HDACs

HDAC
  • 文章类型: Journal Article
    背景:物理,化学,环境中的生物因素不断影响体内和体外的生物过程,包括参与癌症和代谢的多种组蛋白修饰。然而,乙酰化调节的复杂机制仍未阐明。在哺乳动物精子发生中,乙酰化在修复双链DNA断裂中起着至关重要的作用,调节基因转录,和调节各种信号通路。
    结果:这篇综述总结了小鼠睾丸中的组蛋白乙酰化位点,并全面概述了组蛋白乙酰化如何在组蛋白脱乙酰酶的调节下参与精子发生的不同阶段。强调了精子发生过程中各类组蛋白脱乙酰酶的调节功能以及组蛋白乙酰化与其他组蛋白修饰之间的十字路口。了解组蛋白去乙酰化酶调控的组蛋白乙酰化在精子发生中的作用机制,这有助于预防和治疗不孕症相关疾病。
    BACKGROUND: Physical, chemical, and biological factors in the environment constantly influence in vivo and in vitro biological processes, including diverse histone modifications involved in cancer and metabolism. However, the intricate mechanisms of acetylation regulation remain poorly elucidated. In mammalian spermatogenesis, acetylation plays a crucial role in repairing double-strand DNA breaks, regulating gene transcription, and modulating various signaling pathways.
    RESULTS: This review summarizes the histone acetylation sites in the mouse testis and provides a comprehensive overview of how histone acetylation is involved in different stages of spermatogenesis under the regulation by histone deacetylases. The regulatory functions of various class histone deacetylases during spermatogenesis and the crossroad between histone acetylation and other histone modifications are highlighted. It is imperative to understand the mechanisms of histone acetylation regulated by histone deacetylases in spermatogenesis, which facilitates to prevent and treat infertility-related diseases.
    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
    ADORA3主要在肠道表达,并具有促进粘蛋白2(MUC2)表达的潜力,杯状细胞的功能相关因子,在哮喘情况下。本研究旨在证实ADORA3对溃疡性结肠炎(UC)杯状细胞的诱导作用及其机制。在UC患者的粘膜活检和结肠炎小鼠的结肠中发现ADORA3表达显着降低。这种减少与疾病严重程度呈负相关,与杯状细胞数量呈正相关。ADORA3激活减轻葡聚糖硫酸钠(DSS)诱导的结肠炎,并在体内和体外促进ATOH1介导的杯状细胞分化。代谢组学分析揭示了ADORA3激活支持酮生成,导致代谢产物BHB水平升高。随后,BHB增强了HDAC1/2的活性,从而增强了ATOH1基因启动子区域内H3K9ac位点的组蛋白乙酰化。此外,ADORA3激活以增强酮生成的原因归因于控制β-arrestin2,SHP1和PPARγ之间的竞争性结合。这导致PPARγ的非配体依赖性活化,从而促进HMGCS2的转录。使用MRS1191和shHMGCS2质粒阐明了ADORA3促进杯状细胞分化和减轻UC的确切机制。总的来说,ADORA3激活通过“BHB-HDAC1/2-H3K9ac”途径增强酮生成,促进杯状细胞分化并减轻UC。
    ADORA3 is mainly expressed in intestinal tract, and has the potential to promote the expression of mucin 2 (MUC2), the function-related factor of goblet cells, under asthma conditions. This study aims to confirm the induction and mechanisms of ADORA3 activation on goblet cells in ulcerative colitis (UC). A significant decrease in ADORA3 expression was found in mucosal biopsies from UC patients and in the colons of colitis mice. This reduction correlated negatively with disease severity and positively with goblet cell number. ADORA3 activation mitigated dextran sulfate sodium (DSS)-induced colitis and facilitated ATOH1-mediated goblet cell differentiation in both in vivo and in vitro. Metabolomics analysis unveiled that ADORA3 activation bolstered ketogenesis, leading to elevated levels of the metabolite BHB. Subsequently, BHB heightened the activity of HDAC1/2, augmenting histone acetylation at the H3K9ac site within the promoter region of the ATOH1 gene. Furthermore, the reason for ADORA3 activation to enhance ketogenesis was attributed to controlling the competitive binding among β-arrestin2, SHP1 and PPARγ. This results in the non-ligand-dependent activation of PPARγ, thereby promoting the transcription of HMGCS2. The exact mechanisms by which ADORA3 promoted goblet cell differentiation and alleviated UC were elucidated using MRS1191 and shHMGCS2 plasmid. Collectively, ADORA3 activation promoted goblet cell differentiation and alleviated UC by enhancing ketogenesis via the \"BHB-HDAC1/2-H3K9ac\" pathway.
