histones

组蛋白
  • 文章类型: Journal Article
    表观遗传改变,如染色质结构和DNA甲基化,已经在许多肿瘤类型中进行了广泛的研究。但是口腔癌,尤其是口腔腺癌,受到的关注要少得多。这里,我们将激光捕获显微切割和muti-omics小型批量测序相结合,系统地表征口腔癌的表观遗传景观,包括染色质结构,DNA甲基化,H3K27me3修改,和基因表达。在致癌作用中,肿瘤细胞表现出重组的染色质空间结构,包括受损的区室结构和改变的基因-基因相互作用网络。值得注意的是,在表型非恶性癌旁细胞中观察到一些结构改变,但在正常细胞中未观察到。我们开发了转换模型来识别个体基因组基因座的癌症倾向,从而确定每个样品的致癌状态。对癌症表观遗传景观的见解提供了证据,表明染色质重组是口腔癌进展的重要标志。这也与基因组改变和DNA甲基化重编程有关。特别是,癌细胞中拷贝数频繁变化的区域与癌症和正常样本中强烈的空间绝缘相关。口腔鳞状细胞癌中异常甲基化重编程与染色质结构和H3K27me3信号密切相关,进一步受到内在序列属性的影响。我们的发现表明,在两种不同类型的口腔癌中,结构变化既重要又保守。与转录组改变和癌症发展密切相关。值得注意的是,尽管与鳞状细胞癌相比基因组拷贝数改变的发生率相当低,甲基化改变的程度较小,但在口腔腺癌中结构改变仍然明显明显.我们期望对不同类型和亚型的原发性口腔肿瘤的表观遗传重编程的综合分析可以为口腔癌的新型检测和治疗的设计提供额外的指导。
    Epigenetic alterations, such as those in chromatin structure and DNA methylation, have been extensively studied in a number of tumor types. But oral cancer, particularly oral adenocarcinoma, has received far less attention. Here, we combined laser-capture microdissection and muti-omics mini-bulk sequencing to systematically characterize the epigenetic landscape of oral cancer, including chromatin architecture, DNA methylation, H3K27me3 modification, and gene expression. In carcinogenesis, tumor cells exhibit reorganized chromatin spatial structures, including compromised compartment structures and altered gene-gene interaction networks. Notably, some structural alterations are observed in phenotypically non-malignant paracancerous but not in normal cells. We developed transformer models to identify the cancer propensity of individual genome loci, thereby determining the carcinogenic status of each sample. Insights into cancer epigenetic landscapes provide evidence that chromatin reorganization is an important hallmark of oral cancer progression, which is also linked with genomic alterations and DNA methylation reprogramming. In particular, regions of frequent copy number alternations in cancer cells are associated with strong spatial insulation in both cancer and normal samples. Aberrant methylation reprogramming in oral squamous cell carcinomas is closely related to chromatin structure and H3K27me3 signals, which are further influenced by intrinsic sequence properties. Our findings indicate that structural changes are both significant and conserved in two distinct types of oral cancer, closely linked to transcriptomic alterations and cancer development. Notably, the structural changes remain markedly evident in oral adenocarcinoma despite the considerably lower incidence of genomic copy number alterations and lesser extent of methylation alterations compared to squamous cell carcinoma. We expect that the comprehensive analysis of epigenetic reprogramming of different types and subtypes of primary oral tumors can provide additional guidance to the design of novel detection and therapy for oral cancer.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    G9a是一种组蛋白甲基转移酶,催化组蛋白3赖氨酸9(H3K9)的甲基化,参与基因表达的调控。我们先前报道了G9a在体内和体外发育的肌腱中表达,并且G9a缺陷的肌腱细胞在体外表现出受损的增殖和分化。在这项研究中,我们通过使用G9a条件敲除(G9acKO)小鼠研究了G9a在体内肌腱发育中的功能。我们将Sox9Cre/+小鼠与G9afl/fl小鼠交叉以产生G9afl/fl;Sox9Cre/+小鼠。G9acKO小鼠在3周龄时显示出发育不良的肌腱形成。在胚胎第16.5天(E16.5)的溴脱氧尿苷标记显示G9acKO小鼠的肌腱细胞中细胞增殖减少。免疫组织化学分析显示G9a及其底物的表达水平降低,H3K9me2,在G9acKO小鼠的椎体肌腱中。G9acKO小鼠的椎骨和四肢的肌腱组织显示肌腱标记物的表达降低,腱调节素(Tnmd),和col1a1基因,提示肌腱细胞分化被抑制。G9a的过表达导致Tnmd和col1a1在体外肌腱细胞中的表达增强。这些结果表明,G9a在肌腱发育过程中调节肌腱祖细胞的增殖和分化。因此,我们的研究结果表明,G9a在肌腱发育中起着至关重要的作用。
