HDAC8

HDAC8
  • 文章类型: Journal Article
    子宫内膜异位症患者,回流的子宫内膜片段逃避宿主免疫监视,发展为子宫内膜异位病变。然而,这种逃避的潜在机制尚未完全阐明。N-Myc和STATInteractor(NMI)已被确定为宿主免疫监视的关键参与者,包括干扰素(IFN)诱导的细胞死亡信号通路。由于雌激素受体β/组蛋白去乙酰化酶8轴的调节,人子宫内膜异位病变的基质细胞中的NMI水平显着降低。在永生化人子宫内膜基质细胞(IHESC)中敲除NMI导致IFNA治疗后参与细胞间粘附和细胞外基质信号传导的基因的RNA水平升高。此外,NMI敲低抑制IFN调节的经典信号通路,例如由干扰素刺激的基因因子3介导的细胞凋亡和IFNA处理后的坏死。相比之下,用IFNA处理的NMI敲低激活促进增殖的非规范IFN调节的信号通路,包括β-Catenin和AKT信号传导。此外,在小鼠子宫内膜异位症模型中,IHESC中的NMI敲除刺激异位病变的生长。因此,NMI是一种新型的子宫内膜异位症抑制剂,IFN暴露后增强子宫内膜细胞的凋亡并抑制其增殖和细胞粘附。
    In patients with endometriosis, refluxed endometrial fragments evade host immunosurveillance, developing into endometriotic lesions. However, the mechanisms underlying this evasion have not been fully elucidated. N-Myc and STAT Interactor (NMI) have been identified as key players in host immunosurveillance, including interferon (IFN)-induced cell death signaling pathways. NMI levels are markedly reduced in the stromal cells of human endometriotic lesions due to modulation by the Estrogen Receptor beta/Histone Deacetylase 8 axis. Knocking down NMI in immortalized human endometrial stromal cells (IHESCs) led to elevated RNA levels of genes involved in cell-to-cell adhesion and extracellular matrix signaling following IFNA treatment. Furthermore, NMI knockdown inhibited IFN-regulated canonical signaling pathways, such as apoptosis mediated by Interferon Stimulated Gene Factor 3 and necroptosis upon IFNA treatment. In contrast, NMI knockdown with IFNA treatment activated non-canonical IFN-regulated signaling pathways that promote proliferation, including β-Catenin and AKT signaling. Moreover, NMI knockdown in IHESCs stimulated ectopic lesions\' growth in mouse endometriosis models. Therefore, NMI is a novel endometriosis suppressor, enhancing apoptosis and inhibiting proliferation and cell adhesion of endometrial cells upon IFN exposure.
    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
    在这项工作中,我们对化合物N-(2-羟苯基)-2-丙基戊酰胺(HO-AAVPA)对乳腺癌(BC)细胞(MCF-7,SKBR3和三阴性BC(TNBC)MDA-MB-231细胞)进行了抗增殖试验,以探讨其与G蛋白偶联雌激素受体(GPER)表达相关的细胞死亡类型的药理学机制.结果显示,HO-AAVPA在5小时或48小时诱导雌激素依赖性(MCF-7)或非雌激素依赖性BC细胞(SKBR3和MDA-MB-231)中的细胞凋亡。在5小时,MCF-7细胞凋亡率为68.4%,MDA-MB-231细胞凋亡率为56.1%;SKBR3为61.6%,MCF-7细胞为54.9%,MDA-MB-231(TNBC)为43.1%。HO-AAVPA增加MCF-7细胞中的S期,减少MCF-7和MDA-MB-231细胞中的G2/M期。在HO-AAVPA存在下,GPER表达比VPA降低更多。总之,HO-AAVPA对细胞凋亡的影响可通过降低GPER表达来调节表观遗传效应。
    In this work, we performed anti-proliferative assays for the compound N-(2-hydroxyphenyl)-2-propylpentanamide (HO-AAVPA) on breast cancer (BC) cells (MCF-7, SKBR3, and triple-negative BC (TNBC) MDA-MB-231 cells) to explore its pharmacological mechanism regarding the type of cell death associated with G protein-coupled estrogen receptor (GPER) expression. The results show that HO-AAVPA induces cell apoptosis at 5 h or 48 h in either estrogen-dependent (MCF-7) or -independent BC cells (SKBR3 and MDA-MB-231). At 5 h, the apoptosis rate for MCF-7 cells was 68.4% and that for MDA-MB-231 cells was 56.1%; at 48 h, that for SKBR3 was 61.6%, that for MCF-7 cells was 54.9%, and that for MDA-MB-231 (TNBC) was 43.1%. HO-AAVPA increased the S phase in MCF-7 cells and reduced the G2/M phase in MCF-7 and MDA-MB-231 cells. GPER expression decreased more than VPA in the presence of HO-AAVPA. In conclusion, the effects of HO-AAVPA on cell apoptosis could be modulated by epigenetic effects through a decrease in GPER expression.