    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
    核因子红系2相关因子2(Nrf2)的表观遗传调控,一个关键的氧化还原转录因子,在维持细胞稳态中起着至关重要的作用。最近的研究强调了Nrf2的表观遗传修饰在糖尿病足溃疡(DFU)发病机理中的重要性。这项研究调查了在高血糖微环境(HGM)中人类内皮细胞中,蝶芪(PTS)对Nrf2的表观遗传逆转。通过ARE-荧光素酶报告基因测定和核易位研究评估了PTS对Nrf2的激活潜力。暴露于HGM72小时后,Nrf2及其下游靶NAD(P)H醌氧化还原酶1(NQO1)的mRNA表达和蛋白水平,血红素加氧酶1(HO-1),超氧化物歧化酶(SOD),过氧化氢酶(CAT)表现出下降,在PTS预处理的内皮细胞中得到缓解。表观遗传标记,包括组蛋白脱乙酰酶(HDACsI-IV类)和DNA甲基转移酶(DNMTs1/3A和3B),被发现在糖尿病条件下下调。具体来说,Nrf2关联的HDAC,HDAC1、HDAC2、HDAC3和HDAC4在HGM诱导的内皮细胞中上调。这种上调在PTS预处理的细胞中被逆转,除了HDAC2,其在高血糖微环境中的PTS处理的内皮细胞中表现出升高的表达。此外,观察到PTS逆转甲基转移酶DNMT的活性。此外,Nrf2启动子中的CpG岛在暴露于HGM的细胞中高度甲基化,PTS预处理可能抵消的现象,如甲基敏感限制性内切酶PCR(MSRE-qPCR)分析所示。总的来说,我们的发现强调了PTS在高血糖条件下表观遗传调节Nrf2表达的能力,提示其治疗糖尿病并发症的潜力。
    The epigenetic regulation of nuclear factor erythroid 2-related factor 2 (Nrf2), a pivotal redox transcription factor, plays a crucial role in maintaining cellular homeostasis. Recent research has underscored the significance of epigenetic modifications of Nrf2 in the pathogenesis of diabetic foot ulcers (DFUs). This study investigates the epigenetic reversal of Nrf2 by pterostilbene (PTS) in human endothelial cells in a hyperglycemic microenvironment (HGM). The activation potential of PTS on Nrf2 was evaluated through ARE-Luciferase reporter assays and nuclear translocation studies. Following 72 h of exposure to an HGM, mRNA expression and protein levels of Nrf2 and its downstream targets NAD(P)H quinone oxidoreductase 1 (NQO1), heme-oxygenase 1(HO-1), superoxide dismutase (SOD), and catalase (CAT) exhibited a decrease, which was mitigated in PTS-pretreated endothelial cells. Epigenetic markers, including histone deacetylases (HDACs class I-IV) and DNA methyltransferases (DNMTs 1/3A and 3B), were found to be downregulated under diabetic conditions. Specifically, Nrf2-associated HDACs, including HDAC1, HDAC2, HDAC3, and HDAC4, were upregulated in HGM-induced endothelial cells. This upregulation was reversed in PTS-pretreated cells, except for HDAC2, which exhibited elevated expression in endothelial cells treated with PTS in a hyperglycemic microenvironment. Additionally, PTS was observed to reverse the activity of the methyltransferase enzyme DNMT. Furthermore, CpG islands in the Nrf2 promoter were hypermethylated in cells exposed to an HGM, a phenomenon potentially counteracted by PTS pretreatment, as shown by methyl-sensitive restriction enzyme PCR (MSRE-qPCR) analysis. Collectively, our findings highlight the ability of PTS to epigenetically regulate Nrf2 expression under hyperglycemic conditions, suggesting its therapeutic potential in managing diabetic complications.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Editorial
    暂无摘要。
    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
