathogenesis of chronic lung diseases. The presence of various epigenetic factors among the candidates demonstrates that epigenetic regulation in COPD is an exciting research field that holds promise for novel therapeutic avenues for patients.


Assuntos
Doença Pulmonar Obstrutiva Crônica , Transcriptoma , Humanos , Epigênese Genética , Doença Pulmonar Obstrutiva Crônica/genética , Doença Pulmonar Obstrutiva Crônica/patologia , Pulmão/patologia , Perfilação da Expressão Gênica , Metilação de DNA
2.
Front Endocrinol (Lausanne) ; 14: 1134478, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37008919

RESUMO

Introduction: Beta cell dysfunction by loss of beta cell identity, dedifferentiation, and the presence of polyhormonal cells are main characteristics of diabetes. The straightforward strategy for curing diabetes implies reestablishment of pancreatic beta cell function by beta cell replacement therapy. Aristaless-related homeobox (Arx) gene encodes protein which plays an important role in the development of pancreatic alpha cells and is a main target for changing alpha cell identity. Results: In this study we used CRISPR/dCas9-based epigenetic tools for targeted hypermethylation of Arx gene promoter and its subsequent suppression in mouse pancreatic αTC1-6 cell line. Bisulfite sequencing and methylation profiling revealed that the dCas9-Dnmt3a3L-KRAB single chain fusion constructs (EpiCRISPR) was the most efficient. Epigenetic silencing of Arx expression was accompanied by an increase in transcription of the insulin gene (Ins2) mRNA on 5th and 7th post-transfection day, quantified by both RT-qPCR and RNA-seq. Insulin production and secretion was determined by immunocytochemistry and ELISA assay, respectively. Eventually, we were able to induce switch of approximately 1% of transiently transfected cells which were able to produce 35% more insulin than Mock transfected alpha cells. Conclusion: In conclusion, we successfully triggered a direct, transient switch of pancreatic alpha to insulin-producing cells opening a future research on promising therapeutic avenue for diabetes management.


Assuntos
Diabetes Mellitus , Células Secretoras de Glucagon , Camundongos , Animais , Fatores de Transcrição/metabolismo , Proteínas de Homeodomínio/genética , Insulina/metabolismo , Células Secretoras de Glucagon/metabolismo , Metilação de DNA , Diabetes Mellitus/metabolismo
3.
Adv Exp Med Biol ; 1389: 1-19, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36350504

RESUMO

DNA methylation and DNA methyltransferases (MTases)-the enzymes that introduce the methylation mark into the DNA-have been studied for almost 70 years. In this chapter, we review the key developments in the DNA methylation field that have led to our current understanding of the structures and mechanisms of DNA MTases. We discuss the essential biological roles of DNA methylation, including the discovery of DNA methylation, cloning and sequence analysis of the bacterial and eukaryotic MTases, and the elucidation of their structure, mechanism, regulation, and molecular evolution. We describe genetic studies that contributed greatly to the evolving views on the role of DNA methylation in development and diseases, the invention of methods for the genome-wide analysis of DNA methylation, and the biochemical identification of DNA MTases and the TET enzyme family, which is involved in DNA demethylation. We summarize the roles of MTases in bacterial epigenetics and the application of MTases in synthetic biology to generate artificial signaling systems. We finish by highlighting some open questions for the next years of research in the field.


Assuntos
Metilação de DNA , Metilases de Modificação do DNA , Metilação de DNA/genética , Metilases de Modificação do DNA/genética , Metilases de Modificação do DNA/química , Metiltransferases/química , Evolução Molecular , DNA/genética , DNA/metabolismo
4.
Adv Exp Med Biol ; 1389: 69-110, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36350507

