Histone deacetylases

组蛋白脱乙酰酶
  • 文章类型: Journal Article
    活细胞在复杂的机械线索中导航,这些线索影响着它们的行为和命运,来自内部和外部来源。在分子水平上,这些物理刺激转化为细胞反应依赖于机械传感器和传感器的复杂协调,最终影响染色质压实和基因表达。值得注意的是,组蛋白尾部的表观遗传修饰控制着基因调控位点的可及性,从而调节基因表达。在这些修改中,组蛋白乙酰化对机械微环境特别敏感,对细胞活动施加显著控制。然而,由于乙酰化网络的复杂性,组蛋白乙酰化在机械传感和转导中的确切作用仍然难以捉摸。为了解决这个差距,我们的目的是系统地探索组蛋白乙酰化的关键调节因子及其在响应生物力学刺激中的多方面作用。在这次审查中,我们最初引入细胞经历的无处不在的力,然后探索组蛋白乙酰化及其相关辅因子的动态变化,包括HDAC,帽子,和乙酰辅酶A,响应这些生物力学线索。此外,我们深入研究了组蛋白乙酰化和机械传感器/机械传感器之间的复杂相互作用,提供全面的分析。最终,本综述旨在在学术框架内全面了解组蛋白乙酰化与机械力之间的细微差别.
    Living cells navigate a complex landscape of mechanical cues that influence their behavior and fate, originating from both internal and external sources. At the molecular level, the translation of these physical stimuli into cellular responses relies on the intricate coordination of mechanosensors and transducers, ultimately impacting chromatin compaction and gene expression. Notably, epigenetic modifications on histone tails govern the accessibility of gene-regulatory sites, thereby regulating gene expression. Among these modifications, histone acetylation emerges as particularly responsive to the mechanical microenvironment, exerting significant control over cellular activities. However, the precise role of histone acetylation in mechanosensing and transduction remains elusive due to the complexity of the acetylation network. To address this gap, our aim is to systematically explore the key regulators of histone acetylation and their multifaceted roles in response to biomechanical stimuli. In this review, we initially introduce the ubiquitous force experienced by cells and then explore the dynamic alterations in histone acetylation and its associated co-factors, including HDACs, HATs, and acetyl-CoA, in response to these biomechanical cues. Furthermore, we delve into the intricate interactions between histone acetylation and mechanosensors/mechanotransducers, offering a comprehensive analysis. Ultimately, this review aims to provide a holistic understanding of the nuanced interplay between histone acetylation and mechanical forces within an academic framework.
    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
    I类组蛋白脱乙酰酶(HDAC)与多种疾病的发展密切相关。包括癌症,神经退行性疾病,艾滋病毒,和炎症性疾病。考虑到在肿瘤发生中的重要作用,I类HDAC已成为治疗策略的高度理想目标,特别是在抗癌药物开发领域。然而,传统的I类HDAC抑制剂面临着一些挑战,如获得性抗性,固有的毒性,以及抑制HDAC非酶功能的功效有限。为了解决这些问题,新的策略已经出现,包括I类HDAC双作用抑制剂的开发,靶向蛋白质降解(TPD)技术,如PROTACs,分子胶,和HyT降解器,以及共价抑制剂。这篇综述提供了I类HDAC酶和抑制剂的全面概述,通过最初介绍它们的结构和生物学作用。随后,我们专注于I类HDAC调制器的最新进展,包括同工型选择性I类抑制剂,双靶点抑制剂,TPDs,和共价抑制剂,从合理设计原则的角度来看,药效学,药代动力学,和临床进展。最后,我们还提供了挑战,并概述了I类HDAC靶向癌症治疗药物发现领域的未来前景。
    Class I histone deacetylases (HDACs) are closely associated with the development of a diverse array of diseases, including cancer, neurodegenerative disorders, HIV, and inflammatory diseases. Considering the essential roles in tumorigenesis, class I HDACs have emerged as highly desirable targets for therapeutic strategies, particularly in the field of anticancer drug development. However, the conventional class I HDAC inhibitors faced several challenges such as acquired resistance, inherent toxicities, and limited efficacy in inhibiting non-enzymatic functions of HDAC. To address these problems, novel strategies have emerged, including the development of class I HDAC dual-acting inhibitors, targeted protein degradation (TPD) technologies such as PROTACs, molecular glues, and HyT degraders, as well as covalent inhibitors. This review provides a comprehensive overview of class I HDAC enzymes and inhibitors, by initially introducing their structure and biological roles. Subsequently, we focus on the recent advancements of class I HDAC modulators, including isoform-selective class I inhibitors, dual-target inhibitors, TPDs, and covalent inhibitors, from the perspectives of rational design principles, pharmacodynamics, pharmacokinetics, and clinical progress. Finally, we also provide the challenges and outlines future prospects in the realm of class I HDAC-targeted drug discovery for cancer therapeutics.
