histone deacetylases (HDACs)

组蛋白脱乙酰酶 (HDAC)
  • 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
    组蛋白脱乙酰酶(HDACs)是重要的抗癌药物靶点。现有的FDA批准的药物靶向HDAC的催化口袋,在HDAC的亚家族(类)中保守。然而,工程特异性是一个重要的目标。在这里,我们使用分子建模方法在HDAC4和转录辅抑制因子成分蛋白NCoR之间的界面处鉴定和靶向IIA类HDAC-HDAC4特异性的潜在新型口袋。使用集合对接方法结合共识评分来筛选这些口袋,以鉴定具有与目前已知的HDAC调节剂不同的结合机制的化合物。在实验测定中比较了属于不同HDAC家族的HDAC4和HDAC3之间的结合。HDAC4被化合物88402显著抑制,但不被HDAC3抑制。另外两种化合物(67436和134199)对两种HDAC都具有低微摩尔范围的IC50值,这与已知的HDAC4抑制剂SAHA(伏立诺他)相当。然而,这两种化合物的HDAC3抑制剂明显弱于SAHA,因此更具选择性,虽然程度有限。五种化合物对人乳腺癌和/或尿路上皮癌细胞系具有活性。本结果提示开发选择性HDAC4调节剂的潜在机械和化学方法。
    Histone deacetylases (HDACs) are important cancer drug targets. Existing FDA-approved drugs target the catalytic pocket of HDACs, which is conserved across subfamilies (classes) of HDAC. However, engineering specificity is an important goal. Herein, we use molecular modeling approaches to identify and target potential novel pockets specific to Class IIA HDAC-HDAC4 at the interface between HDAC4 and the transcriptional corepressor component protein NCoR. These pockets were screened using an ensemble docking approach combined with consensus scoring to identify compounds with a different binding mechanism than the currently known HDAC modulators. Binding was compared in experimental assays between HDAC4 and HDAC3, which belong to a different family of HDACs. HDAC4 was significantly inhibited by compound 88402 but not HDAC3. Two other compounds (67436 and 134199) had IC50 values in the low micromolar range for both HDACs, which is comparable to the known inhibitor of HDAC4, SAHA (Vorinostat). However, both of these compounds were significantly weaker inhibitors of HDAC3 than SAHA and thus more selective, albeit to a limited extent. Five compounds exhibited activity on human breast carcinoma and/or urothelial carcinoma cell lines. The present result suggests potential mechanistic and chemical approaches for developing selective HDAC4 modulators.
    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
    γ-氨基丁酸A型(GABAA)受体γ2亚基基因突变,GABRG2与难治性癫痫有关。越来越多的证据表明辛二酰苯胺异羟肟酸(SAHA),广谱组蛋白乙酰转移酶(HDAC)抑制剂,可以抑制癫痫发作。然而,所涉及的机制仍然未知。本研究旨在探讨SAHA治疗GABRG2突变所致难治性癫痫的抗癫痫作用及其机制。在表达人突变体GABRG2(F343L)的斑马鱼系中,Tg(hGABRG2F343L),发现SAHA可以减少癫痫发作,游泳活动,和神经元活动。在用GABAA受体亚基转染的Tg(hGABRG2F343L)斑马鱼和HEK293T细胞中,SAHA可以提高全乙酰化水平,降低HDAC1/10的表达。GABAA受体亚基表达的降低可通过SAHA治疗在体内和体外得到挽救。这可能是基因转录和蛋白质运输增加的结果。该过程可能涉及组蛋白H3和H4的上调乙酰化以及Bip的表达。一起来看,我们的数据证明,组蛋白和非组蛋白乙酰化可能有助于SAHA在GABRG2(F343L)突变引起的难治性癫痫中的抗癫痫作用,证明SAHA是难治性癫痫的有前途的治疗剂。
    Mutations in the γ-amino butyric acid type A (GABAA) receptor γ2 subunit gene, GABRG2, have been associated with refractory epilepsy. Increasing evidence indicates that suberoylanilide hydroxamic acid (SAHA), a broad-spectrum histone acetyltransferases (HDACs) inhibitor, can inhibit seizure onset. However, the mechanisms involved remains unknown. The present study aimed to explore the anti-epileptic effect and underlying mechanisms of SAHA in the treatment of refractory epilepsy induced by GABRG2 mutation. In the zebrafish line expressing human mutant GABRG2(F343L), Tg(hGABRG2F343L), SAHA was found to reduce seizure onset, swimming activity, and neuronal activity. In both Tg(hGABRG2F343L) zebrafish and HEK293T cells transfected with GABAA receptor subunits, SAHA could improve the pan-acetylation level and reduce the expression of HDAC1/10. The decreased expressions of GABAA receptor subunits could be rescued by SAHA treatment both in vivo and in vitro, which might be the result of increased gene transcription and protein trafficking. The up-regulated acetylation of histone H3 and H4 as well as Bip expression might be involved in the process. Taken together, our data proved that both histone and non-histone acetylation might contribute to the anti-epileptic effect of SAHA in refractory epilepsy caused by GABRG2(F343L) mutation, demonstrating SAHA as a promising therapeutic agent for refractory epilepsy.