    G9a is a histone methyltransferase that catalyzes the methylation of histone 3 lysine 9 (H3K9), which is involved in the regulation of gene expression. We had previously reported that G9a is expressed in developing tendons in vivo and in vitro and that G9a-deficient tenocytes show impaired proliferation and differentiation in vitro. In this study, we investigated the functions of G9a in tendon development in vivo by using G9a conditional knockout (G9a cKO) mice. We crossed Sox9Cre/+ mice with G9afl/fl mice to generate G9afl/fl; Sox9Cre/+ mice. The G9a cKO mice showed hypoplastic tendon formation at 3 weeks of age. Bromodeoxyuridine labeling on embryonic day 16.5 (E16.5) revealed decreased cell proliferation in the tenocytes of G9a cKO mice. Immunohistochemical analysis revealed decreased expression levels of G9a and its substrate, H3K9me2, in the vertebral tendons of G9a cKO mice. The tendon tissue of the vertebrae and limbs of G9a cKO mice showed reduced expression of a tendon marker, tenomodulin (Tnmd), and col1a1 genes, suggesting that tenocyte differentiation was suppressed. Overexpression of G9a resulted in enhancement of Tnmd and col1a1 expression in tenocytes in vitro. These results suggest that G9a regulates the proliferation and differentiation of tendon progenitor cells during tendon development. Thus, our results suggest that G9a plays an essential role in tendon development.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    赖氨酸特异性组蛋白去甲基酶1(LSD1),将单甲基化或二甲基化组蛋白H3在赖氨酸4(H3K4me1/2)上,对早期胚胎发生和发育至关重要。在这里,我们表明LSD1对于小鼠胚胎干细胞(ESC)的自我更新是不必要的,但对于小鼠ESC的生长和分化是必需的。重新引入催化受损的LSD1(LSD1MUT)可以恢复小鼠ESC的增殖能力,然而,LSD1的酶活性对于确保适当的分化至关重要。的确,Lsd1敲除(KO)小鼠ESC中H3K4me1的增加不会导致与干性相关的全局基因表达程序的重大变化。然而,LSD1而非LSD1MUT的消融导致DNMT1和UHRF1蛋白减少,并伴随整体低甲基化.我们表明,LSD1和LSD1MUT都通过与HDAC1和泛素特异性肽酶7(USP7)的相互作用来控制UHRF1和DNMT1的蛋白质稳定性,因此,促进DNMT1和UHRF1的去乙酰化和去泛素化。我们的研究阐明了LSD1控制小鼠ESCDNA甲基化的机制,独立于其赖氨酸脱甲基酶活性。
    Lysine-specific histone demethylase 1 (LSD1), which demethylates mono- or di- methylated histone H3 on lysine 4 (H3K4me1/2), is essential for early embryogenesis and development. Here we show that LSD1 is dispensable for mouse embryonic stem cell (ESC) self-renewal but is required for mouse ESC growth and differentiation. Reintroduction of a catalytically-impaired LSD1 (LSD1MUT) recovers the proliferation capability of mouse ESCs, yet the enzymatic activity of LSD1 is essential to ensure proper differentiation. Indeed, increased H3K4me1 in Lsd1 knockout (KO) mouse ESCs does not lead to major changes in global gene expression programs related to stemness. However, ablation of LSD1 but not LSD1MUT results in decreased DNMT1 and UHRF1 proteins coupled to global hypomethylation. We show that both LSD1 and LSD1MUT control protein stability of UHRF1 and DNMT1 through interaction with HDAC1 and the ubiquitin-specific peptidase 7 (USP7), consequently, facilitating the deacetylation and deubiquitination of DNMT1 and UHRF1. Our studies elucidate a mechanism by which LSD1 controls DNA methylation in mouse ESCs, independently of its lysine demethylase activity.