    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
    几种致命疾病的过程和病程,比如癌症,炎症性疾病,和神经系统疾病,与HDAC8密切相关。因此,新型HDAC8抑制剂代表了可能有助于治疗这些疾病的有效治疗可能性.到目前为止,没有任何这种特殊的HDAC8抑制剂可供出售。进行此审查是为了检查最近的HDAC8抑制剂,这些抑制剂在过去十年中获得了专利。
    本综述重点介绍了HDAC8抑制剂相关专利及其治疗应用,这些专利在过去十年内已发布,可通过Patentscope和GooglePatents数据库访问。
    在过去的十年中,已经提交了一些与HDAC8抑制剂相关的专利,更具选择性,和特定的HDAC8抑制剂,旨在治疗各种医学疾病。这可能导致开发靶向HDAC8的新型治疗方法。采用理论框架和实验程序可以揭示具有增强的药代动力学特征的新HDAC8抑制剂的产生。彻底了解HDAC8抑制剂在癌症中的作用,包括HDAC8在其他疾病中的作用机制是必要的。
    UNASSIGNED: The processes and course of several fatal illnesses, such as cancer, inflammatory diseases, and neurological disorders, are closely correlated with HDAC8. Therefore, novel HDAC8 inhibitors represent effective therapeutic possibilities that may help treat these conditions. To yet, there aren\'t any such particular HDAC8 inhibitors available for sale. This review was conducted to examine recent HDAC8 inhibitors that have been patented over the last ten years.
    UNASSIGNED: This review focuses on HDAC8 inhibitor-related patents and their therapeutic applications that have been published within the last ten years and are accessible through the Patentscope and Google Patents databases.
    UNASSIGNED: A handful of HDAC8 inhibitor-related patents have been submitted over the previous ten years, more selective, and specific HDAC8 inhibitors that are intended to treat a variety of medical diseases. This could lead to the development of novel treatment approaches that target HDAC8. Employing theoretical frameworks and experimental procedures can reveal the creation of new HDAC8 inhibitors with enhanced pharmacokinetic characteristics. A thorough understanding of the role that HDAC8 inhibitors play in cancer including the mechanisms behind HDAC8 in other disorders is necessary.
    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
    长期使用酪氨酸激酶抑制剂(TKIs)后的耐药性已成为延长透明细胞肾细胞癌(ccRCC)患者生存时间的障碍。这里,应用基于CRISPR的全基因组筛选,揭示HDAC8参与降低ccRCC细胞对舒尼替尼的敏感性.机械上,HDAC8在K245位点脱乙酰ETS1,促进ETS1与HIF-2α的相互作用,增强ETS1/HIF-2α复合物的转录活性。然而,抑制HDAC8对致敏TKI的抗肿瘤作用不是很理想。随后,HDAC8的抑制增加了NEK1的表达,并在T241位点上调NEK1磷酸化ETS1,以通过在ETS1-K245位点的乙酰化阻碍促进ETS1和HIF-2α之间的相互作用。此外,还发现TKI处理通过抑制ccRCC细胞中STAT3磷酸化来增加HDAC8的表达。这两个发现强调了ccRCC中对TKIs和HDAC8抑制剂的获得性抗性的潜在机制。最后,合成了HDAC8-in-PROTACs,通过降解HDAC8并克服ccRCC对TKIs的抗性来优化HDAC8抑制剂的作用。总的来说,结果显示HDAC8是ccRCC靶向治疗耐药的潜在候选药物.
