Histone Methyltransferases

组蛋白甲基转移酶
  • 文章类型: Journal Article
    SETD3是与病毒蛋白酶2A特异性相互作用的多种肠道病毒复制的必需宿主因子。然而,SETD3和2A蛋白酶之间的相互作用尚未得到充分表征。这里,我们使用X射线晶体学和低温电子显微镜来确定与EV71的2A蛋白酶复合的SETD3的结构,分辨率为3.5和3.1,分别。我们发现2A蛋白酶通过两个离散位点占据了SETD3的V形中央裂口。两种蛋白质的相对位置在晶体和低温-EM结构中有所不同,显示动态绑定。生物层干涉测定表明,EV712A蛋白酶超过肌动蛋白与SETD3的结合。我们鉴定了参与SETD3结合的关键2A残基,并证明2A结合SETD3的能力与细胞中EV71的产生相关。在EV71感染和2A表达细胞中的免疫共沉淀实验表明2A干扰SETD3-肌动蛋白复合物,这种复合物的破坏减少了肠道病毒的复制。一起,这些结果揭示了SETD3、肌动蛋白、和病毒2A在病毒复制过程中。
    SETD3 is an essential host factor for the replication of a variety of enteroviruses that specifically interacts with viral protease 2A. However, the interaction between SETD3 and the 2A protease has not been fully characterized. Here, we use X-ray crystallography and cryo-electron microscopy to determine the structures of SETD3 complexed with the 2A protease of EV71 to 3.5 Å and 3.1 Å resolution, respectively. We find that the 2A protease occupies the V-shaped central cleft of SETD3 through two discrete sites. The relative positions of the two proteins vary in the crystal and cryo-EM structures, showing dynamic binding. A biolayer interferometry assay shows that the EV71 2A protease outcompetes actin for SETD3 binding. We identify key 2A residues involved in SETD3 binding and demonstrate that 2A\'s ability to bind SETD3 correlates with EV71 production in cells. Coimmunoprecipitation experiments in EV71 infected and 2A expressing cells indicate that 2A interferes with the SETD3-actin complex, and the disruption of this complex reduces enterovirus replication. Together, these results reveal the molecular mechanism underlying the interplay between SETD3, actin, and viral 2A during virus replication.
    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
    目的:Sotos综合征(SOTOS)是一种罕见的遗传病,表现出以下独特特征:产前过度生长,面部异常,智力残疾。这种疾病通常与核受体结合SET结构域蛋白1(NSD1)基因的单倍体不足有关。我们调查了4例以早发性过度生长和发育迟缓为特征的儿科病例。这项研究的主要目的是实现准确的遗传诊断。
    一种包括染色体核型分析的序贯分析方法,整个外显子组测序,并进行微阵列分析。
    结果:所有4例病例均表现出NSD1基因变异,通过鉴定四个以前未报告的从头变体,每个具体到一个案例。具体来说,案例1携带NSD1(NM_022455):c.2686C>T(p。Q896X)变体,案例2具有NSD1(NM_022455):c.2858_2859delCT(p。3953X)变体,病例3显示染色体畸变,chr5:5q35.2q35.3(176,516,604-176,639,249)×1,包含NSD1的5'-非翻译区,案例4包含NSD1(NM_022455):c.6397T>G(p。C2133G)变体。
    结论:这项研究不仅为这些病例提供了精确的诊断,而且为促进知情咨询提供了重要的证据。此外,我们的发现扩大了与SOTOS相关的突变范围.
    OBJECTIVE: Sotos syndrome (SOTOS) is an uncommon genetic condition that manifests itself with the following distinctive features: prenatal overgrowth, facial abnormalities, and intellectual disability. This disorder is often associated with haploinsufficiency of the nuclear receptor-binding SET domain protein 1 (NSD1)gene. We investigated four pediatric cases characterized by early-onset overgrowth and developmental delay. The primary objective of this study was to achieve accurate genetic diagnoses.