    在给定的蛋白质内的特定残基的化学选择性修饰提出了重大的挑战。因为蛋白质中氨基酸残基的微环境是可变的。开发具有可调化学弹头的通用分子平台可以为精确标记蛋白质中的特定氨基酸提供强大的工具。半胱氨酸和赖氨酸是化学选择性修饰的热门靶标,但是由于交叉反应性和不稳定的反应产物,目前的半胱氨酸/赖氨酸选择性弹头面临挑战。在这项研究中,开发了一种通用的荧光平台,用于在生物相容性条件下对半胱氨酸/赖氨酸进行高选择性修饰。氯-或苯氧基-取代的NBSe衍生物以高特异性有效地标记细胞蛋白质组中的半胱氨酸残基。这一发现还导致了苯氧基-NBSe光热不可知的发展,用于GSH过表达的癌细胞的诊断和可激活的光动力疗法。相反,烷氧基-NBSe衍生物被设计为在细胞环境中选择性地与赖氨酸残基反应,对硫醇具有优异的抗干扰能力。利用接近驱动的方法,成功地设计了烷氧基-NBSe探针以证明其在赖氨酸脱乙酰酶活性的生物成像中的实用性。这项研究还实现了以区域选择性方式将小的光敏剂整合到蛋白质的赖氨酸残基中,实现由过度表达的蛋白质激活的癌细胞的光消融。
    Chemoselective modification of specific residues within a given protein poses a significant challenge, as the microenvironment of amino acid residues in proteins is variable. Developing a universal molecular platform with tunable chemical warheads can provide powerful tools for precisely labeling specific amino acids in proteins. Cysteine and lysine are hot targets for chemoselective modification, but current cysteine/lysine-selective warheads face challenges due to cross-reactivity and unstable reaction products. In this study, a versatile fluorescent platform is developed for highly selective modification of cysteine/lysine under biocompatible conditions. Chloro- or phenoxy-substituted NBSe derivatives effectively labeled cysteine residues in the cellular proteome with high specificity. This finding also led to the development of phenoxy-NBSe phototheragnostic for the diagnosis and activatable photodynamic therapy of GSH-overexpressed cancer cells. Conversely, alkoxy-NBSe derivatives are engineered to selectively react with lysine residues in the cellular environment, exhibiting excellent anti-interfering ability against thiols. Leveraging a proximity-driven approach, alkoxy-NBSe probes are successfully designed to demonstrate their utility in bioimaging of lysine deacetylase activity. This study also achieves integrating a small photosensitizer into lysine residues of proteins in a regioselective manner, achieving photoablation of cancer cells activated by overexpressed proteins.
    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
    组蛋白脱乙酰酶(HDAC)是一类锌依赖性酶。它们维持乙酰化稳态,具有许多生物学功能,并与许多疾病有关。HDAC3严格需要多亚基复合物形成以获得活性。它与许多非传染性疾病的进展有关。它在疾病中的广泛参与使其成为表观遗传药物靶标。先前存在的HDAC3抑制剂有许多用途,强调需要继续研究发现HDAC3选择性抑制剂。
    这篇综述概述了2010-2023年发布的24项专利,重点是抑制HDAC3同工酶的化合物。
    HDAC3选择性抑制剂-作为单一或联合疗法的药物应用至关重要-作为摆脱充满并发症的泛HDAC抑制剂的策略,正在获得牵引力。此外,对于具有替代锌结合基团(ZBG)的HDAC3抑制剂存在未满足的需求,因为一些先前存在的ZBG具有与毒性和副作用相关的局限性.实现HDAC3选择性的困难可能是由于同种型选择性。然而,计算机辅助药物设计和HDAC33D共结晶模型的实验数据的进步可能导致发现新型HDAC3选择性抑制剂。具有对HDAC3的选择性和效力平衡的替代ZBG。
    UNASSIGNED: Histone deacetylases (HDACs) are a class of zinc-dependent enzymes. They maintain acetylation homeostasis, with numerous biological functions and are associated with many diseases. HDAC3 strictly requires multi-subunit complex formation for activity. It is associated with the progression of numerous non-communicable diseases. Its widespread involvement in diseases makes it an epigenetic drug target. Preexisting HDAC3 inhibitors have many uses, highlighting the need for continued research in the discovery of HDAC3-selective inhibitors.
