ASH1L

ASH1L
  • 文章类型: Journal Article
    背景:组蛋白甲基转移酶缺失,小,或同源盘1-like(ASH1L)由su(var)3-9,zeste的增强剂组成,三thorax(SET)域,pleckstrin同源域(PHD)域,中间(MID)域,和溴相邻同源(BAH)结构域。已知ASH1L的SET结构域介导H3K36二甲基化(H3K36me2)修饰。然而,PHD-BAH结构域的特定功能在很大程度上仍未被探索。本研究旨在探讨ASH1L中PHD-BAH结构域的生物学功能。
    方法:我们采用了一系列技术,包括原核融合蛋白表达纯化系统,下拉分析,等温滴定量热法(ITC)聚合酶链反应(PCR),和定点诱变,集群定期间隔短回文重复(CRISPR-Cas9)基因编辑,细胞培养实验,westernblot,细胞增殖试验,和细胞凋亡试验。
    结果:ASH1L中的PHD-BAH结构域相对于H3K4单甲基化(H3K4me1)和H3K4三甲基化(H3K4me3)肽优先结合H3K4me2肽。值得注意的是,PHD-BAH结构域内的W2603A突变可在体外破坏与H3K4me2的相互作用.与野生型胆管癌(CHOL)细胞相比,ASH1L中PHD-BAH结构域的缺失导致CHOL细胞凋亡增加和细胞增殖减少(P<0.001)。此外,W2603A突变影响了蛋白酶体20S亚基β(PSMB)家族基因集的调控。
    结论:W2603A突变对于PHD-BAH结构域与H3K4me2肽之间的相互作用至关重要。ASH1L通过调节PSMB家族基因集的表达,通过其PHD-BAH结构域调节CHOL细胞的存活和增殖。
    BACKGROUND: Histone methyltransferase absent, small, or homeotic discs1-like (ASH1L) is composed of su(var)3-9, enhancer of zeste, trithorax (SET) domain, pleckstrin homology domain (PHD) domain, middle (MID) domain, and bromo adjacent homology (BAH) domain. The SET domain of ASH1L is known to mediate mediate H3K36 dimethylation (H3K36me2) modification. However, the specific functions of the PHD-BAH domain remain largely unexplored. This study aimed to explore the biological function of the PHD-BAH domain in ASH1L.
    METHODS: We employed a range of techniques, including a prokaryotic fusion protein expression purification system, pull-down assay, Isothermal Titration Calorimetry (ITC), polymerase chain reaction (PCR), and sitedirected mutagenesis, Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR-Cas9) gene editing, cell culture experiment, western blot, cell proliferation assay, and cell apoptosis test.
    RESULTS: The PHD-BAH domain in ASH1L preferentially binds to the H3K4me2 peptide over H3K4 monomethylation (H3K4me1) and H3K4 trimethylation (H3K4me3) peptide. Notably, the W2603A mutation within the PHD-BAH domain could disrupt the interaction with H3K4me2 in vitro. Compared with wild-type Cholangiocarcinoma (CHOL) cells, deletion of the PHD-BAH domain in ASH1L led to increased CHOL cell apoptosis and reduced cell proliferation (P < 0.001). Additionally, the W2603A mutation affected the regulation of the proteasome 20S subunit beta (PSMB) family gene set.
    CONCLUSIONS: W2603A mutation was crucial for the interaction between the PHD-BAH domain and the H3K4me2 peptide. ASH1L regulated CHOL cell survival and proliferation through its PHD-BAH domain by modulating the expression of the PSMB family gene set.
    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
    致病性ASH1L变体已在先证者中报道,具有广泛的表型表现,包括智力残疾,自闭症谱系障碍,注意缺陷多动障碍,癫痫发作,先天性异常,和其他骨骼,肌肉,睡眠差异这里,我们回顾了以前发表的具有致病性ASH1L变异体的个体,并报告了另外三个具有新ASH1L变异体和以前未报告的表型特征的先证者。包括混合接受性语言障碍和步态障碍。这些来自大脑基因注册的新数据,一个可访问的临床衍生基因型和表型数据储存库,已经允许扩展这种情况的表型和基因型谱。
    Pathogenic ASH1L variants have been reported in probands with broad phenotypic presentations, including intellectual disability, autism spectrum disorder, attention deficit hyperactivity disorder, seizures, congenital anomalies, and other skeletal, muscular, and sleep differences. Here, we review previously published individuals with pathogenic ASH1L variants and report three further probands with novel ASH1L variants and previously unreported phenotypic features, including mixed receptive language disorder and gait disturbances. These novel data from the Brain Gene Registry, an accessible repository of clinically derived genotypic and phenotypic data, have allowed for the expansion of the phenotypic and genotypic spectrum of this condition.
