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
    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: 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)

  • 文章类型: Journal Article
    海马体的适当形成对于大脑执行记忆和学习功能至关重要。然而,关于海马的锥体神经元(PNs)如何成熟和精确位置,仍然存在许多问题。在这里,我们揭示了Sett2,组蛋白3赖氨酸36三甲基化(H3K36me3)的甲基转移酶,对于海马CA1中PNs的精确定位和成熟至关重要。Sett2在神经祖细胞中的消融导致CA1的不规则分层和地层中PN的数量增加。有丝分裂后神经元中的Sett2缺失会导致CA1PN的定位错误和不成熟。转录组分析显示,SETD2维持了成簇的原钙粘蛋白(cPcdh)基因的表达。一起,Sett2是适当的海马分层和CA1PN成熟所必需的。
    Proper formation of the hippocampus is crucial for the brain to execute memory and learning functions. However, many questions remain regarding how pyramidal neurons (PNs) of the hippocampus mature and precisely position. Here we revealed that Setd2, the methyltransferase for histone 3 lysine 36 trimethylation (H3K36me3), is essential for the precise localization and maturation of PNs in the hippocampal CA1. The ablation of Setd2 in neural progenitors leads to irregular lamination of the CA1 and increased numbers of PNs in the stratum oriens. Setd2 deletion in postmitotic neurons causes mislocalization and immaturity of CA1 PNs. Transcriptome analyses revealed that SETD2 maintains the expressions of clustered protocadherin (cPcdh) genes. Together, Setd2 is required for proper hippocampal lamination and maturation of CA1 PNs.
    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
    聚(ADP-核糖基)化(PARylation)是一种关键的翻译后修饰,通过多种分子机制在维持基因组稳定性中起着至关重要的作用。包括复制应激的激活和DNA损伤反应。nudix水解酶NUDT16最近被鉴定为磷酸二酯酶,负责去除ADP-核糖单元,并在DNA修复中起重要作用。然而,NUDT16在协调复制应激和细胞周期进程中的作用仍然难以捉摸.这里,我们报道了SETD3,它是含有SET结构域的蛋白质(SETD)家族的成员,是NUDT16的新底物,其蛋白质水平在细胞周期进程中波动,并且其稳定性受NUDT16介导的去PARA严格调控。此外,我们的数据表明E3连接酶CHFR以PARP1介导的PAR化依赖性方式负责内源性SETD3的识别和降解.机械上,我们发现SETD3与BRCA2相关,并在复制应激或DNA双链断裂时促进其募集到停滞的复制叉和DNA损伤位点,分别。此外,我们证明了SETD3通过减弱R-loop诱导的复制应激来正向调节细胞周期进程,并促进同源重组(HR)修复以响应DNA双链断裂,导致癌细胞对辐射(IR)治疗的抵抗力显着增加。重要的是,NUDT16缺陷细胞中SETD3的耗竭并没有进一步加剧DNA断裂或增强癌细胞对IR暴露的敏感性,提示NUDT16-SETD3通路可能在诱导放疗耐受中起关键作用.总的来说,这些数据表明,NUDT16通过逆转SETD3的ADP-核糖基化而发挥SETD3蛋白稳定性的关键上游调节因子的作用,并且NUDT16参与复制应激的解决并促进HR修复.
    Poly(ADP-ribosyl)ation (PARylation) is a critical posttranslational modification that plays a vital role in maintaining genomic stability via a variety of molecular mechanisms, including activation of replication stress and the DNA damage response. The nudix hydrolase NUDT16 was recently identified as a phosphodiesterase that is responsible for removing ADP-ribose units and that plays an important role in DNA repair. However, the roles of NUDT16 in coordinating replication stress and cell cycle progression remain elusive. Here, we report that SETD3, which is a member of the SET-domain containing protein (SETD) family, is a novel substrate for NUDT16, that its protein levels fluctuate during cell cycle progression, and that its stability is strictly regulated by NUDT16-mediated dePARylation. Moreover, our data indicated that the E3 ligase CHFR is responsible for the recognition and degradation of endogenous SETD3 in a PARP1-mediated PARylation-dependent manner. Mechanistically, we revealed that SETD3 associates with BRCA2 and promotes its recruitment to stalled replication fork and DNA damage sites upon replication stress or DNA double-strand breaks, respectively. Importantly, depletion of SETD3 in NUDT16-deficient cells did not further exacerbate DNA breaks or enhance the sensitivity of cancer cells to IR exposure, suggesting that the NUDT16-SETD3 pathway may play critical roles in the induction of tolerance to radiotherapy. Collectively, these data showed that NUDT16 functions as a key upstream regulator of SETD3 protein stability by reversing the ADP-ribosylation of SETD3, and NUDT16 participates in the resolution of replication stress and facilitates HR repair.