    UNASSIGNED: A sequential analysis approach comprising chromosomal karyotyping, whole exome sequencing, and microarray analysis was conducted.
    RESULTS: All four cases exhibited variations in the NSD1 gene, with the identification of four previously unreported de novo variants, each specific to one case.Specifically, Case 1 carried the NSD1 (NM_022455): c.2686 C > T(p.Q896X) variant, Case 2 had the NSD1 (NM_022455): c.2858_2859delCT(p.S953X) variant, Case 3 displayed a chromosomal aberration, chr5: 5q35.2q35.3(176,516,604-176,639,249)×1, which encompassed the 5\'-untranslated region of NSD1, and Case 4 harbored the NSD1 (NM_022455): c.6397T > G(p.C2133G) variant.
    CONCLUSIONS: This study not only provided precise diagnoses for these cases but also supplied significant evidence to facilitate informed consultations. Furthermore, our findings expanded the spectrum of mutations associated with SOTOS.
    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
    Graves眼病(GO)是Graves病的甲状腺外并发症,严重时会导致视力丧失。目前,GO的治疗不够有效,所以需要新的治疗策略。由于血小板衍生生长因子(PDGF)-BB在GO轨道成纤维细胞中诱导几种效应机制,包括细胞因子产生和肌成纤维细胞活化,本研究旨在通过HKMTs抑制剂库筛选,探讨组蛋白赖氨酸甲基转移酶(HKMTs)在PDGF-BB激活的GO轨道成纤维细胞中的作用。从库中的十二种选择性HKMT抑制剂中,EZH2,G9a和DOT1L抑制剂,DZNeP,BIX01294和Pinometostat,分别,通过GO轨道成纤维细胞阻止PDGF-BB诱导的增殖和透明质酸的产生。然而,只有EZH2抑制剂,DZNeP,显著阻断促炎细胞因子的产生。对于GO眶成纤维细胞中的HKMTs表达,PDGF-BB显著且时间依赖性地诱导EZH2、G9a和DOT1LmRNA表达。为了证实EZH2在PDGF-BB诱导的眼眶成纤维细胞活化中的作用,EZH2沉默实验显示抑制PDGF-BB诱导的I型胶原蛋白和α-SMA表达,同时降低组蛋白H3赖氨酸27三甲基化(H3K27me3)水平。在比眼眶成纤维细胞培养实验更临床相关的模型中,DZNeP处理的GO眼眶组织显著降低促炎细胞因子产生,同时略微降低ACTA2mRNA表达。我们的数据首次证明,在所有HKMTs中,EZH2主要参与来自GO的PDGF-BB激活的眼眶成纤维细胞中肌成纤维细胞标志物的表达,大概是通过H3K27me3。因此,EZH2可以代表GO的新型治疗靶标。
    Graves\' ophthalmopathy (GO) is an extra-thyroidal complication of Graves\' disease which can lead to vision loss in severe cases. Currently, treatments of GO are not sufficiently effective, so novel therapeutic strategies are needed. As platelet-derived growth factor (PDGF)-BB induces several effector mechanisms in GO orbital fibroblasts including cytokine production and myofibroblast activation, this study aims to investigate the roles of histone lysine methyltransferases (HKMTs) in PDGF-BB-activated GO orbital fibroblasts by screening with HKMTs inhibitors library. From the total of twelve selective HKMT inhibitors in the library, EZH2, G9a and DOT1L inhibitors, DZNeP, BIX01294 and Pinometostat, respectively, prevented PDGF-BB-induced proliferation and hyaluronan production by GO orbital fibroblasts. However, only EZH2 inhibitor, DZNeP, significantly blocked pro-inflammatory cytokine production. For the HKMTs expression in GO orbital fibroblasts, PDGF-BB significantly and time-dependently induced EZH2, G9a and DOT1L mRNA expression. To confirm the role of EZH2 in PDGF-BB-induced orbital fibroblast activation, EZH2 silencing experiments revealed suppression of PDGF-BB-induced collagen type I and α-SMA expression along with decreasing histone H3 lysine 27 trimethylation (H3K27me3) level. In a more clinically relevant model than orbital fibroblast culture experiments, DZNeP treated GO orbital tissues significantly reduced pro-inflammatory cytokine production while slightly reduced ACTA2 mRNA expression. Our data is the first to demonstrate that among all HKMTs EZH2 dominantly involved in the expression of myofibroblast markers in PDGF-BB-activated orbital fibroblast from GO presumably via H3K27me3. Thus, EZH2 may represent a novel therapeutics target for GO.