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    组蛋白H3突变神经胶质瘤是致命的脑肿瘤,其特征是表观基因组失调和分化停滞。与肿瘤细胞上可用的大量数据集相反,关于他们的肿瘤微环境(TME)的信息有限,尤其是免疫渗透。这里,我们表征了H3.3K27M和G34R/V突变神经胶质瘤的免疫TME,和多个H3.3K27M小鼠模型,使用转录组学,蛋白质组学和空间单细胞方法。免疫谱系的分辨率表明H3突变神经胶质瘤具有不同的髓系群体的高浸润,免疫检查点标志物的高水平表达,和稀缺的淋巴样细胞,结果在所有测试的H3.3K27M小鼠模型中一致再现。我们展示了这些髓样群体与H3突变细胞进行通信,介导免疫抑制和维持肿瘤的形成和维持。骨髓细胞和免疫检查点途径的双重抑制在临床前同基因小鼠模型中显示出显著的治疗益处。我们的发现提供了有价值的肿瘤组蛋白突变神经胶质瘤的TME表征,并深入了解调节骨髓浸润的方法,以造福患者。
    Histone H3-mutant gliomas are deadly brain tumors characterized by a dysregulated epigenome and stalled differentiation. In contrast to the extensive datasets available on tumor cells, limited information exists on their tumor microenvironment (TME), particularly the immune infiltrate. Here, we characterize the immune TME of H3.3K27M and G34R/V-mutant gliomas, and multiple H3.3K27M mouse models, using transcriptomic, proteomic and spatial single-cell approaches. Resolution of immune lineages indicates high infiltration of H3-mutant gliomas with diverse myeloid populations, high-level expression of immune checkpoint markers, and scarce lymphoid cells, findings uniformly reproduced in all H3.3K27M mouse models tested. We show these myeloid populations communicate with H3-mutant cells, mediating immunosuppression and sustaining tumor formation and maintenance. Dual inhibition of myeloid cells and immune checkpoint pathways show significant therapeutic benefits in pre-clinical syngeneic mouse models. Our findings provide a valuable characterization of the TME of oncohistone-mutant gliomas, and insight into the means for modulating the myeloid infiltrate for the benefit of patients.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    背景:速效抗抑郁药(RAAD),包括解离性麻醉药,迷幻药,和移态药,通过据称调节神经可塑性来引发精神疾病的快速和持续的治疗改善,神经传递,和豁免权。这些结果可能是由,或导致,表观遗传过程的急性和/或持续夹带,重塑染色质结构并改变DNA可及性以调节基因表达。
    方法:从这个角度来看,我们概述了已知的机制,知识差距,以及围绕RAAD表观遗传效应的未来方向,专注于压力反应性DNA和大脑区域的调节,以及与常规抗抑郁药的比较。
    方法:初步相关证据表明,RAAD的给药伴随着表观遗传效应,这与常规抗抑郁药引起的效应相似。这些包括DNA甲基化的变化,组蛋白的翻译后修饰,和非编码RNA在涉及神经营养的应激反应染色质区域的差异调节,神经传递,和免疫调节,在应激反应的大脑区域。这些表观遗传变化是否对RAAD的治疗效果有因果关系,是其后果,或者是无关的,仍然未知。此外,潜在的细胞类型特异性和相关机制尚未完全阐明。候选机制包括神经元活性和5-羟色胺和原肌苷受体激酶B(TRKB)信号介导的表观遗传变化,与DNA的直接相互作用,组蛋白,或染色质重塑复合物。
    结论:相关证据表明,RAAD诱导的表观遗传变化伴随治疗和副作用,虽然因果关系,机制,和细胞类型特异性仍然很大程度上未知。解决这些研究空白可能会导致新的基于神经表观遗传学的精确疗法的发展。
    BACKGROUND: Rapid-acting antidepressants (RAADs), including dissociative anesthetics, psychedelics, and empathogens, elicit rapid and sustained therapeutic improvements in psychiatric disorders by purportedly modulating neuroplasticity, neurotransmission, and immunity. These outcomes may be mediated by, or result in, an acute and/or sustained entrainment of epigenetic processes, which remodel chromatin structure and alter DNA accessibility to regulate gene expression.