    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
    为了对抗各种传染病,在最近的过去,利用宿主导向疗法作为化疗的替代方案获得了很多关注,因为它绕过了传统疗法的现有局限性。使用宿主表观遗传酶如组蛋白赖氨酸甲基转移酶和赖氨酸去甲基酶作为潜在的药物靶标已经成功地用于控制各种炎性疾病如类风湿性关节炎和急性白血病。在我们之前的研究中,我们已经证明,在内脏利什曼病的实验模型中,KDM6B和ASH1L的功能敲除已导致器官寄生虫负担的显着降低。在这里,我们使用从Maybridge文库和PubChem数据库获得的>53,000种化合物对KDM6B和ASH1L进行了高通量虚拟筛选,然后进行分子对接以评估其对接评分/GlideGscore。根据他们的对接分数,随后评估了所选择的抑制剂的体外抗利什曼钠功效。在针对KDM6B和ASH1L设计的所有抑制剂中,HTS09796,GSK-J4和AS-99特别显示了有希望的体外活性,其IC50<5µM,对多诺瓦尼乳杆菌的细胞外前和细胞内阿马斯蒂戈特形式。这些抑制剂的体外药物相互作用研究进一步证明了它们与两性霉素B和米替福辛的协同相互作用。然而,GSK-J4通过在与米替福辛的不同交互模式中显示一个例外。总的来说,我们的计算机模拟和体外研究为确定这些靶向KDM6B和ASH1L的抑制剂在抗利什曼酶治疗中的适用性提供了平台.
    In order to combat various infectious diseases, the utilization of host-directed therapies as an alternative to chemotherapy has gained a lot of attention in the recent past, since it bypasses the existing limitations of conventional therapies. The use of host epigenetic enzymes like histone lysine methyltransferases and lysine demethylases as potential drug targets has successfully been employed for controlling various inflammatory diseases like rheumatoid arthritis and acute leukemia. In our earlier study, we have already shown that the functional knockdown of KDM6B and ASH1L in the experimental model of visceral leishmaniasis has resulted in a significant reduction of organ parasite burden. Herein, we performed a high throughput virtual screening against KDM6B and ASH1L using > 53,000 compounds that were obtained from the Maybridge library and PubChem Database, followed by molecular docking to evaluate their docking score/Glide Gscore. Based on their docking scores, the selected inhibitors were later assessed for their in vitro anti-leishmanial efficacy. Out of all inhibitors designed against KDM6B and ASH1L, HTS09796, GSK-J4 and AS-99 particularly showed promising in vitro activity with IC50 < 5 µM against both extracellular promastigote and intracellular amastigote forms of L. donovani. In vitro drug interaction studies of these inhibitors further demonstrated their synergistic interaction with amphotericin-B and miltefosine. However, GSK-J4 makes an exception by displaying an in different mode of interaction with miltefosine. Collectively, our in silico and in vitro studies acted as a platform to identify the applicability of these inhibitors targeted against KDM6B and ASH1L for anti-leishmanial 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: Journal Article
    男性不育是一个全球性的健康问题,在高海拔地区尤为普遍。附睾对精子成熟至关重要,但是环境线索对其重塑的影响仍然知之甚少。这里,我们使用单细胞转录组学技术来追踪在常氧或长期缺氧条件下饲养的大鼠表皮细胞的细胞谱。结果表明,缺氧会损害附睾功能,在减少的上皮细胞中很明显,受损的血液附睾屏障完整性,和增加自然杀伤细胞。通过基因调控网络和细胞间相互作用图谱的联合分析,我们鉴定了附睾缺氧敏感细胞,这些细胞通过增加的细胞间粘附分子1(ICAM-1)与自然杀伤(NK)细胞进行通讯,所述细胞间粘附分子1由KLF4将组蛋白甲基转移酶ASL1L募集至Icam1启动子驱动.一起来看,我们的研究提供了缺氧期间附睾变化的详细蓝图,并定义了有助于NK细胞活化的KLF4-ALSH1L-ICAM-1轴,产生针对缺氧诱导的不孕症的潜在治疗方法。