    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
    MYCN激活参与核糖体生物发生的典型MYC靶标,蛋白质合成,并抑制神经元分化基因以驱动神经母细胞瘤(NB)的肿瘤发生。MYCN如何协调全球基因表达仍未完全了解。我们的研究发现,MYCN结合启动子来上调典型的MYC靶标,但同时结合增强子和启动子来抑制分化基因。MYCN结合还增加了典型MYC靶启动子上的H3K4me3和H3K27ac,并减少了神经元分化基因增强子和启动子上的H3K27ac。WDR5促进MYCN启动子结合以激活典型的MYC靶基因,而MYCN招募G9a增强子来抑制神经元分化基因。使用WDR5和G9a抑制剂靶向MYCN的活性和抑制转录活性协同抑制NB生长。我们证明了MYCN与WDR5和G9a合作来协调全局基因转录。这两种辅因子的靶向是间接靶向MYCN的致癌活性的新型治疗策略。
    MYCN activates canonical MYC targets involved in ribosome biogenesis, protein synthesis, and represses neuronal differentiation genes to drive oncogenesis in neuroblastoma (NB). How MYCN orchestrates global gene expression remains incompletely understood. Our study finds that MYCN binds promoters to up-regulate canonical MYC targets but binds to both enhancers and promoters to repress differentiation genes. MYCN binding also increases H3K4me3 and H3K27ac on canonical MYC target promoters and decreases H3K27ac on neuronal differentiation gene enhancers and promoters. WDR5 facilitates MYCN promoter binding to activate canonical MYC target genes, whereas MYCN recruits G9a to enhancers to repress neuronal differentiation genes. Targeting both MYCN\'s active and repressive transcriptional activities using both WDR5 and G9a inhibitors synergistically suppresses NB growth. We demonstrate that MYCN cooperates with WDR5 and G9a to orchestrate global gene transcription. The targeting of both these cofactors is a novel therapeutic strategy to indirectly target the oncogenic activity of MYCN.
    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
    转录因子NRF2通过协调氧化应激依赖性转录程序在维持氧化还原和代谢稳态中起关键作用。尽管越来越多的证据表明,在翻译后阶段,各种细胞成分参与了NRF2活性的调节,对氧化应激反应中决定NRF2转录激活的因素的了解相对较少。在这项研究中,我们报道了MLL1的关键作用,MLL1是一种H3K4特异性甲基转移酶,和UTX,一种H3K27特异性组蛋白去甲基酶,在氧化应激下NRF2依赖性转录程序中。我们发现MLL1或UTX的消耗导致对氧化应激的敏感性增加,伴随着较高的细胞内ROS和抗氧化基因的失败激活,包括NRF2。此外,MLL1和UTX选择性靶向NRF2启动子,和外源FLAG-NRF2表达增加MLL1或UTX耗尽的细胞在暴露于过氧化氢时的活力。RNA-seq分析表明MLL1或UTX的消耗影响响应于氧化应激的NRF2依赖性转录组的变化。此外,ChIP和ChIP-seq分析发现,MLL1和UTX在功能上合作建立了一个染色质环境,该环境有利于响应ROS诱导的氧化应激而在H3K4me3/H3K27me3二价NRF2启动子处进行主动转录。总的来说,这些发现提供了细胞对氧化应激反应的分子机制,并强调了染色质结构和功能在维持氧化还原稳态中的重要性。
    The transcription factor NRF2 plays a pivotal role in maintaining redox and metabolic homeostasis by orchestrating oxidative stress-dependent transcription programs. Despite growing evidence implicating various cellular components in the regulation of NRF2 activity at the posttranslational stage, relatively less is known about the factors dictating the transcriptional activation of NRF2 in response to oxidative stress. In this study, we report the crucial roles of MLL1, an H3K4-specific methyltransferase, and UTX, an H3K27-specific histone demethylase, in the NRF2-dependent transcription program under oxidative stress. We find that the depletion of MLL1 or UTX results in increased susceptibility to oxidative