    METHODS: In this perspective, we present an overview of the known mechanisms, knowledge gaps, and future directions surrounding the epigenetic effects of RAADs, with a focus on the regulation of stress-responsive DNA and brain regions, and on the comparison with conventional antidepressants.
    METHODS: Preliminary correlative evidence indicates that administration of RAADs is accompanied by epigenetic effects which are similar to those elicited by conventional antidepressants. These include changes in DNA methylation, post-translational modifications of histones, and differential regulation of non-coding RNAs in stress-responsive chromatin areas involved in neurotrophism, neurotransmission, and immunomodulation, in stress-responsive brain regions. Whether these epigenetic changes causally contribute to the therapeutic effects of RAADs, are a consequence thereof, or are unrelated, remains unknown. Moreover, the potential cell type-specificity and mechanisms involved are yet to be fully elucidated. Candidate mechanisms include neuronal activity- and serotonin and Tropomyosine Receptor Kinase B (TRKB) signaling-mediated epigenetic changes, and direct interaction with DNA, histones, or chromatin remodeling complexes.
    CONCLUSIONS: Correlative evidence suggests that epigenetic changes induced by RAADs accompany therapeutic and side effects, although causation, mechanisms, and cell type-specificity remain largely unknown. Addressing these research gaps may lead to the development of novel neuroepigenetics-based precision therapeutics.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    脓毒症代表宿主对感染的失调反应导致的器官功能障碍,并可引起急性肾损伤(AKI),显着增加脓毒症患者的发病率和死亡率。这项研究致力于为脓毒症诱导的AKI(SI-AKI)患者确定一种新的治疗策略。对大鼠肾小管上皮NRK-52E细胞进行脂多糖(LPS)暴露以诱导体外SI-AKI。用Westernblot和qRT-PCR检测NRK-52E细胞中E1A结合蛋白p300(EP300)和亚甲基四氢叶酸脱氢酶2(MTHFD2)的表达,通过用H3K27乙酰化抗体(H3K27ac)进行染色质免疫沉淀来探索它们的相互作用。他们对SI-AKI相关的肾小管上皮细胞的线粒体功能障碍的影响进行了研究,使用转染,MTT测定,TUNEL染色,2\',7'-二氯二氢荧光素二乙酸探针测定,Mitosox检测,和JC-1染色。在NRK-52E细胞中,通过LPS暴露上调MTHFD2和EP300。LPS增加了H3组蛋白在MTHFD2启动子区的乙酰化,和EP300抑制LPS的作用。EP300消融抑制MTHFD2的表达。MTHFD2过表达拮抗LPS诱导的活力降低,凋亡促进,活性氧过度产生,NRK-52E细胞的线粒体膜电位崩溃。相比之下,MTHFD2敲低和EP300消融带来了相反的后果。此外,MTHFD2过表达和EP300消融在LPS暴露的NRK-52E细胞中相互抵消作用。EP300介导的H3乙酰化升高MTHFD2表达以减少SI-AKI中肾小管上皮细胞的线粒体功能障碍。
    Sepsis represents an organ dysfunction resulting from the host\'s maladjusted response to infection, and can give rise to acute kidney injury (AKI), which significantly increase the morbidity and mortality of septic patients. This study strived for identifying a novel therapeutic strategy for patients with sepsis-induced AKI (SI-AKI). Rat tubular epithelial NRK-52E cells were subjected to lipopolysaccharide (LPS) exposure for induction of in-vitro SI-AKI. The expressions of E1A binding protein p300 (EP300) and methylenetetrahydrofolate dehydrogenase 2 (MTHFD2) in NRK-52E cells were assessed by western blot and qRT-PCR, and their interaction was explored by chromatin immunoprecipitation performed with antibody for H3K27 acetylation (H3K27ac). The effect of them on SI-AKI-associated mitochondrial dysfunction of tubular epithelial cells was investigated using transfection, MTT assay, TUNEL staining, 2\',7\'-Dichlorodihydrofluorescein diacetate probe assay, Mitosox assay, and JC-1 staining. MTHFD2 and EP300 were upregulated by LPS exposure in NRK-52E cells. LPS increased the acetylation of H3 histone in the MTHFD2 promoter