    Male infertility is a global health problem especially prevalent in high-altitude regions. The epididymis is essential for sperm maturation, but the influence of environmental cues on its reshaping remains poorly understood. Here, we use single-cell transcriptomics to track the cellular profiles of epidydimal cells in rats raised under normoxia or extended hypoxia. The results show that hypoxia impairs epididymal function, evident in reduced epithelial cells, compromised blood-epididymis barrier integrity, and increased natural killer cells. Through combined analysis of gene-regulatory networks and cell-cell interaction maps, we identify epididymal hypoxia-sensitive cells that communicate with natural killer (NK) cells via increased intercellular adhesion molecule 1 (ICAM-1) driven by KLF4 recruitment of the histone methyltransferase ASL1L to the Icam1 promoter. Taken together, our study offers a detailed blueprint of epididymal changes during hypoxia and defines a KLF4-ALSH1L-ICAM-1 axis contributing to NK cell activation, yielding a potential treatment targeting hypoxia-induced infertility.
    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
    目的:银屑病是一种常见的自身免疫性皮肤病,显著降低患者的生活质量。体感系统和皮肤免疫系统相互作用的初级传入基本上介导了银屑病的发病机理。本研究旨在阐明初级感觉神经元如何调节银屑病形成的分子机制。
    方法:在初始和IMQ(咪喹莫特)诱导的银屑病模型条件下,从WT和Ash1l+/-(ASH1样组蛋白赖氨酸甲基转移酶)小鼠中提取皮肤和总RNA。免疫组织化学,然后进行qRT-PCR和FACS。微流体室共培养用于研究体外体感感觉神经元与骨髓树突状细胞(BMDC)之间的相互作用。使用全细胞膜片钳记录来评估原发性感觉神经元中Ash1L单倍体不足后的神经元兴奋性。
    结果:ASH1L的单倍体不足,组蛋白甲基转移酶,在初级感觉神经元中,会导致神经突神经支配过度和神经元兴奋性增加,以神经元活性依赖的方式促进miR-let-7b从皮肤初级传入的释放。具有“GUUGUGU”核心序列,miR-let-7b作为Toll样受体7(TLR7)的内源性配体发挥作用,并刺激皮肤DC和白细胞介素(IL)-23/IL-17轴的激活,最终加剧牛皮癣的症状。因此,通过限制miR-let-7b从初级传入的释放,ASH1L可预防皮肤DC活化并改善银屑病。
    结论:体感神经元ASH1L通过限制miR-let-7b的神经元活性依赖性释放来调节皮肤免疫,可通过TLR7直接激活真皮DCs,最终导致银屑病病变加重。
    OBJECTIVE: Psoriasis is a common autoimmune skin disease that significantly diminishes patients\' quality of life. Interactions between primary afferents of the somatosensory system and the cutaneous immune system mediate the pathogenesis of psoriasis. This study aims to elucidate the molecular mechanisms of how primary sensory neurons regulate psoriasis formation.
    METHODS: Skin and total RNA were extracted from wild-type (WT) and ASH1-like histone lysine methyltransferase (Ash1l+/- ) mice in both naive and imiquimod (IMQ)-induced psoriasis models. Immunohistochemistry, quantitative real-time polymerase chain reaction (qRT-PCR) and fluorescence-activated cell sorting (FACS) were then performed. Microfluidic chamber coculture was used to investigate the interaction between somatosensory neurons and bone marrow dendritic cells (BMDCs) ex vivo. Whole-cell patch clamp recordings were used to evaluate neuronal excitability after Ash1L haploinsufficiency in primary sensory neurons.
