HDAC8

HDAC8
  • 文章类型: Journal Article
    组蛋白脱乙酰酶构成一组参与几种生物过程的酶。值得注意的是,抑制HDAC8已成为各种疾病的治疗策略。目前的HDAC8抑制剂缺乏选择性并靶向多种HDAC。因此,人们越来越认识到需要选择性HDAC8抑制剂来增强治疗性干预措施的有效性.在我们目前的研究中,我们采用了多方面的方法,包括定量结构-活动关系(QSAR)结合定量阅读-跨结构-活动关系(q-RASAR)建模,药效基团作图,分子对接,和分子动力学(MD)模拟。建立的q-RASAR模型具有较高的统计意义和预测能力(Q2F1:0.778,Q2F2:0.775)。详细讨论了重要描述符的贡献,以深入了解HDAC8抑制中的关键结构特征。最佳药效团假设表现出高回归系数(0.969)和低均方根偏差(0.944),强调正确定向氢键受体(HBA)的重要性,环芳族(RA),和锌结合基团(ZBG)在设计有效的HDAC8抑制剂中的特征。为了确认q-RASAR和药效基团作图的结果,对五种有效化合物(44、54、82、102和118)进行分子对接分析,以进一步了解与HDAC8酶相互作用至关重要的这些结构特征。最后,进行了最具活性的化合物(54,用药效团假说正确定位)和最不活性的化合物(34,用药效团假说不良定位)的MD模拟研究,以验证上述研究的观察结果。这项研究不仅完善了我们对HDAC8抑制的基本结构特征的理解,而且为合理设计新型选择性HDAC8抑制剂提供了一个强大的框架,这可能为从事HDAC8靶向疗法开发的药物化学家和研究人员提供见解。
    Histone deacetylases constitute a group of enzymes that participate in several biological processes. Notably, inhibiting HDAC8 has become a therapeutic strategy for various diseases. The current inhibitors for HDAC8 lack selectivity and target multiple HDACs. Consequently, there is a growing recognition of the need for selective HDAC8 inhibitors to enhance the effectiveness of therapeutic interventions. In our current study, we have utilized a multi-faceted approach, including Quantitative Structure-Activity Relationship (QSAR) combined with Quantitative Read-Across Structure-Activity Relationship (q-RASAR) modeling, pharmacophore mapping, molecular docking, and molecular dynamics (MD) simulations. The developed q-RASAR model has a high statistical significance and predictive ability (Q2F1:0.778, Q2F2:0.775). The contributions of important descriptors are discussed in detail to gain insight into the crucial structural features in HDAC8 inhibition. The best pharmacophore hypothesis exhibits a high regression coefficient (0.969) and a low root mean square deviation (0.944), highlighting the importance of correctly orienting hydrogen bond acceptor (HBA), ring aromatic (RA), and zinc-binding group (ZBG) features in designing potent HDAC8 inhibitors. To confirm the results of q-RASAR and pharmacophore mapping, molecular docking analysis of the five potent compounds (44, 54, 82, 102, and 118) was performed to gain further insights into these structural features crucial for interaction with the HDAC8 enzyme. Lastly, MD simulation studies of the most active compound (54, mapped correctly with the pharmacophore hypothesis) and the least active compound (34, mapped poorly with the pharmacophore hypothesis) were carried out to validate the observations of the studies above. This study not only refines our understanding of essential structural features for HDAC8 inhibition but also provides a robust framework for the rational design of novel selective HDAC8 inhibitors which may offer insights to medicinal chemists and researchers engaged in the development of HDAC8-targeted 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
    邻苯二胺对合成和生物学性质有价值。合成了新的乙酰胺3(a-c)和4(a-c),并将其表征为新型N-氨基卤化物5(a-c)和6(a-c)的前体。4a的结构,5(a-b),和6(a-b)通过单晶X射线证实。对接研究确定了具有有利的吉布斯自由能值的化合物,用于结合组蛋白脱乙酰酶8(HDAC8),一种靶向抗癌药物开发的酶。这些化合物与HDAC8的正构和变构袋结合,类似于曲古抑菌素A(TSA),HDAC8抑制剂。图6(a-c)含有羟基乙酰胺部分作为锌结合基团,邻苯二甲酰亚胺部分作为封端基团,和氨基乙酰胺部分作为连接基团,这对配体-受体结合很重要。ΔG值表明化合物5b,6b,与TSA相比,6c对变构口袋中的HDAC8具有更高的亲和力。对HDAC8的抑制活性的体外评估显示,化合物3(a-c)和5(a-c)显示出相似的抑制作用(IC50),范围为0.445至0.751μM。化合物6(a-c)具有较好的亲和力,其中6a(IC50=28nM)和6b(IC50=0.18μM)显示出略低于TSA(IC50=26nM)的有效抑制作用。这些发现表明,所研究的化合物有望成为进一步生物学研究的潜在候选者。