    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)在基因转录中至关重要,从组蛋白中去除乙酰基。它们还影响非组蛋白的去乙酰化,有助于调节各种生物过程。因此,HDAC在各种疾病中起着关键作用,包括癌症,神经退行性疾病,和炎症,突出它们作为治疗靶点的潜力。本文综述了四类人类HDAC的结构和功能。虽然目前有四种HDAC抑制剂可用于治疗血液恶性肿瘤,许多其他人正在进行临床试验。然而,它们的非选择性毒性需要持续研究更安全和更有效的类选择性或同工型选择性抑制剂.计算方法有助于发现具有所需效力和/或选择性的HDAC抑制剂。这些方法包括基于配体的方法,如脚手架跳跃,药效团建模,三维定量结构-活性关系,和基于结构的虚拟筛选(分子对接)。此外,分子动力学模拟领域的最新进展,结合泊松-玻尔兹曼/分子力学广义Born表面积技术,改善了配体结合亲和力的预测。在这次审查中,我们深入研究了这些方法对设计和鉴定HDAC抑制剂的贡献。
    Histone deacetylases (HDACs) are crucial in gene transcription, removing acetyl groups from histones. They also influence the deacetylation of non-histone proteins, contributing to the regulation of various biological processes. Thus, HDACs play pivotal roles in various diseases, including cancer, neurodegenerative disorders, and inflammatory conditions, highlighting their potential as therapeutic targets. This paper reviews the structure and function of the four classes of human HDACs. While four HDAC inhibitors are currently available for treating hematological malignancies, numerous others are undergoing clinical trials. However, their non-selective toxicity necessitates ongoing research into safer and more efficient class-selective or isoform-selective inhibitors. Computational methods have aided the discovery of HDAC inhibitors with the desired potency and/or selectivity. These methods include ligand-based approaches, such as scaffold hopping, pharmacophore modeling, three-dimensional quantitative structure-activity relationships, and structure-based virtual screening (molecular docking). Moreover, recent developments in the field of molecular dynamics simulations, combined with Poisson-Boltzmann/molecular mechanics generalized Born surface area techniques, have improved the prediction of ligand binding affinity. In this review, we delve into the ways in which these methods have contributed to designing and identifying HDAC inhibitors.
    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
    背景:细胞死亡失调是癌症的共同特征,对这一过程的抵抗通常发生在肺癌中。了解异常细胞死亡的分子机制很重要。最近的研究强调钙调蛋白调节的血影蛋白相关蛋白3(CAMSAP3)参与肺癌的侵袭性,其对细胞死亡调节的影响在很大程度上仍未被探索。
    方法:使用CRISPR-Cas9系统在肺癌细胞中敲除CAMSAP3。使用MTT和自噬检测测定法评估细胞死亡和自噬。通过蛋白质组学分析和免疫沉淀进行蛋白质相互作用。通过免疫印迹和免疫荧光技术分析蛋白质表达及其细胞质定位。
    结果:本研究揭示了肺癌患者低CAMSAP3表达与低总生存率之间的显著相关性。蛋白质组学分析鉴定了高迁移率基团盒1(HMGB1)作为参与细胞死亡调节的候选相互作用蛋白。曲古霉素A(TSA)治疗,组蛋白去乙酰化酶(HDACs)的抑制剂导致HMGB1乙酰化增加,并易位到细胞质和分泌,从而诱导自噬性细胞死亡。然而,在CAMSAP3敲除的肺癌细胞中,这一过程减弱.机械上,免疫沉淀表明CAMSAP3和HMGB1之间的相互作用,特别是其乙酰化形式,其中该复合物在TSA存在下升高。
    结论:CAMSAP3通过与细胞质乙酰化HMGB1相互作用并增强其释放,是TSA介导的自噬性细胞死亡的先决条件。
    结论:这一发现为CAMSAP3在调节细胞死亡中的作用提供了分子见解,强调其作为肺癌治疗靶点的潜力。
    BACKGROUND: Deregulation of cell death is a common characteristic of cancer, and resistance to this process often occurs in lung cancer. Understanding the molecular mechanisms underlying an aberrant cell death is important. Recent studies have emphasized the involvement of calmodulin-regulated spectrin-associated protein 3 (CAMSAP3) in lung cancer aggressiveness, its influence on cell death regulation remains largely unexplored.