RESUMO

DNA methylation is a hot topic in basic and biomedical research. Despite tremendous progress in understanding the structures and biochemical properties of the mammalian DNA methyltransferases (DNMTs), principles of their targeting and regulation in cells have only begun to be uncovered. In mammals, DNA methylation is introduced by the DNMT1, DNMT3A, and DNMT3B enzymes, which are all large multi-domain proteins containing a catalytic C-terminal domain and a complex N-terminal part with diverse targeting and regulatory functions. The sub-nuclear localization of DNMTs plays an important role in their biological function: DNMT1 is localized to replicating DNA and heterochromatin via interactions with PCNA and UHRF1 and direct binding to the heterochromatic histone modifications H3K9me3 and H4K20me3. DNMT3 enzymes bind to heterochromatin via protein multimerization and are targeted to chromatin by their ADD, PWWP, and UDR domains, binding to unmodified H3K4, H3K36me2/3, and H2AK119ub1, respectively. In recent years, a novel regulatory principle has been discovered in DNMTs, as structural and functional data demonstrated that the catalytic activities of DNMT enzymes are under a tight allosteric control by their different N-terminal domains with autoinhibitory functions. This mechanism provides numerous possibilities for the precise regulation of the methyltransferases via controlling the binding and release of the autoinhibitory domains by protein partners, chromatin interactions, non-coding RNAs, or posttranslational modifications of the DNMTs. In this chapter, we summarize key enzymatic properties of DNMTs, viz. their specificity and processivity, and afterwards focus on the regulation of their activity and targeting via allosteric processes, protein interactions, and posttranslational modifications.


Assuntos
Metilação de DNA , Heterocromatina , Animais , Heterocromatina/genética , DNA Metiltransferase 3A , DNA (Citosina-5-)-Metiltransferases/genética , DNA (Citosina-5-)-Metiltransferase 1 , Metilases de Modificação do DNA/genética , Cromatina/genética , DNA/metabolismo , Mamíferos/genética
5.
Sci Adv ; 8(36): eabm2427, 2022 09 09.
Artigo em Inglês | MEDLINE | ID: mdl-36070377

RESUMO

TET (ten-eleven translocation) enzymes catalyze the oxidation of 5-methylcytosine bases in DNA, thus driving active and passive DNA demethylation. Here, we report that the catalytic domain of mammalian TET enzymes favor CGs embedded within basic helix-loop-helix and basic leucine zipper domain transcription factor-binding sites, with up to 250-fold preference in vitro. Crystal structures and molecular dynamics calculations show that sequence preference is caused by intrasubstrate interactions and CG flanking sequence indirectly affecting enzyme conformation. TET sequence preferences are physiologically relevant as they explain the rates of DNA demethylation in TET-rescue experiments in culture and in vivo within the zygote and germ line. Most and least favorable TET motifs represent DNA sites that are bound by methylation-sensitive immediate-early transcription factors and octamer-binding transcription factor 4 (OCT4), respectively, illuminating TET function in transcriptional responses and pluripotency support.


Assuntos
5-Metilcitosina , Dioxigenases , 5-Metilcitosina/metabolismo , Animais , Domínio Catalítico , Fenômenos Fisiológicos Celulares , DNA , Dioxigenases/genética , Dioxigenases/metabolismo , Mamíferos/genética
6.
JCI Insight ; 6(6)2021 03 22.
Artigo em Inglês | MEDLINE | ID: mdl-33630765

RESUMO

Complexity of lung microenvironment and changes in cellular composition during disease make it exceptionally hard to understand molecular mechanisms driving development of chronic lung diseases. Although recent advances in cell type-resolved approaches hold great promise for studying complex diseases, their implementation relies on local access to fresh tissue, as traditional tissue storage methods do not allow viable cell isolation. To overcome these hurdles, we developed a versatile workflow that allows storage of lung tissue with high viability, permits thorough sample quality check before cell isolation, and befits sequencing-based profiling. We demonstrate that cryopreservation enables isolation of multiple cell types from both healthy and diseased lungs. Basal cells from cryopreserved airways retain their differentiation ability, indicating that cellular identity is not altered by cryopreservation. Importantly, using RNA sequencing and EPIC Array, we show that gene expression and DNA methylation signatures are preserved upon cryopreservation, emphasizing the suitability of our workflow for omics profiling of lung cells. Moreover, we obtained high-quality single-cell RNA-sequencing data of cells from cryopreserved human lungs, demonstrating that cryopreservation empowers single-cell approaches. Overall, thanks to its simplicity, our workflow is well suited for prospective tissue collection by academic collaborators and biobanks, opening worldwide access to viable human tissue.