stress, accompanied by higher intracellular ROS and the failed activation of antioxidant genes, including NRF2. In addition, MLL1 and UTX selectively target the NRF2 promoter, and exogenous FLAG-NRF2 expression increases the viability of MLL1-or UTX-depleted cells upon exposure to hydrogen peroxide. RNA-seq analysis demonstrates that depletion of MLL1 or UTX affects the changes in NRF2-dependent transcriptome in response to oxidative stress. Furthermore, ChIP and ChIP-seq analyses find that MLL1 and UTX functionally cooperate to establish a chromatin environment that favors active transcription at the H3K4me3/H3K27me3 bivalent NRF2 promoter in response to ROS-induced oxidative stress. Collectively, these findings provide a molecular mechanism underlying the cellular response to oxidative stress and highlight the importance of the chromatin structure and function in maintaining redox homeostasis.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    肺癌,在常见恶性肿瘤中,发病率和高死亡率均居全球第二,尽管新的治疗剂不断涌现,但耐药性的频繁发生仍然是一个重大挑战。这会加剧疾病进展,肿瘤复发,最终导致预后不良。除了遗传突变导致的获得性抗性,越来越多的证据表明,表观遗传机制在这一过程中起着至关重要的作用。大量研究表明组蛋白甲基转移酶(HMTs)在肺癌中表达异常,某些HMTs的异常激活与耐药性密切相关。HMTs通过涉及细胞代谢改变的途径介导肺癌的药物耐受性,肿瘤干细胞相关基因的上调,促进上皮-间质转化,增强迁移能力。HMT抑制剂的使用也为肺癌治疗开辟了新的途径,靶向HMT可能有助于逆转耐药性。本文综述了HMTs在肺癌耐药中的关键作用和分子机制。为治疗策略提供了新的视角。通过彻底检查治疗方法,它为了解肺癌的耐药性提供了新的见解,支持个性化治疗,促进药物开发,并将肺癌治疗推向新的领域。
    Lung cancer, ranking second globally in both incidence and high mortality among common malignant tumors, presents a significant challenge with frequent occurrences of drug resistance despite the continuous emergence of novel therapeutic agents. This exacerbates disease progression, tumor recurrence, and ultimately leads to poor prognosis. Beyond acquired resistance due to genetic mutations, mounting evidence suggests a critical role of epigenetic mechanisms in this process. Numerous studies have indicated abnormal expression of Histone Methyltransferases (HMTs) in lung cancer, with the abnormal activation of certain HMTs closely linked to drug resistance. HMTs mediate drug tolerance in lung cancer through pathways involving alterations in cellular metabolism, upregulation of cancer stem cell-related genes, promotion of epithelial-mesenchymal transition, and enhanced migratory capabilities. The use of HMT inhibitors also opens new avenues for lung cancer treatment, and targeting HMTs may contribute to reversing drug resistance. This comprehensive review delves into the pivotal roles and molecular mechanisms of HMTs in drug resistance in lung cancer, offering a fresh perspective on therapeutic strategies. By thoroughly examining treatment approaches, it provides new insights into understanding drug resistance in lung cancer, supporting personalized treatment, fostering drug development, and propelling lung cancer therapy into novel territories.