    METHODS: CAMSAP3 was knockout in lung cancer cells using CRISPR-Cas9 system. Cell death and autophagy were evaluated using MTT and autophagic detection assays. Protein interactions were performed by proteomic analysis and immunoprecipitation. Protein expressions and their cytoplasmic localization were analyzed through immunoblotting and immunofluorescence techniques.
    RESULTS: This study reveals a significant correlation between low CAMSAP3 expression and poor overall survival rates in lung cancer patients. Proteomic analysis identified high mobility group box 1 (HMGB1) as a candidate interacting protein involved in the regulation of cell death. Treatment with trichostatin A (TSA), an inhibitor of histone deacetylases (HDACs) resulted in increased HMGB1 acetylation and its translocation to the cytoplasm and secretion, thereby inducing autophagic cell death. However, this process was diminished in CAMSAP3 knockout lung cancer cells. Mechanistically, immunoprecipitation indicated an interaction between CAMSAP3 and HMGB1, particularly with its acetylated form, in which this complex was elevated in the presence of TSA.
    CONCLUSIONS: CAMSAP3 is prerequisite for TSA-mediated autophagic cell death by interacting with cytoplasmic acetylated HMGB1 and enhancing its release.
    CONCLUSIONS: This finding provides molecular insights into the role of CAMSAP3 in regulating cell death, highlighting its potential as a therapeutic target for lung cancer 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
    与糖尿病患者中枢神经系统并发症相关的表观遗传机制在很大程度上是未知的。本研究调查了组蛋白乙酰化机制在1型和2型糖尿病小鼠模型中触发认知功能障碍的作用。糖尿病参数的动态变化,如空腹血糖水平,葡萄糖耐量试验,糖尿病诱导后观察胰岛素水平。通过Morris水迷宫检查的T1D和T2D小鼠的认知功能显着减弱,新颖的物体识别测试,和Y迷宫与对照组相比。组蛋白谱分析显示,与对照组相比,T1D和T2D小鼠的皮质和海马中的H3K9/14和H4K12乙酰化显着降低。虽然组蛋白去乙酰化酶(HDAC)活性在T1D和T2D小鼠的脑区显著升高,组蛋白乙酰转移酶(HAT)活性保持不变。在T1D和T2D脑区观察到的HDAC2,HDAC3蛋白和mRNA表达显着增加可能证实HDAC活性增加。在糖尿病脑中HDAC1、5、6和7的蛋白和mRNA表达没有观察到显著变化。减少H3K9/14和H4K12乙酰化平行转录抑制记忆相关标记BDNF,SYP,T1D和T2D的皮质和海马中的PSD-95。TrichostatinA对HDAC活性的药理学抑制通过改善BDNF增强了T1D和T2D中观察到的认知变化,SYP,psd-95.本研究为与糖尿病依赖性记忆变化有关的分子机制提供了更好的见解,这可以帮助在该领域开发疗法的新进展。
    Epigenetic mechanisms related to diabetes-afflicted CNS complications are largely unknown. The present study investigated the role of histone acetylation mechanisms triggering cognitive dysfunction in the Type 1 and 2 diabetic mice model. Dynamic changes in diabetic parameters like fasting blood glucose levels, glucose tolerance test, and insulin levels were observed after the induction of diabetes. Cognitive performance was significantly diminished in T1D and T2D mice examined by the Morris water maze, novel object recognition test, and Y Maze as compared to controls. Histone profiling revealed a significant reduction in H3K9/14 and H4K12 acetylation in the cortex and hippocampus of T1D and T2D mice vs Controls. While histone deacetylase (HDAC) activity was significantly elevated in brain regions of T1D and T2D mice, the histone acetyltransferase (HAT) activity remain unchanged. Significantly increased HDAC 2, HDAC 3 protein and mRNA expression observed in T1D and T2D brain regions may corroborate for increased HDAC activity. No significant change was observed in protein and mRNA expression of HDAC 1, 5, 6, and 7 in diabetic brains. Reduced H3K9/14 and H4K12 acetylation paralleled transcriptional repression of memory-related markers BDNF, SYP, and PSD-95 in the cortex and hippocampus of T1D and T2D. Pharmacological inhibition of HDAC activity by Trichostatin A enhanced the cognitive changes observed in T1D and T2D by ameliorating BDNF, SYP, Psd-95. The present study provides a better insight into molecular mechanisms related to diabetes-dependent memory changes that can help to generate new advances for therapeutics to be developed in this area.