    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
    前列腺癌(PCa)是最常见的恶性肿瘤之一,同时表现出耐药性和复发。SUV39H2在许多类型的人类肿瘤中高表达,但其在PCa发展和发展中的作用从未得到澄清。本研究的目的是阐明SUV39H2在PCa发生发展中的作用,它与AKT/FOXO信号通路的关联,及其对PCa诊断和治疗的潜在影响。在癌症基因组图谱(TCGA)和基因型组织表达泛癌症数据中分析SUV39H2表达。评估TCGA数据库的SUV39H2富集及其与免疫细胞浸润的相关性。使用qPCR和IHC在30名患者中测定PCa组织和对照组织中的SUV39H2水平。通过癌症基因组图谱(TCGA)评估临床相关性。体外评估,包括集落形成测定,西方印迹分析,CCK-8测定,和流式细胞术用于确定SUV39H2对PCa细胞生长的贡献。通过细胞系衍生的异种移植模型评估SUV39H2对PC3和DU145细胞增殖的影响。球体形成测定和qPCR用于描述SUV39H2在PCa干性和化学敏感性中的作用。体外巨噬细胞极化试验提供了对SUV39H2与M2巨噬细胞关联的见解,而富集分析揭示了其在FOXO信号传导中的作用。PCa组织表达的SUV39H2水平高于正常组织。通过敲低SUV39H2,PCa细胞对多西他赛的化学敏感性更高,细胞增殖和干细胞被抑制。此外,SUV39H2敲低可显著抑制体内PCa细胞的生长,抑制巨噬细胞的极化。此外,发现SUV39H2通过增加Akt和FOXO3a磷酸化来调节AKT/FOXO信号传导。我们的发现强调了SUV39H2主要通过调节AKT/FOXO信号通路在PCa细胞凋亡和化疗敏感性中的作用,并表明SUV39H2可能是PCa诊断和治疗的潜在靶标。
    Prostate cancer (PCa) is one of the most common malignant tumors that exhibit both chemoresistance and recurrence. SUV39H2 is highly expressed in many types of human tumors, but its role in the development and progression of PCa has never been clarified. The aim of this study is to elucidate the role of SUV39H2 in the development and progression of PCa, its association with the AKT/FOXO signaling pathway, and its potential implications for PCa diagnosis and treatment. SUV39H2 expression was analyzed in The Cancer Genome Atlas (TCGA) and genotype tissue expression pan-cancer data. The TCGA database was evaluated for SUV39H2 enrichment and its correlation to immune cell infiltration. SUV39H2 levels in PCa tissues and control tissues were determined in 30 patients using qPCR and IHC. Clinical relevance was assessed via The Cancer Genome Atlas (TCGA). In vitro assessments including colony formation assays, Western Blot analysis, CCK-8 assays, and flow cytometry were utilized to establish SUV39H2\'s contribution to PCa cell growth. The influence of SUV39H2 on PC3 and DU145 cell proliferation was assessed through a cell line-derived xenograft model. Sphere formation assays and qPCR were employed to delineate SUV39H2\'s role in PCa stemness and chemosensitivity. In vitro macrophage polarization assays provided insights into SUV39H2\'s association with M2 macrophages, while enrichment analysis shed light on its role in FOXO signaling. PCa tissues expressed higher levels of SUV39H2 than normal tissues. By knocking down SUV39H2, PCa cells were made more chemosensitive to docetaxel and cell proliferation and stemness were inhibited. Additionally, SUV39H2 knockdown significantly inhibited in vivo PCa cell growth and inhibited the polarization of macrophages. Furthermore, SUV39H2 was found to regulate AKT/FOXO signaling by increasing Akt and FOXO3a phosphorylation. Our findings highlight SUV39H2\'s role in PCa cell apoptosis and chemosensitivity mainly by regulating the AKT/FOXO signaling pathway and suggest that SUV39H2 could be a potential target for PCa diagnosis and treatment.