    RESULTS: The haploinsufficiency of ASH1L, a histone methyltransferase, in primary sensory neurons causes both neurite hyperinnervation and increased neuronal excitability, which promote miR-let-7b release from primary afferents in the skin in a neuronal activity-dependent manner. With a \'GUUGUGU\' core sequence, miR-let-7b functions as an endogenous ligand of toll-like receptor 7 (TLR7) and stimulates the activation of dermal dendritic cells (DCs) and interleukin (IL)-23/IL-17 axis, ultimately exacerbating the symptoms of psoriasis. Thus, by limiting miR-let-7b release from primary afferents, ASH1L prevents dermal DC activation and ameliorates psoriasis.
    CONCLUSIONS: Somatosensory neuron ASH1L modulates the cutaneous immune system by limiting neuronal activity-dependent release of miR-let-7b, which can directly activate dermal DCs via TLR7 and ultimately lead to aggravated psoriatic lesion.
    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
    ASH1L是一种组蛋白甲基转移酶,通过赖氨酸K36上组蛋白H3的甲基化来调节基因表达。虽然ASH1L的催化SET域具有较低的固有活性,一些研究发现,它可以通过与MRG15蛋白的相互作用而大大增强,并提出了释放其自抑制构象的变构机制。这里,我们发现全长MRG15,而不是MRG域,可以增强ASH1LSET域的活性。此外,我们表明MRG15-ASH1L的催化活性取决于MRG15色结构域介导的核小体结合。我们发现在溶液中MRG15与ASH1L结合,但对SET结构域自抑制环或S-腺苷甲硫氨酸辅因子结合位点的构象没有影响。此外,MRG15结合不损害ASH1L的小分子抑制剂的效力。这些发现表明MRG15充当通过募集核小体底物来增强ASH1L催化活性的衔接子。
    ASH1L is a histone methyltransferase that regulates gene expression through methylation of histone H3 on lysine K36. While the catalytic SET domain of ASH1L has low intrinsic activity, several studies found that it can be vastly enhanced by the interaction with MRG15 protein and proposed allosteric mechanism of releasing its autoinhibited conformation. Here, we found that full-length MRG15, but not the MRG domain alone, can enhance the activity of the ASH1L SET domain. In addition, we showed that catalytic activity of MRG15-ASH1L depends on nucleosome binding mediated by MRG15 chromodomain. We found that in solution MRG15 binds to ASH1L, but has no impact on the conformation of the SET domain autoinhibitory loop or the S-adenosylmethionine cofactor binding site. Moreover, MRG15 binding did not impair the potency of small molecule inhibitors of ASH1L. These findings suggest that MRG15 functions as an adapter that enhances ASH1L catalytic activity by recruiting nucleosome substrate.
    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
    甲基转移酶如甲基转移酶3(METTL3),主要的N6-甲基腺苷(m6A)甲基转移酶,已经牵涉到各种生物和病理过程,包括免疫反应。然而,METTL3在致病性T辅助细胞(Th)17中的功能和机制知之甚少。在这里,我们发现实验性自身免疫性葡萄膜炎(EAU)的眼球和T细胞中METTL3表达显着降低,m6A水平降低。METTL3的过表达改善了EAU的发育,并在体内和体外抑制了致病性Th17细胞反应。机械上,METTL3促进缺失的表达,小,或同源样1(ASH1L)通过增强其在含有2(YTHDC2)依赖性方式的YT521-B同源域中的稳定性,进一步降低IL-17和IL-23受体(IL-23R)的表达,导致致病性Th17反应减少。一起,我们的数据揭示了METTL3在调节致病性Th17反应中的关键作用,这可能有助于人类葡萄膜炎的治疗。
    Methyltransferase like 3 (METTL3), a primary N6-methyladenosine (m6A) methyltransferase, has been implicated in various biological and pathological processes including immune responses. However, the functions and mechanisms of METTL3 in pathogenic T helper (Th)17 cells are poorly understood. Here we found significantly decreased METTL3 expression along with reduced m6A levels in eyeballs and T cells of experimental autoimmune uveitis (EAU). Overexpression of METTL3 ameliorated the development of EAU and suppressed pathogenic Th17 cell responses in vivo and in