    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)在妇科肿瘤的肿瘤免疫微环境中的作用仍未被探索。我们整合了来自癌症基因组图谱和人类蛋白质图谱的数据,以检查乳腺中的HDAC表达,子宫颈,卵巢,和子宫内膜癌。HDAC表达升高与不良预后和高度恶性癌症亚型相关。基因集富集分析显示HDAC表达与肿瘤增殖特征呈正相关,而与肿瘤炎症特征呈负相关。HDAC表达增加与自然杀伤(NK)浸润减少有关,NKT,和CD8+T细胞,与PSMB10,NKG7,CCL5,CD27,HLA-DQA1和HLA-DQB1的表达呈负相关。在小鼠4T1乳腺癌模型中,用辛二酰苯胺异羟肟酸(SAHA;HDAC抑制剂)和PD-1抗体治疗可显着抑制肿瘤的生长以及CD3和CD8T细胞的浸润。实时聚合酶链反应显示Psmb10,Nkg7,Ccl5,Cd8a的表达上调,Cxcr6和Cxcl9基因,而Ctnnb1和Myc基因被抑制,表明肿瘤抑制和免疫微环境激活。我们的研究表明,HDAC在妇科癌症中发挥促进肿瘤和免疫抑制的作用。建议HDAC抑制剂作为这些癌症的潜在治疗剂。
    The role of histone deacetylases (HDACs) in the tumor immune microenvironment of gynecologic tumors remains unexplored. We integrated data from The Cancer Genome Atlas and Human Protein Atlas to examine HDAC expression in breast, cervical, ovarian, and endometrial cancers. Elevated HDAC expression correlated with poor prognosis and highly malignant cancer subtypes. Gene Set Enrichment Analysis revealed positive associations between HDAC expression and tumor proliferation signature, while negative associations were found with tumor inflammation signature. Increased HDAC expression was linked to reduced infiltration of natural killer (NK), NKT, and CD8+ T cells, along with negative associations with the expression of PSMB10, NKG7, CCL5, CD27, HLA-DQA1, and HLA-DQB1. In a murine 4T1 breast cancer model, treatment with suberoylanilide hydroxamic acid (SAHA; HDAC inhibitor) and PD-1 antibody significantly inhibited tumor growth and infiltration of CD3+ and CD8+ T cells. Real-time polymerase chain reaction revealed upregulated expressions of Psmb10, Nkg7, Ccl5, Cd8a, Cxcr6, and Cxcl9 genes, while Ctnnb1 and Myc genes were inhibited, indicating tumor suppression and immune microenvironment activation. Our study revealed that HDACs play tumor-promoting and immunosuppressive roles in gynecologic cancers, suggesting HDAC inhibitors as potential therapeutic agents for these 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
    脱髓鞘病变中未达到的临床目标是在神经变性之前恢复髓鞘。N-乙酰天冬氨酸(NAA)是天冬氨酸的乙酰化衍生物形式,在健康的大脑中丰富,但在创伤性脑损伤和神经退行性病变患者中严重减少。细胞外NAA变化如何影响髓鞘再生过程,因此,少突胶质细胞对髓鞘再生轴突的能力仍未被探索。这里,我们评估了少突胶质(OL)小鼠细胞系Oli-neuM在不同浓度的NAA下的髓鞘再生特性,qPCR,免疫荧光测定,和体外参与测试,NAA剂量与健康大脑和脑损伤期间观察到的剂量一致。我们观察到少突胶质细胞通过刺激分化和以时间调节的方式促进髓磷脂蛋白的基因表达来响应NAA水平的降低。低剂量的NAA可有效刺激Oli-neuM与合成轴突接合。此外,我们显示了在NAA或Clobetasol处理下MBP基因表达所必需的特定组蛋白脱乙酰酶的浓度依赖性表达。这些数据与少突胶质细胞通过经由脱乙酰酶激活激活髓鞘再生过程来响应降低NAA浓度的想法一致。
    An unmet clinical goal in demyelinating pathologies is to restore the myelin sheath prior to neural degeneration. N-acetylaspartate (NAA) is an acetylated derivative form of aspartate, abundant in the healthy brain but severely reduced during traumatic brain injury and in patients with neurodegenerative pathologies. How extracellular NAA variations impact the remyelination process and, thereby, the ability of oligodendrocytes to remyelinate axons remains unexplored. Here, we evaluated the remyelination properties of the oligodendroglial (OL) mouse cell line Oli-neuM under different concentrations of NAA using a combination of biochemical, qPCR, immunofluorescence assays, and in vitro engagement tests, at NAA doses compatible with those observed in healthy brains and during brain injury. We observed that oligodendroglia cells respond to decreasing levels of