    UNASSIGNED: This review provides an overview of 24 patents published from 2010 to 2023, focusing on compounds that inhibit the HDAC3 isoenzyme.
    UNASSIGNED: HDAC3-selective inhibitors - pivotal for pharmacological applications, as single or combination therapies - are gaining traction as a strategy to move away from complications laden pan-HDAC inhibitors. Moreover, there is an unmet need for HDAC3 inhibitors with alternative zinc-binding groups (ZBGs) because some preexisting ZBGs have limitations related to toxicity and side effects. Difficulties in achieving HDAC3 selectivity may be due to isoform selectivity. However, advancements in computer-aided drug design and experimental data of HDAC3 3D co-crystallized models could lead to the discovery of novel HDAC3-selective inhibitors, which bear alternative ZBGs with balanced selectivity for HDAC3 and potency.
    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
    糖尿病是一种损害新陈代谢的慢性疾病,其患病率已达到全球流行比例。大多数受影响的人患有2型糖尿病(T2DM),这是由胰腺内分泌胰岛细胞的数量或功能下降引起的,特别是β-细胞释放足够量的胰岛素以克服代谢组织的任何胰岛素抗性。遗传和表观遗传因素是T2DM的主要贡献者。表观遗传修饰剂,组蛋白脱乙酰酶(HDAC),是从组蛋白中去除乙酰基的酶,在各种分子过程中发挥重要作用,包括胰腺细胞的命运,胰岛素释放,胰岛素生产,胰岛素信号,和葡萄糖代谢。HDAC还管理与糖尿病相关的其他监管过程,如氧化应激,炎症,凋亡,和纤维化,通过网络和功能分析揭示。这篇综述解释了目前对HDAC在糖尿病病理生理学中的功能的理解。各种HDAC抑制剂(HDACi)的抑制作用,以及它们作为T2DM生物标志物和可能的治疗靶点的功能重要性。虽然它们在T2DM中的作用仍在显现,更好地了解HDACi的作用可能与改善胰岛素敏感性有关,保护β细胞并减少T2DM相关并发症,在其他人中。
    Diabetes mellitus is a chronic disease that impairs metabolism, and its prevalence has reached an epidemic proportion globally. Most people affected are with type 2 diabetes mellitus (T2DM), which is caused by a decline in the numbers or functioning of pancreatic endocrine islet cells, specifically the β-cells that release insulin in sufficient quantity to overcome any insulin resistance of the metabolic tissues. Genetic and epigenetic factors have been implicated as the main contributors to the T2DM. Epigenetic modifiers, histone deacetylases (HDACs), are enzymes that remove acetyl groups from histones and play an important role in a variety of molecular processes, including pancreatic cell destiny, insulin release, insulin production, insulin signalling, and glucose metabolism. HDACs also govern other regulatory processes related to diabetes, such as oxidative stress, inflammation, apoptosis, and fibrosis, revealed by network and functional analysis. This review explains the current understanding of the function of HDACs in diabetic pathophysiology, the inhibitory role of various HDAC inhibitors (HDACi), and their functional importance as biomarkers and possible therapeutic targets for T2DM. While their role in T2DM is still emerging, a better understanding of the role of HDACi may be relevant in improving insulin sensitivity, protecting β-cells and reducing T2DM-associated complications, among others.