Assuntos
Criopreservação , Epigênese Genética , Pulmão/metabolismo , Transcrição Gênica , Metilação de DNA , Expressão Gênica , Humanos , Pulmão/citologia , Análise de Sequência de RNA/métodos , Fluxo de Trabalho
7.
Nat Commun ; 11(1): 3355, 2020 07 03.
Artigo em Inglês | MEDLINE | ID: mdl-32620778

RESUMO

Mammalian DNA methylation patterns are established by two de novo DNA methyltransferases, DNMT3A and DNMT3B, which exhibit both redundant and distinctive methylation activities. However, the related molecular basis remains undetermined. Through comprehensive structural, enzymology and cellular characterization of DNMT3A and DNMT3B, we here report a multi-layered substrate-recognition mechanism underpinning their divergent genomic methylation activities. A hydrogen bond in the catalytic loop of DNMT3B causes a lower CpG specificity than DNMT3A, while the interplay of target recognition domain and homodimeric interface fine-tunes the distinct target selection between the two enzymes, with Lysine 777 of DNMT3B acting as a unique sensor of the +1 flanking base. The divergent substrate preference between DNMT3A and DNMT3B provides an explanation for site-specific epigenomic alterations seen in ICF syndrome with DNMT3B mutations. Together, this study reveals distinctive substrate-readout mechanisms of the two DNMT3 enzymes, implicative of their differential roles during development and pathogenesis.


Assuntos
Ilhas de CpG/genética , DNA (Citosina-5-)-Metiltransferases/metabolismo , Metilação de DNA , Animais , Domínio Catalítico , Linhagem Celular , DNA (Citosina-5-)-Metiltransferases/genética , DNA (Citosina-5-)-Metiltransferases/ultraestrutura , DNA Metiltransferase 3A , Células-Tronco Embrionárias , Ensaios Enzimáticos , Epigênese Genética , Face/anormalidades , Humanos , Camundongos , Mutação , Doenças da Imunodeficiência Primária/genética , Relação Estrutura-Atividade , Especificidade por Substrato/genética , Difração de Raios X , DNA Metiltransferase 3B
9.
Mol Syst Biol ; 15(12): e8983, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31885201

RESUMO

Arrayed CRISPR-based screens emerge as a powerful alternative to pooled screens making it possible to investigate a wide range of cellular phenotypes that are typically not amenable to pooled screens. Here, we describe a solid-phase transfection platform that enables CRISPR-based genetic screens in arrayed format with flexible readouts. We demonstrate efficient gene knockout upon delivery of guide RNAs and Cas9/guide RNA ribonucleoprotein complexes into untransformed and cancer cell lines. In addition, we provide evidence that our platform can be easily adapted to high-throughput screens and we use this approach to study oncogene addiction in tumor cells. Finally demonstrating that the human primary cells can also be edited using this method, we pave the way for rapid testing of potential targeted therapies.


Assuntos
Edição de Genes/instrumentação , Neoplasias/genética , RNA Guia de Cinetoplastídeos/farmacologia , Sistemas CRISPR-Cas , Linhagem Celular Tumoral , Predisposição Genética para Doença , Ensaios de Triagem em Larga Escala , Humanos , Fenótipo , Transfecção
10.
Sci Rep ; 8(1): 13242, 2018 09 05.
Artigo em Inglês | MEDLINE | ID: mdl-30185810