    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
    组蛋白甲基转移酶(HMTs)在基因翻译后调控和多种生理过程中发挥重要作用。并与许多人类疾病有关,尤其是癌症。越来越多的证据表明,HMT可以作为癌症治疗的潜在治疗靶点。因此,在过去的十年中,HMTs抑制剂的开发一直受到越来越多的关注.然而,临床疗效不足等缺点,适度的选择性,和获得性抗性的倾向阻碍了常规HMT抑制剂的发展。新的技术和方法对于增强HMT抑制剂的抗癌活性至关重要。在这次审查中,我们首先回顾了几种重要的HMT的结构和生物学功能,如EZH2、G9a、PRMT5和DOT1L。阐述了这些HMTs与癌症的内在联系。接下来,我们主要关注包括双靶点抑制剂在内的HMT调节剂开发的最新进展,从合理设计等角度出发的靶向蛋白质降解剂和共价抑制剂,药效学,药代动力学,和临床状态。最后,我们还讨论了基于HMT的癌症治疗药物发现的挑战和未来方向.
    Histone methyltransferases (HMTs) play a critical role in gene post-translational regulation and diverse physiological processes, and are implicated in a plethora of human diseases, especially cancer. Increasing evidences demonstrate that HMTs may serve as a potential therapeutic target for cancer treatment. Thus, the development of HMTs inhibitor have been pursued with steadily increasing interest over the past decade. However, the disadvantages such as insufficient clinical efficacy, moderate selectivity, and propensity for acquired resistance have hindered the development of conventional HMT inhibitors. New technologies and methods are imperative to enhance the anticancer activity of HMT inhibitors. In this review, we first review the structure and biological functions of the several essential HMTs, such as EZH2, G9a, PRMT5, and DOT1L. The internal relationship between these HMTs and cancer is also expounded. Next, we mainly focus on the latest progress in the development of HMT modulators encompassing dual-target inhibitors, targeted protein degraders and covalent inhibitors from perspectives such as rational design, pharmacodynamics, pharmacokinetics, and clinical status. Lastly, we also discuss the challenges and future directions for HMT-based drug discovery for cancer therapy.
    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: Case Reports
    背景:Sotos综合征是一种先天性过度生长综合征,其主要特征包括过度生长,独特的面部特征,学习障碍,并伴随着各种第二特征。NSD1缺失或突变是主要致病原因。尽管世界上有一些关于治疗这种疾病的报道,在中国,正在接受治疗的病例较少。
    方法:一个1岁男孩患有大头畸形,巨型,过高的身体高度,特殊的面孔和延迟的发展,具有NSD1的致病基因(外显子5中的NM_022455.5:c.3536delA)。
    方法:患儿明确诊断为Sotos综合征,有3个月的中医康复综合治疗。
    结果:孩子在全球发展中取得了巨大进步。
    结论:本病例首次描述了中国治疗Sotos综合征的中医康复治疗方法。没有根治方法,但我们的治疗可以改善患者的预后和生活质量。因此,为Sotos综合征的临床治疗提供参考。
    BACKGROUND: Sotos syndrome is an congenital overgrowth syndrome characterized by the primary features including overgrowth, distinctive facial features, learning disability, and accompanied with various second features. NSD1 deletion or mutation is a major pathogenic cause. Although there are some reports on treatment of this disease worldwide, less cases under treatment have been published in China.
    METHODS: A 1-year-old boy had macrocephaly, gigantism, excessive high body height, a particular face and delayed development, with a pathogenic gene of NSD1 (NM_022455.5:c.3536delA in exon 5).
    METHODS: The child was definitely diagnosed as Sotos syndrome and have 3 months\' combination treatment of traditional Chinese medicine and rehabilitation.
    RESULTS: The child made a great progress in global development.
    CONCLUSIONS: This case firstly describes the traditional Chinese medicine and rehabilitation to treat Sotos syndrome in China. There is no radical cure, but our therapy could improve the prognosis and the life quality of the patient. Therefore, this case provides a reference to the clinical treatment of Sotos syndrome.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号