    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
    赖氨酸去乙酰化酶抑制剂(KDACis)是批准用于皮肤T细胞淋巴瘤(CTCL)的药物,外周T细胞淋巴瘤(PTCL),和多发性骨髓瘤,但其细胞作用机制(MoA)和实质性毒性的许多方面尚不清楚。为了进一步阐明KDACis如何引发细胞反应,我们系统地测量了乙酰化的剂量依赖性变化,磷酸化,和响应21种临床和临床前KDACis的蛋白质表达。得到的862,000个剂量反应曲线显示,例如,组蛋白去乙酰化酶(HDAC)1,2,3和6抑制剂的细胞特异性有限;乙酰化和磷酸化途径之间的强交叉;大多数药物反应乙酰化位点定位于内在无序区域(IDR);乙酰化在蛋白质结构中的作用被低估;EP300蛋白质丰度在细胞质和细胞核之间的变化.这个全面的数据集可以作为研究细胞中KDACi作用的分子机制的资源,可以在ProteomicsDB中进行在线交互探索。
    Lysine deacetylase inhibitors (KDACis) are approved drugs for cutaneous T cell lymphoma (CTCL), peripheral T cell lymphoma (PTCL), and multiple myeloma, but many aspects of their cellular mechanism of action (MoA) and substantial toxicity are not well understood. To shed more light on how KDACis elicit cellular responses, we systematically measured dose-dependent changes in acetylation, phosphorylation, and protein expression in response to 21 clinical and pre-clinical KDACis. The resulting 862,000 dose-response curves revealed, for instance, limited cellular specificity of histone deacetylase (HDAC) 1, 2, 3, and 6 inhibitors; strong cross-talk between acetylation and phosphorylation pathways; localization of most drug-responsive acetylation sites to intrinsically disordered regions (IDRs); an underappreciated role of acetylation in protein structure; and a shift in EP300 protein abundance between the cytoplasm and the nucleus. This comprehensive dataset serves as a resource for the investigation of the molecular mechanisms underlying KDACi action in cells and can be interactively explored online in ProteomicsDB.
    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
    中性粒细胞弹性蛋白酶(NE)被巨噬细胞吸收,保留细胞内蛋白酶活性,并诱导促炎表型。然而,NE诱导的巨噬细胞促炎极化的机制尚不清楚.我们假设细胞内NE降解组蛋白脱乙酰酶(HDAC)和Sirtuins,破坏赖氨酸乙酰化和去乙酰化的平衡,导致主要alarmin的细胞核向细胞质易位,高流动性组方框1(HMGB1),巨噬细胞的促炎反应。从健康供体或患有囊性纤维化(CF)或慢性阻塞性肺病(COPD)的受试者获得人血单核细胞。单核细胞在体外分化成血液单核细胞衍生的巨噬细胞(BMDMs)。人类BMDM暴露于NE或对照载体,HDAC和Sirtuins的丰度通过总细胞裂解物或核提取物的Western印迹或通过ELISA测定。HDAC,Sirtuin,测定组蛋白乙酰转移酶(HAT)活性。NE降解大多数HDAC和Sirtuin(Sirt)1,导致HDAC和Sirtuin活性降低,HAT活性变化最小。然后我们评估了NE诱导的Sirt活性丧失或HDAC活性丧失是否会改变HMGB1的细胞定位。NE治疗或用曲古抑菌素A(TSA)治疗,全球HDAC抑制剂,两者都增加了HMGB1从细胞核到细胞质的易位,与HMGB1激活一致。NE显着降解I类和II类HDAC家族成员和Sirt1,使BMDM转变为促炎表型。
    Neutrophil elastase (NE) is taken up by macrophages, retains intracellular protease activity, and induces a pro-inflammatory phenotype. However, the mechanism of NE-induced pro-inflammatory polarization of macrophages is not well understood. We hypothesized that intracellular NE degrades histone deacetylases (HDAC) and Sirtuins, disrupting the balance of lysine acetylation and deacetylation and resulting in nuclear to cytoplasmic translocation of a major alarmin, High Mobility Group Box 1 (HMGB1), a pro-inflammatory response in macrophages. Human blood monocytes were obtained from healthy donors or from subjects with cystic fibrosis (CF) or chronic obstructive pulmonary disease (COPD). Monocytes were differentiated into blood monocyte