    Phthalimides are valuable for synthesis and biological properties. New acetamides 3(a-c) and 4(a-c) were synthesized and characterized as precursors for novel N-aminophalimides 5(a-c) and 6(a-c). Structures of 4a, 5(a-b), and 6(a-b) were confirmed by single crystal X-ray. Docking studies identified compounds with favorable Gibbs free energy values for binding to histone deacetylase 8 (HDAC8), an enzyme targeted for anticancer drug development. These compounds bound to both the orthosteric and allosteric pockets of HDAC8, similar to Trichostatin A (TSA), an HDAC8 inhibitor. 6(a-c) contain hydroxyacetamide moiety as a zinc-binding group, a phthalimide moiety as a capping group, and aminoacetamide moiety as a linker group, which are important for ligand-receptor binding. ΔG values indicated that compounds 5b, 6b, and 6c had higher affinity for HDAC8 in the allosteric pocket compared to TSA. In vitro evaluation of inhibitory activities on HDAC8 revealed that compounds 3(a-c) and 5(a-c) showed similar inhibitory effects (IC50 ) ranging from 0.445 to 0.751 μM. Compounds 6(a-c) showed better affinity, with 6a (IC50  = 28 nM) and 6b (IC50  = 0.18 μM) showing potent inhibitory effects slightly lower than TSA (IC50  = 26 nM). These findings suggest that the studied compounds hold promise as potential candidates for further biological investigations.
    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
    背景:三阴性乳腺癌(TNBC)是没有任何批准的靶向治疗的最具侵袭性的癌症类型。表观遗传过程在癌细胞进展中起着关键作用,而组蛋白去乙酰化酶8(HDAC8)已被证明是乳腺癌的潜在癌基因,其潜在的分子机制尚不清楚。因此,本研究,旨在评估HDAC8癌变在乳腺癌进展中的潜在机制。
    方法:HDAC8在细胞凋亡等癌细胞过程中的潜在作用,入侵,迁移,血管生成,和癌症干细胞(CSC)标志物通过使用流式细胞术膜联蛋白V-FITC/碘化丙啶(PI)进行评估,逆转录-聚合酶链反应(RT-qPCR),Matrigel包被的transwell板和两种细胞系的伤口愈合测定。还使用乳腺癌异种移植模型研究了HDAC8对肿瘤发展的影响。
    结果:HDAC8表达在细胞系中显著下调,用KO载体转染后。HDAC8的下调显著降低了细胞迁移,血管生成,在MDAMB-468和MDA-MB-231细胞系中诱导凋亡和侵袭。HDAC8敲除的TNBC细胞系具有较低水平的癌症干性标志物,例如malogin-1(CD133),CD44、BMI1和醛脱氢酶1(ALDH1)。此外,在乳腺癌异种移植模型中,HDAC8基因敲除抑制肿瘤生长.
    结论:研究结果表明,敲除HDAC8在BC细胞中保留了几种抗癌作用,如诱导细胞凋亡,减少迁移,入侵,血管生成和去除CSC标志物。
    BACKGROUND: Triple-negative breast cancer (TNBC) is the most aggressive type of cancer without any approved targeted therapy. Epigenetic processes have a pivotal role in cancer cell progression and while histone deacetylase 8 (HDAC8) has been proven as a potential oncogene in breast cancer, its underlying molecular mechanism is not known. Therefore, the present study, aimed to evaluate the underlying mechanism of the HDAC8 carcinogenesis in breast cancer progression.
    METHODS: The potential role of HDAC8 in cancer cell processes such as apoptosis, invasion, migration, angiogenesis, and cancer stem cells (CSCs) markers were evaluated by using flow cytometry Annexin V-FITC/propidium iodide (PI), reverse transcription-polymerase chain reaction (RT-qPCR), Matrigel-coated transwell plates and wound healing assay on both cell lines. The impact of HDAC8 on tumor development was also studied using a breast cancer xenograft model.