vitro. Mechanistically, METTL3 promoted the expression of absent, small, or homeotic-like 1 (ASH1L) via enhancing its stability in a YT521-B homology domain containing 2 (YTHDC2)-dependent manner, which further decreased the expression of IL-17 and IL-23 receptor (IL-23R), resulting in reduced pathogenic Th17 responses. Together, our data reveal a pivotal role of METTL3 in regulating pathogenic Th17 responses, which may contribute to human uveitis 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: Journal Article
    在雌性哺乳动物中,卵巢内最初建立的原始卵泡池的大小决定了生殖寿命。有趣的是,由于不清楚的原因,原始卵泡池的建立伴随着显着的程序性卵母细胞丢失。这里,我们确定了ASH1样组蛋白赖氨酸甲基转移酶(ASH1L)在小鼠减数分裂前期I期控制卵母细胞凋亡中的新作用。我们的结果表明,Ash1l的过表达通过凋亡导致胎儿卵母细胞的急剧损失,随后导致原始卵泡池的容量降低。Ash1l的过表达也导致与p63和磷酸化检查点激酶2(p-CHK2)的过早上调相关的DNA双链断裂修复缺陷,女性种系的主要基因组监护人,在胎儿卵巢中Ash1l过表达后。总之,ASH1L是不可或缺的表观遗传分子之一,充当基因组的守护者。它通过调节小鼠减数分裂前期I期p63和p-CHK2的表达来保护卵母细胞基因组完整性并去除具有严重DNA损伤的卵母细胞。我们的研究为DNA损伤检查点信号在胎儿卵母细胞监护和女性生育中的生理调节作用提供了视角。
    In female mammals, the size of the initially established primordial follicle pool within the ovaries determines the reproductive life span. Interestingly, the establishment of the primordial follicle pool is accompanied by a remarkable programmed oocyte loss for unclear reasons. Here, we identify a new role of ASH1-like histone lysine methyltransferase (ASH1L) in controlling the apoptosis of oocytes during meiotic prophase I in mice. Our results showed that overexpression of Ash1l led to a dramatic loss of fetal oocytes via apoptosis, which subsequently resulted in a reduced capacity of the primordial follicle pool. Overexpression of Ash1l also led to a deficiency in DNA double-strand break repair associated with premature upregulation of p63 and phosphorylated checkpoint kinase 2 (p-CHK2), the major genome guardian of the female germline, following Ash1l overexpression in fetal ovaries. In summary, ASH1L is one of the indispensable epigenetic molecules that acts as a guardian of the genome. It protects oocyte genome integrity and removes oocytes with serious DNA damage by regulating the expression of p63 and p-CHK2 during meiotic prophase I in mice. Our study provides a perspective on the physiological regulatory role of DNA damage checkpoint signaling in fetal oocyte guardianship and female fertility.
    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
    ASH1L是Trithorax组蛋白的成员,可作为组蛋白甲基转移酶用于基因转录激活。已知ASH1L在其基因靶标处调节H3K4me3和H3K36me2/3,但其具体的组蛋白识别机制尚不清楚。在这项研究中,我们发现ASH1L植物同源域(PHD)手指与单相互作用,di-,和具有相当亲和力的H3K4肽的三甲基化状态,表明ASH1LPHD非选择性地结合H3K4的所有三种甲基化状态。我们解决了核磁共振结构,描绘了ASH1LPHD指与二甲基化H3K4肽的结合,并发现甲基化赖氨酸结合袋中的狭窄结合沟和残基组成限制了与K4的二甲基铵部分的必要相互作用。此外,我们发现,与PCaLNCaP细胞相比,ASH1L蛋白在去势耐药前列腺癌(PCa)PC3和DU145细胞中过表达.ASH1L的敲低调节了参与细胞凋亡和细胞周期调控的基因表达和细胞通路,从而诱导了细胞周期停滞,细胞凋亡,PC3和DU145细胞的集落形成能力降低。ASH1L的C末端核心而不是PHD缺失突变体的过表达增加了整体H3K36me2水平,但对H3K4me2/3水平没有影响。总的来说,我们的研究将ASH1LPHD手指确定为第一个非选择性识别H3K4三种甲基化状态的天然阅读器。此外,ASH1L是PCas中细胞周期失调和存活所必需的。