derived macrophages (BMDMs) in vitro. Human BMDMs were exposed to NE or control vehicle, and the abundance of HDACs and Sirtuins was determined by Western blotting of total cell lysates or nuclear extracts or determined by ELISA. HDAC, Sirtuin, and Histone acetyltransferase (HAT) activities were measured. NE degraded most HDACs and Sirtuin (Sirt)1, resulting in decreased HDAC and sirtuin activities, with minimal change in HAT activity. We then evaluated whether the NE-induced loss of Sirt activity or loss of HDAC activities would alter the cellular localization of HMGB1. NE treatment or treatment with Trichostatin A (TSA), a global HDAC inhibitor, both increased HMGB1 translocation from the nucleus to the cytoplasm, consistent with HMGB1 activation. NE significantly degraded Class I and II HDAC family members and Sirt 1, which shifted BMDMs to a pro-inflammatory phenotype.
    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
    三阴性乳腺癌(TNBC)是一种转移性疾病,是一项艰巨的治疗挑战,因为它对现有疗法没有反应。表观遗传调节因子通过调节抗凋亡细胞的表达在进展和转移中起关键作用,TNBC细胞中的促凋亡标志物和相关miRNA。我们已经研究了膳食类黄酮“Apigenin”及其与伏立诺他的组合对MDA-MB-231细胞的抗TNBC潜力。在芹菜素中产生ROS,抑制细胞迁移,细胞周期阻滞在subG0/G1期,并诱导凋亡介导的细胞死亡。芹菜素在转录组和蛋白质组水平上降低I类HDAC的表达。在免疫印迹研究中,芹菜素具有上调的促凋亡标志物和下调的抗凋亡蛋白。芹菜素抑制HDAC/DNMT的酶活性并增加HAT活性。芹菜素通过上调肿瘤抑制因子miR-200b和下调comiR-21表现出对miRNA表达的影响。组合研究通过调节表观遗传和凋亡调节因子的表达协同地降低了TNBC细胞的生长。分子对接和MD模拟探索了HDAC1和HDAC3的催化抑制机制,并支持了体外研究。总体研究证明了芹菜素的抗TNBC潜力,可能有助于设计治疗TNBC转移性表型的有效策略。
    Triple-negative breast cancer (TNBC) is a metastatic disease and a formidable treatment challenge as it does not respond to existing therapies. Epigenetic regulators play a crucial role in the progression and metastasis by modulating the expression of anti-apoptotic, pro-apoptotic markers and related miRNAs in TNBC cells. We have investigated the anti-TNBC potential of dietary flavonoid \'Apigenin\' and its combination with Vorinostat on MDA-MB-231 cells. At Apigenin generated ROS, inhibited cell migration, arrested the cell cycle at subG0/G1 phases, and induced apoptotic-mediated cell death. Apigenin reduced the expression of the class-I HDACs at the transcriptomic and proteomic levels. In the immunoblotting study, Apigenin has upregulated pro-apoptotic markers and downregulated anti-apoptotic proteins. Apigenin inhibited the enzymatic activity of HDAC/DNMT and increased HAT activity. Apigenin has manifested its effect on miRNA expression by upregulating the tumor-suppressor miR-200b and downregulation oncomiR-21. Combination study reduced the growth of TNBC cells synergistically by modulating the expression of epigenetic and apoptotic regulators. Molecular docking and MD simulations explored the mechanism of catalytic inhibition of HDAC1 and HDAC3 and supported the in-vitro studies. The overall studies demonstrated an anti-TNBC potential of Apigenin and may help to design an effective strategy to treat metastatic phenotype of TNBC.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号