    RESULTS: HDAC8 expression was significantly downregulated in the cell lines, post-transfection with KO-vector. Downregulation of HDAC8 dramatically decreased cell migration, angiogenesis, and invasion while inducing apoptosis in MDAMB-468 and MDA-MB-231 cell lines. HDAC8 knocked out TNBC cell lines had lower levels of cancer stemness markers, such as prominin-1 (CD133), CD44, BMI1, and Aldehyde dehydrogenase 1 (ALDH1). Additionally, the knockout of HDAC8 inhibited tumor growth in a breast cancer xenograft model.
    CONCLUSIONS: The findings show that knocking out HDAC8 retains several anticancer actions in BC cells, such as inducing apoptosis, reducing migration, invasion, angiogenesis and removing CSCs markers.
    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
    以乳腺癌(BC)为目标,表观遗传调节可能是一种有前途的治疗策略,由于其在起源中的作用,增长,和BC的转移。丙戊酸(VPA)是一种众所周知的组蛋白脱乙酰酶抑制剂(HDACi),由于其表观遗传焦点,需要深入研究以了解它可能在BC细胞中引起的影响。这项工作的目的是有助于探索VPA使用MCF-7杀死癌细胞的完整药理机制。LC-MS/MS代谢组学研究应用于用VPA处理的MCF-7。结果表明,VPA通过改变代谢途径促进细胞死亡,主要是磷酸戊糖途径(PPP)和2'脱氧-α-D-核糖-1-磷酸降解与降低细胞增殖和细胞生长的代谢物有关,干扰能源并提高活性氧(ROS)水平。我们甚至认为,由于L-半胱氨酸的失调,可能涉及铁凋亡等机制。这些结果表明,VPA在杀死癌细胞方面具有不同的药理学机制,包括凋亡和非凋亡机制。由于HDACis在细胞中的广泛影响,代谢组学方法是为这种分子产生新见解的重要信息来源。
    To target breast cancer (BC), epigenetic modulation could be a promising therapy strategy due to its role in the genesis, growth, and metastases of BC. Valproic acid (VPA) is a well-known histone deacetylase inhibitor (HDACi), which due to its epigenetic focus needs to be studied in depth to understand the effects it might elicit in BC cells. The aim of this work is to contribute to exploring the complete pharmacological mechanism of VPA in killing cancer cells using MCF-7. LC-MS/MS metabolomics studies were applied to MCF-7 treated with VPA. The results show that VPA promote cell death by altering metabolic pathways principally pentose phosphate pathway (PPP) and 2\'deoxy-α-D-ribose-1-phosphate degradation related with metabolites that decrease cell proliferation and cell growth, interfere with energy sources and enhance reactive oxygen species (ROS) levels. We even suggest that mechanisms such as ferropoptosis could be involved due to deregulation of L-cysteine. These results suggest that VPA has different pharmacological mechanisms in killing cancer cells including apoptotic and nonapoptotic mechanisms, and due to the broad impact that HDACis have in cells, metabolomic approaches are a great source of information to generate new insights for this type of 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
    组蛋白去乙酰化酶8(HDAC8)的过度表达可引起多种疾病,和HDAC8的选择性抑制已被吹捧为一个有前途的治疗策略,由于其副作用少。然而,HDAC8选择性抑制的机制尚不清楚。在这项研究中,柔性对接和模拟突变用于探索HDAC8与抑制剂结合过程中甲硫氨酸(M274)的结构变化,以及这种变化的原因。同时,采用转向和常规分子动力学模拟来探索结构变化的稳定性。研究结果表明,M274充当“开关”来控制HDAC8选择性口袋的暴露。仅当L形抑制剂与HDAC8结合时,M274的结构才从翻转变为翻转。这种结构变化形成允许这些抑制剂进入选择性口袋的凹槽。在其他HDAC中,亮氨酸残基在原位取代M274,并且没有观察到相同的结构变化。研究结果揭示了HDAC8的选择性抑制机制,为新型选择性抑制剂的开发提供了指导。
    The overexpression of histone deacetylase 8 (HDAC8) causes several diseases, and the selective inhibition of HDAC8 has been touted as a promising therapeutic strategy due to its fewer side effects. However, the mechanism of HDAC8 selective inhibition remains unclear. In this study, flexible docking and in silico mutation were used to explore the structural change of methionine (M274) during HDAC8 binding to inhibitors, along with the reason for this change. Meanwhile, steered and conventional molecular dynamics simulations were employed to explore the stability of the structural change. The findings suggest that M274 acts as a \"switch\" to control the exposure of the HDAC8-selective pocket. The structure of M274 changes from flipped-out to flipped-in only when L-shaped inhibitors bind to HDAC8. This structural change forms a groove that allows these inhibitors to enter the selective pocket. In other HDACs, a leucine residue replaces M274 in situ, and the same structural change is not observed. The findings reveal the mechanism of selective HDAC8 inhibition and provide guidance for the development of novel selective inhibitors.