    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
    暴露于颗粒物(PM)可引起气道炎症并使各种气道疾病恶化。然而,PM引发气道炎症的潜在分子机制尚未完全阐明,缺乏有效的干预措施。我们的研究表明,PM暴露通过METTL3/m6A甲基化/IGF2BP3途径增加了人支气管上皮细胞和小鼠气道上皮中组蛋白脱乙酰酶9(HDAC9)的表达。功能实验表明,HDAC9上调可促进PM诱导的气道炎症和MAPK信号通路的激活。机械上,HDAC9调控双特异性磷酸酶9(DUSP9)启动子区K12(H4K12)组蛋白4乙酰化去乙酰化抑制DUSP9的表达,激活MAPK信号通路,从而促进PM诱导的气道炎症。此外,HDAC9与MEF2A结合以削弱其对PM诱导的气道炎症的抗炎作用。然后,我们开发了一种新型的吸入脂质纳米颗粒系统,用于将HDAC9siRNA递送到气道,为PM引起的气道炎症提供有效的治疗。总的来说,我们阐明了HDAC9在PM诱导的气道炎症中的关键调节机制,并介绍了一种针对HDAC9的吸入治疗方法.这些发现有助于减轻由PM暴露引起的各种气道疾病的负担。
    Exposure to particulate matter (PM) can cause airway inflammation and worsen various airway diseases. However, the underlying molecular mechanism by which PM triggers airway inflammation has not been completely elucidated, and effective interventions are lacking. Our study revealed that PM exposure increased the expression of histone deacetylase 9 (HDAC9) in human bronchial epithelial cells and mouse airway epithelium through the METTL3/m6A methylation/IGF2BP3 pathway. Functional assays showed that HDAC9 upregulation promoted PM-induced airway inflammation and activation of MAPK signaling pathway in vitro and in vivo. Mechanistically, HDAC9 modulated the deacetylation of histone 4 acetylation at K12 (H4K12) in the promoter region of dual specificity phosphatase 9 (DUSP9) to repress the expression of DUSP9 and resulting in the activation of MAPK signaling pathway, thereby promoting PM-induced airway inflammation. Additionally, HDAC9 bound to MEF2A to weaken its anti-inflammatory effect on PM-induced airway inflammation. Then, we developed a novel inhaled lipid nanoparticle system for delivering HDAC9 siRNA to the airway, offering an effective treatment for PM-induced airway inflammation. Collectively, we elucidated the crucial regulatory mechanism of HDAC9 in PM-induced airway inflammation and introduced an inhaled therapeutic approach targeting HDAC9. These findings contribute to alleviating the burden of various airway diseases caused by PM exposure.