RESUMO

The DNA methyltransferase DNMT3A R882H mutation is observed in 25% of all AML patients. DNMT3A is active as tetramer and the R882H mutation is located in one of the subunit/subunit interfaces. Previous work has reported that formation of mixed wildtype/R882H complexes leads to a strong loss of catalytic activity observed in in vitro DNA methylation assays (Russler-Germain et al., 2014, Cancer Cell 25:442-454). To investigate this effect further, we have prepared mixed wildtype/R882H DNMT3A complexes by incubation of individually purified subunits of the DNMT3A catalytic domain and full-length DNMT3A2. In addition, we have used a double affinity tag approach and specifically purified mixed catalytic domain complexes formed after co-expression of R882H and wildtype subunits in E. coli cells. Afterwards, we determined the catalytic activity of the mixed complexes and compared it to that of purified complexes only consisting of one subunit type. In both settings, the expected catalytic activities of mixed R882H/wildtype complexes were observed demonstrating an absence of a dominant negative effect of the R882H mutation in purified DNMT3A enzymes. This result suggests that heterocomplex formation of DNMT3A and R882H is unlikely to cause dominant negative effects in human cells as well. The limitations of this conclusion and its implications are discussed.


Assuntos
DNA (Citosina-5-)-Metiltransferases/química , DNA (Citosina-5-)-Metiltransferases/genética , Mutação , Domínio Catalítico , DNA (Citosina-5-)-Metiltransferases/metabolismo , Metilação de DNA , DNA Metiltransferase 3A , Humanos , Modelos Moleculares , Multimerização Proteica
11.
Nucleic Acids Res ; 46(17): 9044-9056, 2018 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-30102379

RESUMO

Despite their central importance in mammalian development, the mechanisms that regulate the DNA methylation machinery and thereby the generation of genomic methylation patterns are still poorly understood. Here, we identify the 5mC-binding protein MeCP2 as a direct and strong interactor of DNA methyltransferase 3 (DNMT3) proteins. We mapped the interaction interface to the transcriptional repression domain of MeCP2 and the ADD domain of DNMT3A and find that binding of MeCP2 strongly inhibits the activity of DNMT3A in vitro. This effect was reinforced by cellular studies where a global reduction of DNA methylation levels was observed after overexpression of MeCP2 in human cells. By engineering conformationally locked DNMT3A variants as novel tools to study the allosteric regulation of this enzyme, we show that MeCP2 stabilizes the closed, autoinhibitory conformation of DNMT3A. Interestingly, the interaction with MeCP2 and its resulting inhibition were relieved by the binding of K4 unmodified histone H3 N-terminal tail to the DNMT3A-ADD domain. Taken together, our data indicate that the localization and activity of DNMT3A are under the combined control of MeCP2 and H3 tail modifications where, depending on the modification status of the H3 tail at the binding sites, MeCP2 can act as either a repressor or activator of DNA methylation.


Assuntos
Cromatina/metabolismo , DNA (Citosina-5-)-Metiltransferases/genética , DNA/química , Epigênese Genética , Histonas/genética , Proteína 2 de Ligação a Metil-CpG/genética , Regulação Alostérica , Animais , Sítios de Ligação , Química Encefálica , Cromatina/química , Clonagem Molecular , DNA/metabolismo , DNA (Citosina-5-)-Metiltransferases/química , DNA (Citosina-5-)-Metiltransferases/metabolismo , Metilação de DNA , DNA Metiltransferase 3A , Escherichia coli/genética , Escherichia coli/metabolismo , Expressão Gênica , Vetores Genéticos/química , Vetores Genéticos/metabolismo , Células HEK293 , Histonas/química , Histonas/metabolismo , Humanos , Proteína 2 de Ligação a Metil-CpG/química , Proteína 2 de Ligação a Metil-CpG/metabolismo , Camundongos , Mutagênese Sítio-Dirigida/métodos , Ligação Proteica , Engenharia de Proteínas/métodos , Domínios e Motivos de Interação entre Proteínas , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
12.
Nucleic Acids Res ; 46(6): 3130-3139, 2018 04 06.
Artigo em Inglês | MEDLINE | ID: mdl-29518238