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    Diketopiperazines (DKPs) have been regarded as an important scaffold from the viewpoint of synthesis due to their biological properties for the treatment of several diseases, including cancer. In this work, two novel series of enantiomeric 2,6-DKPs derived from α-amino acids were synthesized through nucleophilic substitution and intramolecular cyclization reactions. All the compounds were docked against histone deacetylase 8 (HDAC8), which is a promising target for the development of anticancer drugs. These compounds bound into the active site of HDAC8 in a similar way to Trichostatin A (TSA), which is an HDAC8 inhibitor. This study showed that the conformation of the 2,6-DKP ring, stereochemistry, and the type of substituent on the chiral center had an important role in the binding modes. The Gibbs free energies and dissociation constants values of HDAC8-ligand complexes showed that compounds (S)-4hBn, (S)-4m, (R)-4h, and (R)-4m were more stable and affine towards HDAC8 than TSA. The inhibitory activities of 4a, (S)-4h, (S)- and (R)-4(g, l, m) were evaluated in vitro on HDAC8. It was found that compounds (R)-4g (IC50 = 21.54 nM) and (R)-4m (IC50 = 10.81 nM) exhibited better inhibitory activities than TSA (IC50 = 28.32 nM). These results suggested that 2,6-DKPs derivatives may be promising anticancer agents for further biological studies.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    Histone deacetylase 8 (HDAC8) is one of the crucial HDACs responsible for influencing the epigenetic functions of the body. Overexpression of HDAC8 is found to be involved in numerous disease conditions such as tumorigenesis, cell proliferation, cancer, viral infections, neuronal disorders and other epigenetic diseases. Therefore, inhibition of HDAC8 is a primary method to combat these diseases. In this article, a multi-QSAR modeling study on tetrahydroisoquinoline derivatives was conducted to identify important contributions of the structural features of these compounds toward HDAC8 inhibition. All these QSAR modeling techniques were individually validated and justified the observations of each other. The results implied that the tetrahydroisoquinoline moiety may be effective as a cap group than as a linker moiety for HDAC8 inhibition. Different substitutions at the tetrahydroisoquinoline scaffold were also found to be crucial in modulating HDAC8 inhibition.Communicated by Ramaswamy H. Sarma.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    The X-ray crystal structures of HDAC8 complexed with largazole thiol (LAR, PubChem CID: 56663191) and its synthetic variants (Ligand ID in PDB, PubChem CID: L6G, 91667418; L7G, 91667421; L8G, 91667420) (PDB codes: 3RQD, 4RN0, 4RN2 and 4RN1) were analyzed using molecular dynamics simulations to comprehend protein-ligand nonbonding energies (NBEs). The NBEs of ligands\' substructures vis-à-vis active site indicated that pyridyl fragment (F2B4) in L7G and L8G, and amide fragment (F2B5) in LAR and L6G are in high energy states. Based on ligands\' substructures and active site residues properties new compounds were designed by introducing phenolic and amidine moieties, respectively, for F2B4 and F2B5. This improved NBEs of new compounds (NC2, -60.93 kcal/mol; NC3, -42.42 kcal/mol). Also, Zn2+ group (substructure F1) of largazoles was modified with that of SAHA and Trapoxin A. Here, the results indicated in favor of Zn2+ group of Trapoxin A. New compound NC6 incorporating aforesaid modifications i.e. phenolic moiety for F2B4, amidine moiety for F2B5 and Zn2+ group of Trapoxin A in F1, offered best interactions with HDAC8 (-89.75 kcal/mol). Thus, the study revealed new depsipeptides as potential HDAC8 inhibitors. AbbreviationsCAScomposite active siteCHARMMchemistry at Harvard Macromolecular MechanicsCUDAcompute unified device architectureHAThistone acetyletransferaseHDAChistone deacetylaseLARlargazole thiol (or) (2R,5R,8R,11R)-5-methyl-8-(propan-2-yl)-11-[(1E)-4-sulfanylbut-1-en-1-yl]-10-oxa-3,17-dithia-7,14,19,20-tetraazatricyclo[14.2.1.1 ∼ 2,5∼]icosa-1(18),16(19)-diene-6,9,13-trioneL6G(5R, 8S,11S)-5-methyl-8-(propan-2-yl)-11-[(1E)-4-sulfanylbut-1-en-1-yl]-3,17-dithia-7,10,14,19,20-pentaazatricyclo[14.2.1.1 ∼ 2,5∼]icosa-1(18),2(20),16(19)-triene-6,9,13-trione)L7G(5R,8S,11S)-5-methyl-8-(propan-2-yl)-11-[(1E)-4-sulfanylbut-1-en-1-yl]-3-thia-7,10,14,17,21-pentaazatricyclo[14.3.1.1 ∼ 2,5∼]henicosa-1(20),2 (21),16,18-tetraene-6,9,13-trioneL8G(5R,8S,11S)-5-methyl-8-(propan-2-yl)-11-[(1E)-4-sulfanylbut-1-en-1-yl]-3-thia-7,10,14,20,21-pentaazatricyclo[14.3.1.1 ∼ 2,5∼]henicosa-1(20),2(21),16,18-tetraene-6,9,13-trioneMDmolecular dynamicsMOEmolecular operating environmentNAMDnanoscale molecular dynamicsNBEnonbonding energyNBEEelectrostatic nonbonding energyNBEVVan der Waals nonbonding energyNBEFnonbonding energy of fragmentNBEFEelectrostatic nonbonding energy of fragmentNBEFVVan der Waals nonbonding energy of fragmentNCnew compound; Rg: radius of gyration;RMSDroot mean square deviationRMSFroot mean square fluctuationVMDvisual molecular dynamics.Communicated by Ramaswamy H. Sarma.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    UNASSIGNED: The altered expression of histone deacetylase family member 8 (HDAC8) has been found to be linked with various cancers, thereby making its selective inhibition a potential strategy in cancer therapy. Recently, plant secondary metabolites, particularly phenolic compounds, have been shown to possess HDAC inhibitory activity.
    UNASSIGNED: In the present work, we have evaluated the potential of cinnamaldehyde (CAL), cinnamic acid (CA), and cinnamyl alcohol (CALC) (bioactives of Cinnamomum) as well as aqueous cinnamon extract (ACE), to inhibit HDAC8 activity in vitro and in silico.
    UNASSIGNED: HDAC8 inhibitory activity of ACE and cinnamon bioactives was determined in vitro using HDAC8 inhibitor screening kit. Trichostatin A (TSA), a well-known anti-cancer agent and HDAC inhibitor, was used as a positive control. In silico studies included molecular descriptor Analysis molecular docking absorption, distribution, metabolism, excretion, and toxicity prediction, density function theory calculation and synthetic accessibility program.
    UNASSIGNED: Pharmacoinformatics studies implicated that ACE and its Bioactives (CAL, CA, and CALC) exhibited comparable activity with that of TSA. The highest occupied molecular orbitals and lowest unoccupied molecular orbitals along with binding energy of cinnamon bioactives were comparable with that of TSA. Molecular docking results suggested that all the ligands maintained two hydrogen bond interactions within the active site of HDAC8. Finally, the synthetic accessibility values showed that cinnamon bioactives were easy to synthesize compared to TSA.
    UNASSIGNED: It was evident from both the experimental and computational data that cinnamon bioactives exhibited significant HDAC8 inhibitory activity, thereby suggesting their potential therapeutic implications against cancer.
    CONCLUSIONS: Pharmacoinformatics studies revealed that cinnamon bioactives bound to the active site of HDAC8 enzyme in a way similar to that of TSAThe molecular descriptors of cinnamon compounds successfully correlated with TSA values. The binding interactions and energies were also found to be close to TSASynthetic accessibility values showed that cinnamon bioactives were easy to synthesize compared to TSA. Abbreviations used: ACE: Aqueous Cinnamon Extract; DFT: Density Function Theory; CAL: Cinnamaldehyde; CA: Cinnamic Acid; CALC: Cinnamyl Alcohol; MW: Molecular Weight; ROTBs: Rotatable Bonds; ROF: Lipinski\'s Rule of Five; TSA: Trichostatin A; PDB: Protein Data Bank; RMSD: Root Mean Square Deviation; HBA: Hydrogen Bond Acceptor; HBD: Hydrogen Bond Donor; ADMET: Absorption, Distribution, Metabolism, Excretion and Toxicity; FO: Frontier Orbital; HOMOs: Highest Occupied Molecular Orbitals; LUMOs: Lowest Unoccupied Molecular Orbitals; BE: Binding Energy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号