NAA by stimulating differentiation and promoting gene expression of myelin proteins in a temporally regulated manner. Low doses of NAA potently stimulate Oli-neuM to engage with synthetic axons. Furthermore, we show a concentration-dependent expression of specific histone deacetylases essential for MBP gene expression under NAA or Clobetasol treatment. These data are consistent with the idea that oligodendrocytes respond to lowering the NAA concentration by activating the remyelination process via deacetylase activation.
    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
    严重急性呼吸道综合症冠状病毒2(SARS-CoV-2)在全球造成约6.72亿感染和685万人死亡。SARS-CoV-2感染后,组蛋白脱乙酰酶(HDAC)过度激活促炎反应,导致刺激乙酰辅酶A和胆固醇进入病毒。HDAC3抑制导致抗炎活性和可能限制COVID-19进展的促炎细胞因子减少。这里,我们通过列举测试其对HDAC3的结合偏好的二面角的扭转,设计了44个先前已知的HD-TAC7的构象集合。通过仔细检查它们在活性位点的位置和结合亲和力,三击被隔离。Cereblon(CRBN)是一种众所周知的E3连接酶,可促进蛋白水解靶向嵌合体(PROTACs)靶向。三个实体,包括HDAC3结合部分(4-乙酰氨基-N-(2-氨基-4氟苯基)苯甲酰胺),一个6碳接头,和CRBN结合配体(泊马度胺)组装以设计4个PROTACs,然后能量最小化并与HDAC3和CRBN对接,分别。随后的分子动力学(MD)和自由能分析证实了类似的结合趋势和有利的能量值。在所有案件中,CRBN的Met88、GLu106、Pro352、Trp380和Trp388残基,HDAC3的Pro23、Arg28、Lys194、Phe199、Leu266、Thr299和Ile346残基参与PROTAC结合。因此,HDAC3和CRBN部分的构象动力学对于PROTAC的放置至关重要,导致目标降解。总的来说,提出的双功能小分子可能有效地靶向HDAC3,刺激先天免疫反应以限制COVID-19过度炎症。这项研究通过限制构象搜索空间来支持设计新的PROTACs的基础,这可能证明对靶向感兴趣的蛋白质更有效。由RamaswamyH.Sarma沟通。
    Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is responsible for about 672 million infections and 6.85 million deaths worldwide. Upon SARS-CoV-2 infection, Histone deacetylases (HDACs) hyperactivate the pro-inflammatory response resulting in stimulation of Acetyl-Coenzyme A and cholesterol for viral entry. HDAC3 inhibition results in the anti-inflammatory activity and reduction of pro-inflammatory cytokines that may restrict COVID-19 progression. Here, we have designed 44 conformational ensembles of previously known HD-TAC7 by enumerating torsions of dihedral angles tested for their binding preferences against HDAC3. Through scrutinizing their placements at active site and binding affinities, three hits were isolated. Cereblon (CRBN) is a well-known E3 ligase that facilitates Proteolysis Targeting Chimeras (PROTACs) targeting. Three entities, including HDAC3-binding moiety (4-acetamido-N-(2-amino-4 fluorophenyl) benzamide), a 6-carbon linker, and CRBN binding ligand (pomalidomide) were assembled to design 4 PROTACs followed by energy minimization and docking against HDAC3 and CRBN, respectively. Subsequent molecular dynamics (MD) and free energy analyses corroborated similar binding trends and favorable energy values. Among all cases, Met88, GLu106, Pro352, Trp380 and Trp388 residues of CRBN, and Pro23, Arg28, Lys194, Phe199, Leu266, Thr299 and Ile346 residues of HDAC3 were engaged in PROTAC binding. Thus, conformational dynamics of both HDAC3 and CRBN moieties are essential for the placement of PROTAC, resulting in target degradation. Overall, the proposed bifunctional small molecules may effectively target HDAC3, stimulating innate immune response to restrict COVID-19 hyperinflammation. This study supports the basis for designing new PROTACs by limiting the conformational search space that may prove more efficient for targeting the protein of interest.Communicated by Ramaswamy H. Sarma.