    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
    III类组蛋白脱乙酰酶SIRT1是研究最广泛的去乙酰化酶。它对广泛的脱乙酰酶抑制剂曲古抑菌素A具有抗性,并且依赖于氧化的烟酰胺腺嘌呤核苷酸(NAD)。SIRT1在许多类型癌症的肿瘤发生中起着至关重要的作用。包括结直肠癌(CRC)。越来越多的证据表明SIRT1是CRC的治疗靶点;然而,SIRT1在CRC中的功能和潜在机制仍有待阐明。在这里,我们提供了一个详细的和更新的审查,以说明SIRT1调节许多在CRC细胞中出错的过程,如细胞凋亡,自噬,扩散,迁移,入侵,转移,氧化应激,对化学放射疗法的抗性,免疫逃避,和代谢重编程。此外,我们将我们的审查与CRC治疗的临床实践紧密联系起来,总结SIRT1抑制剂在CRC治疗中的作用机制及前景。SIRT1抑制剂作为CRC的单一疗法或与化疗联合使用,放射治疗,和免疫疗法进行了全面讨论。从表观遗传调控到其潜在的治疗效果,我们希望对SIRT1在CRC中的作用提供新的见解和全面的理解。
    The class-III histone deacetylase SIRT1 is the most extensively investigated sirtuin deacetylase. It is resistant to the broad deacetylase inhibitor trichostatin A and depends on oxidized nicotinamide adenine nucleotide (NAD+). SIRT1 plays a crucial role in the tumorigenesis of numerous types of cancers, including colorectal cancer (CRC). Accumulating evidence indicates that SIRT1 is a therapeutic target for CRC; however, the function and underlying mechanism of SIRT1 in CRC still need to be elucidated. Herein, we provide a detailed and updated review to illustrate that SIRT1 regulates many processes that go awry in CRC cells, such as apoptosis, autophagy, proliferation, migration, invasion, metastasis, oxidative stress, resistance to chemo-radio therapy, immune evasion, and metabolic reprogramming. Moreover, we closely link our review to the clinical practice of CRC treatment, summarizing the mechanisms and prospects of SIRT1 inhibitors in CRC therapy. SIRT1 inhibitors as monotherapy in CRC or in combination with chemotherapy, radiotherapy, and immune therapies are comprehensively discussed. From epigenetic regulation to its potential therapeutic effect, we hope to offer novel insights and a comprehensive understanding of SIRT1\'s role in CRC.
    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:大量证据表明组蛋白脱乙酰酶(HDAC)抑制剂可减少骨关节炎(OA)动物模型中的软骨破坏。肿瘤坏死因子(TNF)-α阻断治疗OA可通过减缓关节损伤提供有效的关节保护。探讨依那西普(TNF-α抑制剂)对大鼠OA发生发展的影响及对大鼠伤害性行为和HDACs表达的影响。软骨中的RUNX2和MMP13。
    方法:通过前交叉韧带横断(ACLT)诱导Wistar大鼠OA。ACLT+依那西普(1和5mg/kg)组,在ACLT后连续5周腹膜内给予1或5毫克(mg)依那西普。分析痛觉行为和膝关节宽度的变化。对软骨进行组织学和免疫组织化学评估。
    结果:与单独使用ACLT相比,ACLT+依那西普显著改善了机械异常性疼痛和负重分布。在用依那西普治疗的OA大鼠中,软骨变性和滑膜炎明显低于ACLT大鼠。受OA影响的软骨还显示响应依那西普的HDAC6、7、RUNX-2和MMP-13的表达降低,但HDAC4的表达增加。
    结论:我们的研究表明依那西普治疗(1)减轻了大鼠OA和滑膜炎的发展,(2)减少伤害感受,和(3)调节软骨细胞代谢,可能通过抑制细胞HDAC6和HDAC7,RUNX2和MMP13并增加HDAC4表达。根据新的证据,依那西普可能在OA中具有治疗潜力。
    OBJECTIVE: Mounting evidence suggests that histone deacetylases (HDAC) inhibitors reduce cartilage destruction in animal models of osteoarthritis (OA). Tumor necrosis factor (TNF)-α-blocking treatment for OA may provide effective joint protection by slowing joint damage. To investigate the effects of intraperitoneal administration of etanercept (a TNF-α inhibitor) on OA development in rats and changes in the nociceptive behavior of rats and expression of HDACs, RUNX2, and MMP13 in cartilage.
    METHODS: Induction of OA in Wistar rats was accomplished through anterior cruciate ligament transection (ACLT). One or five milligrams (mg) of etanercept was administered intraperitoneally for 5 consecutive weeks after ACLT to the ACLT + etanercept (1 and 5 mg/kg) groups. Nociceptive behavior and changes in knee joint width were analyzed. Cartilage was evaluated histologically and immunohistochemically.