    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
    胃肠道(GI)癌症被认为主要是遗传性恶性肿瘤,由一系列渐进的遗传改变引起的。越来越多的证据表明组蛋白甲基化,表观遗传修饰计划,在胃肠道癌症进展的不同病理阶段中起着至关重要的作用,如癌前病变,肿瘤发生,和肿瘤转移。组蛋白甲基化修饰酶,包括组蛋白甲基转移酶(HMTs)和去甲基酶(HDMs),是转录后修饰的主要执行者。组蛋白甲基化修饰酶的异常表达表征具有复杂发病机制和进展的GI癌症。上游控制器和组蛋白甲基化修饰酶之间的相互作用最近被揭示,并提供了许多机会来深入和清楚地阐明胃肠道癌症的发病机制。在这里,我们专注于组蛋白甲基化修饰酶和它们的控制器之间的关联,旨在为胃肠道肿瘤的分子研究和临床管理提供新的视角。
    Gastrointestinal (GI) cancers have been considered primarily genetic malignancies, caused by a series of progressive genetic alterations. Accumulating evidence shows that histone methylation, an epigenetic modification program, plays an essential role in the different pathological stages of GI cancer progression, such as precancerous lesions, tumorigenesis, and tumor metastasis. Histone methylation-modifying enzymes, including histone methyltransferases (HMTs) and demethylases (HDMs), are the main executor of post-transcriptional modification. The abnormal expression of histone methylation-modifying enzymes characterizes GI cancers with complex pathogenesis and progression. Interactions between upstream controllers and histone methylation-modifying enzymes have recently been revealed, and have provided numerous opportunities to elucidate the pathogenesis of GI cancers in depth and clearly. Here we focus on the association between histone methylation-modifying enzymes and their controllers, aiming to provide a new perspective on the molecular research and clinical management of GI cancers.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    比较方法揭示了实现共同发展成果的不同和趋同路径。因此,只有通过组合多个案例研究,我们才能理解生物学原理。然而,尽管认识到发展网络的保护或缺乏保护,调节这些网络的表观遗传机制的保守性知之甚少。线虫Pristionchuspacificus已成为可塑性和表观遗传调控的模型系统,因为它根据其环境表现出细菌或杂食性形态。这里,我们确定了可用于P.pacificus的“表观遗传工具包”,作为未来可塑性功能工作的资源,并与秀丽隐杆线虫进行比较,以研究表观遗传机制的保守性。广义上,我们观察到在秀丽隐杆线虫和太平洋疟原虫之间具有类似的表观遗传功能的基因。然而,我们还发现了惊人的差异。最值得注意的是,组蛋白甲基转移酶复合物PRC2似乎在太平洋疟原虫中缺失。我们描述了PRC2基因mes-2和mes-6的缺失/假基因化,并得出结论,这两个基因都在P.pacificus和相关物种P.arcanus的最后一个共同祖先中丢失。有趣的是,我们通过质谱和免疫荧光法观察了PRC2(H3K27me3)的酶产物,表明目前未知的甲基转移酶已被选择用于异染色质沉默。总之,我们提供了pacificus的表观遗传基因清单,以与秀丽隐杆线虫进行比较。该清单将使与可塑性相关的反向遗传实验成为可能,并揭示了多细胞生物中PRC2的首次丢失。
    Comparative approaches have revealed both divergent and convergent paths to achieving shared developmental outcomes. Thus, only through assembling multiple case studies can we understand biological principles. Yet, despite appreciating the conservation-or lack thereof-of developmental networks, the conservation of epigenetic mechanisms regulating these networks is poorly understood. The nematode Pristionchus pacificus has emerged as a model system of plasticity and epigenetic regulation as it exhibits a bacterivorous or omnivorous morph depending on its environment. Here, we determined the \"epigenetic toolkit\" available to P. pacificus as a resource for future functional work on plasticity, and as a comparison with Caenorhabditis elegans to investigate the conservation of epigenetic mechanisms. Broadly, we observed a similar cast of genes with putative epigenetic function between C. elegans and P. pacificus. However, we also found striking differences. Most notably, the histone methyltransferase complex PRC2 appears to be missing in P. pacificus. We described the deletion/pseudogenization of the PRC2 genes mes-2 and mes-6 and concluded that both were lost in the last common ancestor of P. pacificus and a related species P. arcanus. Interestingly, we observed the enzymatic product of PRC2 (H3K27me3) by mass spectrometry and immunofluorescence, suggesting that a currently unknown methyltransferase has been co-opted for heterochromatin silencing. Altogether, we have provided an inventory of epigenetic genes in P. pacificus to compare with C. elegans. This inventory will enable reverse-genetic experiments related to plasticity and has revealed the first loss of PRC2 in a multicellular organism.