region, and EP300 suppressed the effect of LPS. EP300 ablation inhibited the expression of MTHFD2. MTHFD2 overexpression antagonized LPS-induced viability reduction, apoptosis promotion, reactive oxygen species overproduction, and mitochondrial membrane potential collapse of NRK-52E cells. By contrast, MTHFD2 knockdown and EP300 ablation brought about opposite consequences. Furthermore, MTHFD2 overexpress and EP300 ablation counteracted each other\'s effect in LPS-exposed NRK-52E cells. EP300-mediated H3 acetylation elevates MTHFD2 expression to reduce mitochondrial dysfunction of tubular epithelial cells in SI-AKI.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    伤口愈合受损是2型糖尿病(T2D)的主要临床并发症之一,也是下肢截肢的主要原因。糖尿病伤口表现出持续的炎症状态,减少炎症对糖尿病伤口管理至关重要。巨噬细胞是伤口愈合的关键调节剂,它们的功能障碍会导致糖尿病伤口的炎症加剧和愈合不良。组蛋白修饰引起的基因调控可影响糖尿病伤口愈合过程中的巨噬细胞表型和功能。最近的研究表明,靶向组蛋白修饰酶在局部,巨噬细胞特异性方式可以减轻炎症反应,改善糖尿病创面愈合。本文将对巨噬细胞表型和功能在创面愈合中的意义进行综述,以及说明组蛋白修饰如何影响糖尿病伤口中的巨噬细胞极化。用组蛋白修饰酶靶向巨噬细胞表型可能为糖尿病伤口愈合的治疗提供新的治疗策略。
    Impaired wound healing is one of the main clinical complications of type 2 diabetes (T2D) and a major cause of lower limb amputation. Diabetic wounds exhibit a sustained inflammatory state, and reducing inflammation is crucial to diabetic wounds management. Macrophages are key regulators in wound healing, and their dysfunction would cause exacerbated inflammation and poor healing in diabetic wounds. Gene regulation caused by histone modifications can affect macrophage phenotype and function during diabetic wound healing. Recent studies have revealed that targeting histone-modifying enzymes in a local, macrophage-specific manner can reduce inflammatory responses and improve diabetic wound healing. This article will review the significance of macrophage phenotype and function in wound healing, as well as illustrate how histone modifications affect macrophage polarization in diabetic wounds. Targeting macrophage phenotype with histone-modifying enzymes may provide novel therapeutic strategies for the treatment of diabetic wound healing.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    在由组蛋白修饰或蛋白质结合的基因组区域测量染色质接触的方法是研究染色质组织的重要工具。然而,这些方法不能捕获其他表观基因组特征的可能参与,例如G-四链体DNA二级结构(G4s).为了弥合这个差距,我们介绍ViCAR(观点HiCAR),用于折叠G4s处染色质相互作用的基于抗体的直接捕获。通过ViCAR,我们展示了第一个G4-3D交互景观。使用组蛋白标记,我们还展示了ViCAR如何改进早期的方法,从而提高信噪比。ViCAR是探索表观遗传标记和3D基因组相互作用的实用和强大的工具。
    Methods to measure chromatin contacts at genomic regions bound by histone modifications or proteins are important tools to investigate chromatin organization. However, such methods do not capture the possible involvement of other epigenomic features such as G-quadruplex DNA secondary structures (G4s). To bridge this gap, we introduce ViCAR (viewpoint HiCAR), for the direct antibody-based capture of chromatin interactions at folded G4s. Through ViCAR, we showcase the first G4-3D interaction landscape. Using histone marks, we also demonstrate how ViCAR improves on earlier approaches yielding increased signal-to-noise. ViCAR is a practical and powerful tool to explore epigenetic marks and 3D genome interactomes.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    DELLA蛋白是保守的主要生长调节剂,在响应内部和环境线索控制植物发育中起核心作用。DELLA作为转录调节因子,通过其GRAS结构域与转录因子(TF)和组蛋白H2A结合而募集到靶启动子。最近的研究表明,DELLA的稳定性通过两种机制在翻译后受到调节,植物激素赤霉素诱导的聚泛素化快速降解,和小泛素样修饰剂(SUMO)-缀合以增加其积累。此外,DELLA活性由两种不同的糖基化动态调节:O-岩藻糖基化增强DELLA-TF相互作用,但受到O-连接的N-乙酰葡糖胺(O-GlcNAc)修饰的抑制。然而,DELLA磷酸化的作用目前尚不清楚,因为之前的研究显示结果相互矛盾,从表明磷酸化促进或减少DELLA降解的发现到表明它对其稳定性没有影响的其他发现.这里,我们确定了ga1-3的REPRESSOR中的磷酸化位点(RGA,AtDELLA)通过质谱分析从拟南芥中纯化,并且显示PolyS和PolyS/T区域中两个RGA肽的磷酸化通过促进H2A结合和RGA与靶启动子的缔合来增强RGA活性。值得注意的是,磷酸化不影响RGA-TF相互作用或RGA稳定性。我们的研究揭示了磷酸化诱导DELLA活性的分子机制。