    RESULTS: ACLT + etanercept significantly improved mechanical allodynia and weight-bearing distribution compared to ACLT alone. In OA rats treated with etanercept, cartilage degeneration and synovitis were significantly less pronounced than those in ACLT rats. OA-affected cartilage also showed reduced expression of HDAC 6, 7, RUNX-2, and MMP-13 in response to etanercept but increased expression of HDAC4.
    CONCLUSIONS: Our study demonstrated that etanercept therapy (1) attenuated the development of OA and synovitis in rats, (2) reduced nociception, and (3) regulated chondrocyte metabolism, possibly by inhibiting cell HDAC6 and HDAC7, RUNX2, and MMP13 and increasing HDAC4 expression. Based on new evidence, etanercept may have therapeutic potential in OA.
    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
    在现代癌症治疗中,阻断多个目标是一种标准方法,并且已经有许多双靶标药物可以通过单个分子实现多重抑制。在这里,我们通过基于STAT3抑制剂E28和HDAC抑制剂MS-275的药效团组合策略,设计并合成了一系列具有信号转导和转录激活因子3(STAT3)和组蛋白去乙酰化酶(HDAC)抑制活性的新型衍生物。其中,化合物24(IC50=8.22±0.27μM)在MCF-7乳腺癌细胞中显示出比临床I类HDAC抑制剂MS-275(IC50=14.65±0.24μM)更好的抗肿瘤活性。此外,通过蛋白质印迹分析验证了化合物24对HDAC和STAT3的双重抑制作用.该研究为进一步探索用单个分子实现的STAT3-HDAC途径抑制剂提供了新的工具化合物。
    In modern cancer therapy, blockage of more than one target is a standard approach, and there are already many dual-target drugs that can achieve multiple inhibition through a single molecule. Herein, we designed and synthesized a series of novel derivatives with signal transducer and activator of transcription 3 (STAT3) and histone deacetylase (HDAC) inhibitory activity through strategy of combining pharmacophore based on the STAT3 inhibitor E28 and HDAC inhibitor MS-275. Among them, compound 24 (IC50 = 8.22 ± 0.27 μM) showed better anti-tumor activity than the clinical Class I HDAC inhibitor MS-275 (IC50 = 14.65 ± 0.24 μM) in MCF-7 breast cancer cells. Furthermore, the dual inhibition to HDAC and STAT3 of compound 24 was validated by western blot analysis. The study provides new tool compounds for further exploration of STAT3-HDAC pathway inhibitor achieved with a single molecule.
    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(HDACs)是药物开发过程中极具吸引力的靶标,亚型选择性HDAC抑制剂的开发是HDAC抑制剂的研究方向。作为HDAC家族的重要成员,HDAC3因其异常表达而被发现与许多疾病的病理进展密切相关。在以往的研究中,我们发现了化合物13a,对HDAC1、2和3具有有效的抑制活性。在这项工作中,我们提高了13a的HDAC3同种型选择性,通过合理的药物设计获得了化合物9c。图9c显示HDAC3相对于HDAC1的71倍的选择性,并且可以在体外显著抑制MV4-11细胞的增殖活性。此外,当与维奈托克合用时,9c在体外能有效诱导MV4-11细胞凋亡,降低抗凋亡蛋白的表达,HDAC3选择性抑制剂的开发可能是逆转维奈托克耐药的潜在先导化合物。
    Histone deacetylases (HDACs) are highly attractive targets in the drug development process, and the development of subtype-selective HDAC inhibitors is the research direction for HDAC inhibitors. As an important member of the HDAC family, HDAC3 has been found to be closely related to the pathological progression of many diseases due to its abnormal expression. In previous studies, we discovered compound 13a, which has potent inhibitory activity against HDAC1, 2, and 3. In this work, we improved the HDAC3 isotype selectivity of 13a, and obtained compound 9c through rational drug design. 9c shows a selectivity of 71 fold for HDAC3 over HDAC1 and can significantly inhibit the proliferation activity of MV4-11 cells in vitro. Furthermore, when combined with Venetoclax, 9c can effectively induce apoptosis in MV4-11 cells in vitro and reduce the expression of anti-apoptotic proteins, the development of HDAC3 selective inhibitors may serve as a potential lead compound to reverse Venetoclax resistance.