    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赖氨酸4单甲基化(H3K4me1)标志着平衡的或有活性的增强子。KMT2C(MLL3)和KMT2D(MLL4)催化H3K4me1,但它们的组蛋白甲基转移酶活性对于哺乳动物早期胚胎发生过程中的转录在很大程度上是不必要的。为了更好地理解H3K4me1在增强剂功能中的作用,我们分析了小鼠胚胎干细胞神经分化过程中的动态增强子-启动子(E-P)相互作用和基因表达。我们发现KMT2C/D催化活性仅对于H3K4me1和E-P在候选增强剂的子集接触是必需的,诱导神经分化。相比之下,大多数增强子在KMT2C/D催化突变细胞中保留H3K4me1。令人惊讶的是,这些KMT2C/D独立位点的H3K4me1信号在KMT2B急性耗竭后减少,导致转录缺陷加剧。因此,我们的观察结果暗示KMT2B在增强子上催化H3K4me1,并为H3K4me1在增强子-启动子相互作用和哺乳动物细胞转录中的积极作用提供了额外的支持。
    Histone H3 lysine 4 mono-methylation (H3K4me1) marks poised or active enhancers. KMT2C (MLL3) and KMT2D (MLL4) catalyze H3K4me1, but their histone methyltransferase activities are largely dispensable for transcription during early embryogenesis in mammals. To better understand the role of H3K4me1 in enhancer function, we analyze dynamic enhancer-promoter (E-P) interactions and gene expression during neural differentiation of the mouse embryonic stem cells. We found that KMT2C/D catalytic activities were only required for H3K4me1 and E-P contacts at a subset of candidate enhancers, induced upon neural differentiation. By contrast, a majority of enhancers retained H3K4me1 in KMT2C/D catalytic mutant cells. Surprisingly, H3K4me1 signals at these KMT2C/D-independent sites were reduced after acute depletion of KMT2B, resulting in aggravated transcriptional defects. Our observations therefore implicate KMT2B in the catalysis of H3K4me1 at enhancers and provide additional support for an active role of H3K4me1 in enhancer-promoter interactions and transcription in mammalian cells.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Case Reports
    NSD1的种系突变与Sotos综合征有关,以独特的面部特征为特征,过度生长,和发育迟缓。大约3%的Sotos综合征患者会发展成肿瘤。在这项研究中,我们描述了一个患有面部异常的松果体母细胞瘤的婴儿,学习障碍和轻度自闭症在1年时被诊断为Sotos综合征,这是由于携带新的突变,从头生殖系NSD1可能是致病变异。该患者扩展了Sotos综合征的突变和表型谱,并为潜在的松果体母细胞瘤病理的潜在机制提供了新的临床见解。
    Germline mutations of NSD1 are associated with Sotos syndrome, characterized by distinctive facial features, overgrowth, and developmental delay. Approximately 3% of individuals with Sotos syndrome develop tumors. In this study, we describe an infant in pineoblastoma with facial anomalies, learning disability and mild autism at 1 years diagnosed as Sotos syndrome owing to carrying a novel mutation de novo germline NSD1 likely pathogenic variant. This patient expands both the mutation and phenotype spectrum of the Sotos Syndrome and provides new clinical insights into the potential mechanism of underlying pinealoblastoma pathology.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号