    DELLA proteins are conserved master growth regulators that play a central role in controlling plant development in response to internal and environmental cues. DELLAs function as transcription regulators, which are recruited to target promoters by binding to transcription factors (TFs) and histone H2A via their GRAS domain. Recent studies showed that DELLA stability is regulated post-translationally via two mechanisms, phytohormone gibberellin-induced polyubiquitination for its rapid degradation, and Small Ubiquitin-like Modifier (SUMO)-conjugation to increase its accumulation. Moreover, DELLA activity is dynamically modulated by two distinct glycosylations: DELLA-TF interactions are enhanced by O-fucosylation, but inhibited by O-linked N-acetylglucosamine (O-GlcNAc) modification. However, the role of DELLA phosphorylation remains unclear as previous studies showing conflicting results ranging from findings that suggest phosphorylation promotes or reduces DELLA degradation to others indicating it has no effect on its stability. Here, we identify phosphorylation sites in REPRESSOR OF ga1-3 (RGA, an AtDELLA) purified from Arabidopsis by mass spectrometry analysis, and show that phosphorylation of two RGA peptides in the PolyS and PolyS/T regions enhances RGA activity by promoting H2A binding and RGA association with target promoters. Notably, phosphorylation does not affect RGA-TF interactions or RGA stability. Our study has uncovered a molecular mechanism of phosphorylation-induced DELLA activity.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    DNMT1是一种必需的DNA甲基转移酶,可催化甲基转移到DNA中的CpG岛,并产生突出的表观遗传标记。DNMT1的催化活性依赖于其构象可塑性和将构象从自抑制状态改变为活化状态的能力。这里,我们提出了apoDNMT1和DNTM1的四种低温EM重建:非生产性DNA,DNTM1:H3Ub2-肽,DNTM1:生产性DNA复合物。我们的结构证明了DNMT1的N端调节域在从apo“自动抑制”过渡到DNA结合的“非生产性”以及最终的DNA结合的“生产性”过渡过程中的灵活性。DNMT1的状态。此外,我们讨论了DNMT1选择性小分子抑制剂和泛素化组蛋白H3对DNMT1甲基转移酶活性的调节。我们观察到,尽管存在抑制剂,DNMT1仍以“非生产性”状态结合DNA,并在与双泛素化H3肽类似物的复合物中呈现全长DNMT1的低温EM重建。一起来看,我们的结果提供了DNMT1反应循环的结构见解。
    DNMT1 is an essential DNA methyltransferase that catalyzes the transfer of methyl groups to CpG islands in DNA and generates a prominent epigenetic mark. The catalytic activity of DNMT1 relies on its conformational plasticity and ability to change conformation from an auto-inhibited to an activated state. Here, we present four cryo-EM reconstructions of apo DNMT1 and DNTM1: non-productive DNA, DNTM1: H3Ub2-peptide, DNTM1: productive DNA complexes. Our structures demonstrate the flexibility of DNMT1\'s N-terminal regulatory domains during the transition from an apo \'auto-inhibited\' to a DNA-bound \'non-productive\' and finally a DNA-bound \'productive\' state of DNMT1. Furthermore, we address the regulation of DNMT1\'s methyltransferase activity by a DNMT1-selective small-molecule inhibitor and ubiquitinated histone H3. We observe that DNMT1 binds DNA in a \'non-productive\' state despite the presence of the inhibitor and present the cryo-EM reconstruction of full-length DNMT1 in complex with a di-ubiquitinated H3 peptide analogue. Taken together, our results provide structural insights into the reaction cycle of DNMT1.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号