RESUMO

The DNMT3A R882H mutation is frequently observed in acute myeloid leukemia (AML). It is located in the subunit and DNA binding interface of DNMT3A and has been reported to cause a reduction in activity and dominant negative effects. We investigated the mechanistic consequences of the R882H mutation on DNMT3A showing a roughly 40% reduction in overall DNA methylation activity. Biochemical assays demonstrated that R882H does not change DNA binding affinity, protein stability or subnuclear distribution of DNMT3A. Strikingly, DNA methylation experiments revealed pronounced changes in the flanking sequence preference of the DNMT3A-R882H mutant. Based on these results, different DNA substrates with selected flanking sequences were designed to be favored or disfavored by R882H. Kinetic analyses showed that the R882H favored substrate was methylated by R882H with 45% increased rate when compared with wildtype DNMT3A, while methylation of the disfavored substrate was reduced 7-fold. Our data expand the model of the potential carcinogenic effect of the R882H mutation by showing CpG site specific activity changes. This result suggests that R882 is involved in the indirect readout of flanking sequence preferences of DNMT3A and it may explain the particular enrichment of the R882H mutation in cancer patients by revealing mutation specific effects.


Assuntos
DNA (Citosina-5-)-Metiltransferases/genética , Metilação de DNA , DNA/metabolismo , Mutação de Sentido Incorreto , Doença Aguda , Sítios de Ligação/genética , Ilhas de CpG/genética , DNA/química , DNA/genética , DNA (Citosina-5-)-Metiltransferases/química , DNA (Citosina-5-)-Metiltransferases/metabolismo , DNA Metiltransferase 3A , Humanos , Leucemia Mieloide/enzimologia , Leucemia Mieloide/genética , Modelos Moleculares , Conformação de Ácido Nucleico , Ligação Proteica , Domínios Proteicos , Especificidade por Substrato
13.
Nat Commun ; 8(1): 2057, 2017 12 12.
Artigo em Inglês | MEDLINE | ID: mdl-29234025

RESUMO

SETDB1 is an essential H3K9 methyltransferase involved in silencing of retroviruses and gene regulation. We show here that its triple Tudor domain (3TD) specifically binds to doubly modified histone H3 containing K14 acetylation and K9 methylation. Crystal structures of 3TD in complex with H3K14ac/K9me peptides reveal that peptide binding and K14ac recognition occurs at the interface between Tudor domains (TD) TD2 and TD3. Structural and biochemical data demonstrate a pocket switch mechanism in histone code reading, because K9me1 or K9me2 is preferentially recognized by the aromatic cage of TD3, while K9me3 selectively binds to TD2. Mutations in the K14ac/K9me binding sites change the sub-nuclear localization of 3TD. ChIP-seq analyses show that SETDB1 is enriched at H3K9me3 regions and K9me3/K14ac is enriched at SETDB1 binding sites overlapping with LINE elements, suggesting that recruitment of the SETDB1 complex to K14ac/K9me regions has a role in silencing of active genomic regions.


Assuntos
Histonas/metabolismo , Elementos Nucleotídeos Longos e Dispersos/fisiologia , Proteínas Metiltransferases/fisiologia , Processamento de Proteína Pós-Traducional/fisiologia , Domínio Tudor/fisiologia , Acetilação , Animais , Sítios de Ligação/fisiologia , Cristalografia por Raios X , Células HEK293 , Histona-Lisina N-Metiltransferase , Histonas/genética , Humanos , Metilação , Camundongos , Células-Tronco Embrionárias Murinas , Ligação Proteica/fisiologia , Proteínas Metiltransferases/química , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo
14.
Nucleic Acids Res ; 45(4): 1703-1713, 2017 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-27899645