    Drug resistance after long-term use of Tyrosine kinase inhibitors (TKIs) has become an obstacle for prolonging the survival time of patients with clear cell renal cell carcinoma (ccRCC). Here, genome-wide CRISPR-based screening to reveal that HDAC8 is involved in decreasing the sensitivity of ccRCC cells to sunitinib is applied. Mechanically, HDAC8 deacetylated ETS1 at the K245 site to promote the interaction between ETS1 and HIF-2α and enhance the transcriptional activity of the ETS1/HIF-2α complex. However, the antitumor effect of inhibiting HDAC8 on sensitized TKI is not very satisfactory. Subsequently, inhibition of HDAC8 increased the expression of NEK1, and up-regulated NEK1 phosphorylated ETS1 at the T241 site to promote the interaction between ETS1 and HIF-2α by impeded acetylation at ETS1-K245 site is showed. Moreover, TKI treatment increased the expression of HDAC8 by inhibiting STAT3 phosphorylation in ccRCC cells is also found. These 2 findings highlight a potential mechanism of acquired resistance to TKIs and HDAC8 inhibitors in ccRCC. Finally, HDAC8-in-PROTACs to optimize the effects of HDAC8 inhibitors through degrading HDAC8 and overcoming the resistance of ccRCC to TKIs are synthesized. Collectively, the results revealed HDAC8 as a potential therapeutic candidate for resistance to ccRCC-targeted therapies.
    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
    HDAC8,I类HDAC的成员,通过使粘附素亚基SMC3脱乙酰在细胞周期调节中起关键作用。虽然细胞周期蛋白和CDK是公认的细胞周期调节剂,我们对其他监管机构的了解仍然有限。在这里,我们揭示了HDAC8中K202的乙酰化作用作为对应激的关键细胞周期调节剂。主要由Tip60催化的HDAC8中的K202乙酰化限制了HDAC8的活性,导致增加的SMC3乙酰化和细胞周期阻滞。此外,表达模拟K202乙酰化的HDAC8突变形式的细胞在基因表达中显示出显著的改变,可能与3D基因组结构的变化有关,包括增强的染色单体环相互作用。K202乙酰化通过破坏细胞周期相关基因的表达和姐妹染色单体的内聚力来损害细胞周期进程,导致G2/M阶段停滞。这些发现表明HDAC8作为细胞周期调节剂的可逆乙酰化,扩大我们对应激反应细胞周期动力学的理解。
    HDAC8, a member of class I HDACs, plays a pivotal role in cell cycle regulation by deacetylating the cohesin subunit SMC3. While cyclins and CDKs are well-established cell cycle regulators, our knowledge of other regulators remains limited. Here we reveal the acetylation of K202 in HDAC8 as a key cell cycle regulator responsive to stress. K202 acetylation in HDAC8, primarily catalyzed by Tip60, restricts HDAC8 activity, leading to increased SMC3 acetylation and cell cycle arrest. Furthermore, cells expressing the mutant form of HDAC8 mimicking K202 acetylation display significant alterations in gene expression, potentially linked to changes in 3D genome structure, including enhanced chromatid loop interactions. K202 acetylation impairs cell cycle progression by disrupting the expression of cell cycle-related genes and sister chromatid cohesion, resulting in G2/M phase arrest. These findings indicate the reversible acetylation of HDAC8 as a cell cycle regulator, expanding our understanding of stress-responsive cell cycle dynamics.
    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
    双特异性酪氨酸磷酸化调节激酶2(DYRK2)和组蛋白脱乙酰酶8(HDAC8)已显示与几种癌症的发展有关。这里,我们通过联合虚拟筛选方案鉴定了双靶点DYRK2/HDAC8抑制剂(DYC-1).DYC-1表现出对DYRK2(IC50=5.27±0.13nM)和HDAC8(IC50=8.06±0.47nM)的纳摩尔抑制活性。分子动力学模拟显示DYC-1与DYRK2和HDAC8具有正的结合稳定性。重要的是,细胞毒性实验表明DYC-1对人肝癌具有优越的抗增殖活性,尤其是SK-HEP-1细胞,对正常肝细胞无明显抑制作用。此外,DYC-1对SK-HEP-1异种移植瘤的生长表现出较强的抑制作用,且无明显副作用。这些数据表明DYC-1是用于治疗肝细胞癌的高效低毒性抗肿瘤剂。
    Dual-specificity tyrosine phosphorylation-regulated kinase 2 (DYRK2) and histone deacetylase 8 (HDAC8) have been shown to be associated with the development of several cancers. Here, we identified a dual-target DYRK2/HDAC8 inhibitor (DYC-1) through a combined virtual screening protocol. DYC-1 exhibited nanomolar inhibitory activity against both DYRK2 (IC50 = 5.27 ± 0.13 nM) and HDAC8 (IC50 = 8.06 ± 0.47 nM). Molecular dynamics simulations showed that DYC-1 had positive binding stability with DYRK2 and HDAC8. Importantly, the cytotoxicity assay indicated that DYC-1 exhibited superior antiproliferative activity against human liver cancer, especially SK-HEP-1 cells, and had no significant inhibition on normal liver cells. Moreover, DYC-1 showed a strong inhibitory effect on the growth of SK-HEP-1 xenograft tumors with no significant side effects. These data suggest that DYC-1 is a high-efficacy and low-toxic antitumor agent for the treatment of hepatocellular carcinoma.