    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
    已经研究了组蛋白去乙酰化家族蛋白在通过去乙酰化非组蛋白蛋白调节病毒复制中的功能。RIG-I(维甲酸诱导基因I)是RNA病毒诱导的先天抗病毒信号通路中的关键蛋白。我们之前的研究表明,HDAC8(组蛋白去乙酰化酶8)参与先天抗病毒免疫反应,但病毒感染过程中的潜在机制仍不清楚。在这项研究中,我们表明HDAC8参与了水泡性口炎病毒(VSV)复制的调节。HDAC8的过表达抑制,而敲低促进VSV复制。进一步的探索表明,HDAC8与RIG-I相互作用并脱乙酰,这最终导致增强先天抗病毒免疫反应。总的来说,我们的数据清楚地表明,HDAC8通过促进RIG-I介导的干扰素产生和下游信号通路抑制VSV复制.
    Histone deacetylates family proteins have been studied for their function in regulating viral replication by deacetylating non-histone proteins. RIG-I (Retinoic acid-inducible gene I) is a critical protein in RNA virus-induced innate antiviral signaling pathways. Our previous research showed that HDAC8 (histone deacetylase 8) involved in innate antiviral immune response, but the underlying mechanism during virus infection is still unclear. In this study, we showed that HDAC8 was involved in the regulation of vesicular stomatitis virus (VSV) replication. Over-expression of HDAC8 inhibited while knockdown promoted VSV replication. Further exploration demonstrated that HDAC8 interacted with and deacetylated RIG-I, which eventually lead to enhance innate antiviral immune response. Collectively, our data clearly demonstrated that HDAC8 inhibited VSV replication by promoting RIG-I mediated interferon production and downstream signaling pathway.
    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
    靶向程序性细胞死亡-1/配体1(PD-1/PD-L1)途径是最有前途的癌症治疗策略之一。研究表明,HDAC抑制剂可以通过调节PD-L1的表达来增强抗肿瘤免疫反应。在这里,我们设计并合成了一系列新型的基于酰肼的小分子HDAC抑制剂,化合物HQ-30显示出选择性的HDAC3抑制作用(IC50=89nM)和显着的PD-L1降解活性(DC50=5.7μM,在10μM时Dmax=80%)。进一步的研究表明,HQ-30通过调节溶酶体中的组织蛋白酶B(CTSB)诱导PD-L1的降解。Further,HQ-30可以增强肿瘤中CD3+CD4+辅助性T细胞和CD3+CD8+细胞毒性T细胞的浸润,从而激活肿瘤免疫微环境。此外,HQ-30具有良性毒性特征(LD50>1000mg/kg)和良好的药代动力学性质(F=57%)。一起来看,HQ-30作为肿瘤免疫治疗的小分子表观遗传调节剂值得进一步研究。
    Targeting the programmed cell death-1/ligand 1 (PD-1/PD-L1) pathway is one of the most promising cancer treatment strategies. Studies have shown that HDAC inhibitors can enhance the antitumor immune response by modulating the expression of PD-L1. Herein, we designed and synthesized a series of novel hydrazide-based small molecule HDAC inhibitors; among them, compound HQ-30 showed selective HDAC3 inhibition (IC50 = 89 nM) and remarkable PD-L1-degrading activity (DC50 = 5.7 μM, Dmax = 80% at 10 μM). Further studies revealed that HQ-30 induced the degradation of PD-L1 by regulating cathepsin B (CTSB) in the lysosomes. Further, HQ-30 could enhance the infiltration of CD3+ CD4+ helper T and CD3+ CD8+ cytotoxic T cells in tumors, thus activating the tumor immune microenvironment. Moreover, HQ-30 possessed a benign toxicity profile (LD50 > 1000 mg/kg) and favorable pharmacokinetic properties (F = 57%). Taken together, HQ-30 is worthy of further investigation as a small molecule-based epigenetic modulator of tumor immunotherapy.
    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)

公众号