RESUMO

DNA methylation plays a critical role in the regulation and maintenance of cell-type specific transcriptional programs. Targeted epigenome editing is an emerging technology to specifically regulate cellular gene expression in order to modulate cell phenotypes or dissect the epigenetic mechanisms involved in their control. In this work, we employed a DNA methyltransferase Dnmt3a-Dnmt3L construct fused to the nuclease-inactivated dCas9 programmable targeting domain to introduce DNA methylation into the human genome specifically at the EpCAM, CXCR4 and TFRC gene promoters. We show that targeting of these loci with single gRNAs leads to efficient and widespread methylation of the promoters. Multiplexing of several guide RNAs does not increase the efficiency of methylation. Peaks of targeted methylation were observed around 25 bp upstream and 40 bp downstream of the PAM site, while 20-30 bp of the binding site itself are protected against methylation. Potent methylation is dependent on the multimerization of Dnmt3a/Dnmt3L complexes on the DNA. Furthermore, the introduced methylation causes transcriptional repression of the targeted genes. These new programmable epigenetic editors allow unprecedented control of the DNA methylation status in cells and will lead to further advances in the understanding of epigenetic signaling.


Assuntos
DNA (Citosina-5-)-Metiltransferases/metabolismo , Metilação de DNA , DNA/genética , DNA/metabolismo , Endonucleases/metabolismo , Animais , Linhagem Celular Tumoral , Análise por Conglomerados , Ilhas de CpG , DNA (Citosina-5-)-Metiltransferases/química , DNA (Citosina-5-)-Metiltransferases/genética , Endonucleases/genética , Epigênese Genética , Epigenômica/métodos , Regulação da Expressão Gênica , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Camundongos , Modelos Biológicos , Regiões Promotoras Genéticas , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo
15.
Adv Exp Med Biol ; 945: 1-17, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27826832

RESUMO

DNA methylation and DNA methyltransferases (MTases) - the enzymes that introduce the methylation mark into the DNA - have been studied for almost 70 years. In this chapter, we review key developments in the field that led to our current understanding of the structures and mechanisms of DNA MTases and the essential biological role of DNA methylation, including the discovery of DNA methylation and DNA MTases, the cloning and sequence analysis of bacterial and eukaryotic MTases, and the elucidation of their structure, mechanism, and regulation. We describe genetic studies that contributed greatly to the evolving views on the role of DNA methylation in human development and diseases, the invention of methods for the genome-wide analysis of DNA methylation, and the biochemical identification of DNA MTases and the family of TET enzymes, which are involved in DNA demethylation. We finish by highlighting critical questions for the next years of research in the field.


Assuntos
DNA (Citosina-5-)-Metiltransferases/genética , Metilação de DNA/genética , Metilases de Modificação do DNA/genética , Evolução Molecular , Animais , DNA (Citosina-5-)-Metiltransferases/química , Metilases de Modificação do DNA/química , Humanos , Mamíferos/genética , Oxigenases de Função Mista/genética , Proteínas Proto-Oncogênicas/genética
16.
Adv Exp Med Biol ; 945: 87-122, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27826836

RESUMO

DNA methylation is currently one of the hottest topics in basic and biomedical research. Despite tremendous progress in understanding the structures and biochemical properties of the mammalian DNA nucleotide methyltransferases (DNMTs), principles of their regulation in cells have only begun to be uncovered. In mammals, DNA methylation is introduced by the DNMT1, DNMT3A, and DNMT3B enzymes, which are all large multi-domain proteins. These enzymes contain a catalytic C-terminal domain with a characteristic cytosine-C5 methyltransferase fold and an N-terminal part with different domains that interacts with other proteins and chromatin and is involved in targeting and regulation of the DNMTs. The subnuclear localization of the DNMT enzymes plays an important role in their biological function: DNMT1 is localized to replicating DNA via interaction with PCNA and UHRF1. DNMT3 enzymes bind to heterochromatin via protein multimerization and are targeted to chromatin by their ADD and PWWP domains. Recently, a novel regulatory mechanism has been discovered in DNMTs, as latest structural and functional data demonstrated that the catalytic activities of all three enzymes are under tight allosteric control of their N-terminal domains having autoinhibitory functions. This mechanism provides numerous possibilities for the precise regulation of the methyltransferases via controlling the binding and release of autoinhibitory domains by protein factors, noncoding RNAs, or by posttranslational modifications of the DNMTs. In this chapter, we summarize key enzymatic properties of DNMTs, including their specificity and processivity, and afterward we focus on the regulation of their activity and targeting via allosteric processes, protein interactors, and posttranslational modifications.