    ASH1L is a member of the Trithorax-group protein and acts as a histone methyltransferase for gene transcription activation. It is known that ASH1L modulates H3K4me3 and H3K36me2/3 at its gene targets, but its specific mechanism of histone recognition is insufficiently understood. In this study, we found that the ASH1L plant homeodomain (PHD) finger interacts with mono-, di-, and trimethylated states of H3K4 peptides with comparable affinities, indicating that ASH1L PHD non-selectively binds to all three methylation states of H3K4. We solved nuclear magnetic resonance structures picturing the ASH1L PHD finger binding to the dimethylated H3K4 peptide and found that a narrow binding groove and residue composition in the methylated-lysine binding pocket restricts the necessary interaction with the dimethyl-ammonium moiety of K4. In addition, we found that the ASH1L protein is overexpressed in castrate-resistant prostate cancer (PCa) PC3 and DU145 cells in comparison to PCa LNCaP cells. The knockdown of ASH1L modulated gene expression and cellular pathways involved in apoptosis and cell cycle regulation and consequently induced cell cycle arrest, cell apoptosis, and reduced colony-forming abilities in PC3 and DU145 cells. The overexpression of the C-terminal core of ASH1L but not the PHD deletion mutant increased the overall H3K36me2 level but had no effect on the H3K4me2/3 level. Overall, our study identifies the ASH1L PHD finger as the first native reader that non-selectively recognizes the three methylation states of H3K4. Additionally, ASH1L is required for the deregulation of cell cycle and survival in PCas.
    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在赖氨酸36(H3K36me)的甲基化对于维持基因组稳定性至关重要。的确,这个甲基化标记对于正确的转录是必不可少的,重组,和DNA损伤反应。H3K36甲基转移酶中的功能缺失和获得突变与人类发育障碍和各种癌症密切相关。结构分析表明,核小体成分,如接头DNA和由组蛋白H2A和H3构成的疏水斑块,除了组蛋白H3尾之外,可能是H3K36甲基化的决定因素。其包括H3K36和催化SET结构域。H3K36甲基转移酶与核小体的相互作用与其自抑制性变化的调节相配合,微调了H3K36me介导NSD2和NSD3的二甲基化以及Set2/SETD2的三甲基化的精确度。鉴定与不同形式的H3K36me结合的特定结构特征和各种顺式作用因子,特别是H3K36的二(H3K36me2)和三(H3K36me3)甲基化形式,突出了H3K36me功能意义的复杂性。这里,我们巩固了这些发现,并为H3K36me2向H3K36me3转化的调节提供了结构见解。我们还讨论了H3K36me与其他染色质修饰之间合作的机制(特别是,H3K27me3,H3乙酰化,RNA中的DNA甲基化和N6-甲基腺苷)在染色质表观基因组功能的生理调节中。
    The methylation of histone H3 at lysine 36 (H3K36me) is essential for maintaining genomic stability. Indeed, this methylation mark is essential for proper transcription, recombination, and DNA damage response. Loss- and gain-of-function mutations in H3K36 methyltransferases are closely linked to human developmental disorders and various cancers. Structural analyses suggest that nucleosomal components such as the linker DNA and a hydrophobic patch constituted by histone H2A and H3 are likely determinants of H3K36 methylation in addition to the histone H3 tail, which encompasses H3K36 and the catalytic SET domain. Interaction of H3K36 methyltransferases with the nucleosome collaborates with regulation of their auto-inhibitory changes fine-tunes the precision of H3K36me in mediating dimethylation by NSD2 and NSD3 as well as trimethylation by Set2/SETD2. The identification of specific structural features and various cis-acting factors that bind to different forms of H3K36me, particularly the di-(H3K36me2) and tri-(H3K36me3) methylated forms of H3K36, have highlighted the intricacy of H3K36me functional significance. Here, we consolidate these findings and offer structural insight to the regulation of H3K36me2 to H3K36me3 conversion. We also discuss the mechanisms that underlie the cooperation between H3K36me and other chromatin modifications (in particular, H3K27me3, H3 acetylation, DNA methylation and N6-methyladenosine in RNAs) in the physiological regulation of the epigenomic functions of chromatin.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号