    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
    越来越多的证据(主要来自实验研究)表明,二甲双胍通过诱导细胞凋亡和抑制癌细胞的生长和增殖而具有抗癌特性。然而,它对负责组蛋白乙酰化状态的酶的影响,在致癌过程中起着关键作用,尚不清楚。因此,我们研究的目的是评估二甲双胍对组蛋白乙酰转移酶(HAT)的影响(即,p300/CBP相关因子(PCAF),p300和CBP)和组蛋白脱乙酰酶(HDAC)(即,人胰腺癌(PC)细胞系中的SIRT-1,1.2B4和PANC-1)。细胞暴露于二甲双胍,HAT抑制剂(HATi),或HATi与二甲双胍的组合24、48或72小时。使用MTT测定确定细胞活力,用膜联蛋白V-Cy3凋亡检测试剂盒测定早期凋亡细胞的百分比。还评估了胱天蛋白酶-9活性。SIRT-1,PCAF,使用RT-PCR和蛋白质印迹方法在mRNA和蛋白质水平上确定p300和CBP表达,分别。我们的结果表明,二甲双胍对caspase-9的反应增加,表明它诱导了1.2B4和PANC-1细胞的凋亡死亡。当单独使用HATi或HATi与二甲双胍的组合治疗时,早期凋亡中的细胞数量和caspase-9的活性降低,与单独的二甲双胍相比。此外,二甲双胍,一个HATI,HATi与二甲双胍的组合也修饰了SIRT-1,PCAF的mRNA表达,CBP,p300然而,二甲双胍没有改变1.2B4细胞中研究基因的表达。Westernblot分析结果显示二甲双胍可降低1.2B4和PANC-1细胞中PCAF的蛋白表达。因此,PCAF可能参与了二甲双胍介导的PC细胞凋亡。
    Accumulating evidence (mainly from experimental research) suggests that metformin possesses anticancer properties through the induction of apoptosis and inhibition of the growth and proliferation of cancer cells. However, its effect on the enzymes responsible for histone acetylation status, which plays a key role in carcinogenesis, remains unclear. Therefore, the aim of our study was to evaluate the impact of metformin on histone acetyltransferases (HATs) (i.e., p300/CBP-associated factor (PCAF), p300, and CBP) and on histone deacetylases (HDACs) (i.e., SIRT-1 in human pancreatic cancer (PC) cell lines, 1.2B4, and PANC-1). The cells were exposed to metformin, an HAT inhibitor (HATi), or a combination of an HATi with metformin for 24, 48, or 72 h. Cell viability was determined using an MTT assay, and the percentage of early apoptotic cells was determined with an Annexin V-Cy3 Apoptosis Detection Assay Kit. Caspase-9 activity was also assessed. SIRT-1, PCAF, p300, and CBP expression were determined at the mRNA and protein levels using RT-PCR and Western blotting methods, respectively. Our results reveal an increase in caspase-9 in response to the metformin, indicating that it induced the apoptotic death of both 1.2B4 and PANC-1 cells. The number of cells in early apoptosis and the activity of caspase-9 decreased when treated with an HATi alone or a combination of an HATi with metformin, as compared to metformin alone. Moreover, metformin, an HATi, and a combination of an HATi with metformin also modified the mRNA expression of SIRT-1, PCAF, CBP, and p300. However, metformin did not change the expression of the studied genes in 1.2B4 cells. The results of the Western blot analysis showed that metformin diminished the protein expression of PCAF in both the 1.2B4 and PANC-1 cells. Hence, it appears possible that PCAF may be involved in the metformin-mediated apoptosis of PC cells.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号