Assuntos
DNA (Citosina-5-)-Metiltransferases/química , Metilação de DNA/genética , Animais , Domínio Catalítico/genética , Cromatina/química , Cromatina/genética , DNA (Citosina-5-)-Metiltransferase 1 , DNA (Citosina-5-)-Metiltransferases/genética , DNA Metiltransferase 3A , Heterocromatina/química , Heterocromatina/genética , Humanos , Processamento de Proteína Pós-Traducional/genética , DNA Metiltransferase 3B
17.
Nucleic Acids Res ; 44(18): 8556-8575, 2016 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-27521372

RESUMO

In mammals, DNA methylation is introduced by the DNMT1, DNMT3A and DNMT3B methyltransferases, which are all large multi-domain proteins containing a catalytic C-terminal domain and an N-terminal part with regulatory functions. Recently, two novel regulatory principles of DNMTs were uncovered. It was shown that their catalytic activity is under allosteric control of N-terminal domains with autoinhibitory function, the RFT and CXXC domains in DNMT1 and the ADD domain in DNMT3. Moreover, targeting and activity of DNMTs were found to be regulated in a concerted manner by interactors and posttranslational modifications (PTMs). In this review, we describe the structures and domain composition of the DNMT1 and DNMT3 enzymes, their DNA binding, catalytic mechanism, multimerization and the processes controlling their stability in cells with a focus on their regulation and chromatin targeting by PTMs, interactors and chromatin modifications. We propose that the allosteric regulation of DNMTs by autoinhibitory domains acts as a general switch for the modulation of the function of DNMTs, providing numerous possibilities for interacting proteins, nucleic acids or PTMs to regulate DNMT activity and targeting. The combined regulation of DNMT targeting and catalytic activity contributes to the precise spatiotemporal control of DNMT function and genome methylation in cells.


Assuntos
DNA (Citosina-5-)-Metiltransferases/metabolismo , Mamíferos/metabolismo , Regulação Alostérica , Animais , Cromatina , DNA (Citosina-5-)-Metiltransferases/química , Humanos , Modelos Biológicos , Ligação Proteica
18.
BMC Biochem ; 17: 7, 2016 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-27001594

RESUMO

BACKGROUND: Dnmt3a is a DNA methyltransferase that establishes de novo DNA methylation in mammals. The structure of the Dnmt3a C-terminal domain is similar to the bacterial M. HhaI enzyme, a well-studied prokaryotic DNA methyltransferase. No X-ray structure is available for the complex of Dnmt3a with DNA and the mechanistic details of DNA recognition and catalysis by mammalian Dnmts are not completely understood. RESULTS: Mutant variants of the catalytic domain of the murine Dnmt3a carrying substitutions of highly conserved N167, R200, and R202 have been generated by site directed mutagenesis and purified. Their methylation activity, DNA binding affinity, ability to flip the target cytosine out of the DNA double helix and covalent complex formation with DNA have been examined. Substitutions of N167 lead to reduced catalytic activity and reduced base flipping. Catalytic activity, base flipping, and covalent conjugate formation were almost completely abolished for the mutant enzymes with substitutions of R200 or R202. CONCLUSIONS: We conclude that R202 plays a similar role in catalysis in Dnmt3a-CD as R232 in M.SssI and R165 in M.HhaI, which could be positioning of the cytosine for nucleophilic attack by a conserved Cys. R200 of Dnmt3a-CD is important in both catalysis and cytosine flipping. Both conserved R200 and R202 are involved in creating and stabilizing of the transient covalent intermediate of the methylation reaction. N167 might contribute to the positioning of the residues from the motif VI, but does not play a direct role in catalysis.


Assuntos
DNA (Citosina-5-)-Metiltransferases/metabolismo , Metilação de DNA , Sequência de Aminoácidos , Animais , Domínio Catalítico , DNA/química , DNA/metabolismo , DNA (Citosina-5-)-Metiltransferases/genética , DNA Metiltransferase 3A , Fluoresceína/química , Cinética , Camundongos , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/química , Proteínas Recombinantes/isolamento & purificação , Alinhamento de Sequência
19.
J Mol Biol ; 427(10): 1903-15, 2015 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-25823553

RESUMO

The HELLS (helicase, lymphoid specific, also known as lymphoid-specific helicase) protein is related to the SNF2 (sucrose non-fermentable 2) family of chromatin remodeling ATPases. It is required for efficient DNA methylation in mammals, particularly at heterochromatin-located repetitive sequences. In this study, we investigated the interaction of HELLS with chromatin and used an ATPase-deficient HELLS variant to address the role of ATP hydrolysis in this process. Chromatin fractionation experiments demonstrated that, in the absence of the ATPase activity, HELLS is retained at the nuclear matrix compartment, defined in part by lamin B1. Microscopy studies revealed a stronger association of the ATPase-deficient mutant with heterochromatin. These results were further supported by fluorescence recovery after photobleaching measurements, which showed that, at heterochromatic sites, wild-type HELLS is very dynamic, with a recovery half-time of 0.8s and a mobile protein fraction of 61%. In contrast, the ATPase-deficient mutant displayed 4.5-s recovery half-time and a reduction in the mobile fraction to 30%. We also present evidence suggesting that, in addition to the ATPase activity, a functional H3K9me3 signaling pathway contributes to an efficient release of HELLS from pericentromeric chromatin. Overall, our results show that a functional ATPase activity is not required for the recruitment of HELLS to heterochromatin, but it is important for the release of the enzyme from these sites.


Assuntos
Adenosina Trifosfatases/deficiência , DNA Helicases/metabolismo , Embrião de Mamíferos/metabolismo , Fibroblastos/metabolismo , Heterocromatina/metabolismo , Metiltransferases/fisiologia , Proteínas Mutantes/metabolismo , Proteínas Repressoras/fisiologia , Animais , Western Blotting , Células Cultivadas , DNA Helicases/genética , Metilação de DNA , Embrião de Mamíferos/citologia , Fibroblastos/citologia , Recuperação de Fluorescência Após Fotodegradação , Histonas/metabolismo , Camundongos , Camundongos Knockout , Proteínas Mutantes/genética , Mutação/genética , Transcrição Gênica
20.
Genome Res ; 24(11): 1842-53, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25301795

RESUMO

Post-translational modifications (PTMs) of histones constitute a major chromatin indexing mechanism, and their proper characterization is of highest biological importance. So far, PTM-specific antibodies have been the standard reagent for studying histone PTMs despite caveats such as lot-to-lot variability of specificity and binding affinity. Herein, we successfully employed naturally occurring and engineered histone modification interacting domains for detection and identification of histone PTMs and ChIP-like enrichment of different types of chromatin. Our results demonstrate that histone interacting domains are robust and highly specific reagents that can replace or complement histone modification antibodies. These domains can be produced recombinantly in Escherichia coli at low cost and constant quality. Protein design of reading domains allows for generation of novel specificities, addition of affinity tags, and preparation of PTM binding pocket variants as matching negative controls, which is not possible with antibodies.


Assuntos
Anticorpos/metabolismo , Histonas/metabolismo , Mapeamento de Interação de Proteínas/métodos , Processamento de Proteína Pós-Traducional , Sequência de Aminoácidos , Anticorpos/imunologia , Sítios de Ligação/genética , Western Blotting , Imunoprecipitação da Cromatina , DNA (Citosina-5-)-Metiltransferases/genética , DNA (Citosina-5-)-Metiltransferases/metabolismo , DNA Metiltransferase 3A , Células HEK293 , Histonas/imunologia , Humanos , Lisina/metabolismo , Metilação , Peptídeos/metabolismo , Complexo Repressor Polycomb 1/genética , Complexo Repressor Polycomb 1/metabolismo , Análise Serial de Proteínas/métodos , Ligação Proteica , Reprodutibilidade dos Testes
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...