    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
    CorneliadeLange综合征(CdLS)是一种罕见的先天性多系统发育障碍。临床表现变化很大,但是经典的表型,以独特的颅面特征为特征,出生前和出生后生长迟缓,四肢减少缺陷,多毛症和智力障碍可以与非经典表型区分开来,通常更温和,更难诊断。此外,临床特征与其他神经发育障碍的特征重叠,因此,使用共识临床标准和人工智能工具可能有助于确认诊断。NIPBL的致病变异,编码一种与粘附蛋白复合物相关的蛋白质,已经在超过60%的患者中被确认,与该复合物相关的其他基因中的致病变异还有15%:SMC1A,SMC3、RAD21和HDAC8。大规模测序的技术进步允许描述其他基因(BRD4,ANKRD11,MAU2),但15%的个体缺乏分子诊断以及该综合征的实质性临床异质性提示可能涉及其他基因和机制。虽然没有治愈性的治疗,儿科医生应该注意对症/姑息治疗.经典SCdL的主要并发症是胃食管反流(GER),应该及早治疗。
    Cornelia de Lange syndrome (CdLS) is a rare congenital developmental disorder with multisystemic involvement. The clinical presentation is highly variable, but the classic phenotype, characterized by distinctive craniofacial features, pre- and postnatal growth retardation, extremity reduction defects, hirsutism and intellectual disability can be distinguished from the nonclassic phenotype, which is generally milder and more difficult to diagnose. In addition, the clinical features overlap with those of other neurodevelopmental disorders, so the use of consensus clinical criteria and artificial intelligence tools may be helpful in confirming the diagnosis. Pathogenic variants in NIPBL, which encodes a protein related to the cohesin complex, have been identified in more than 60% of patients, and pathogenic variants in other genes related to this complex in another 15%: SMC1A, SMC3, RAD21, and HDAC8. Technical advances in large-scale sequencing have allowed the description of additional genes (BRD4, ANKRD11, MAU2), but the lack of molecular diagnosis in 15% of individuals and the substantial clinical heterogeneity of the syndrome suggest that other genes and mechanisms may be involved. Although there is no curative treatment, there are symptomatic/palliative treatments that paediatricians should be aware of. The main medical complication in classic SCdL is gastro-esophageal reflux (GER), which should be treated early.
    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
    宫颈癌,全球女性死亡的主要原因,表现出不同的分子畸变影响基因表达和信号通路。表观遗传因素,包括组蛋白脱乙酰酶(HDACs),如HDAC8和HDAC6,以及microRNAs(miRNAs),在宫颈癌进展中起关键作用。最近的研究揭示了miRNA作为HDAC的潜在调节因子,提供了一个有希望的治疗途径。本研究采用计算机内miRNA预测,qRT-PCR共表达研究,和双荧光素酶报告基因分析,以鉴定HeLa中控制HDAC8和HDAC6的miRNA,宫颈癌细胞。结果确定miR-497-3p和miR-324-3p分别是HDAC8和HDAC6的新型负调节因子。功能测定表明,miR-497-3p在HeLa细胞中的过表达抑制HDAC8,导致下游靶标p53和α-微管蛋白的乙酰化增加。同样,miR-324-3p过表达抑制HDAC6mRNA和蛋白表达,增强Hsp90和α-微管蛋白的乙酰化。值得注意的是,通过与细胞活力降低相关的miRNA过表达抑制HDAC8,上皮-间质转化(EMT)减少,并增加了HeLa细胞中微管束的形成。总之,miR-497-3p和miR-324-3p分别成为HDAC8和HDAC6的新型负调节因子。具有潜在的治疗意义。这些miRNA在宫颈癌细胞中的表达升高有望抑制转移,为宫颈恶性肿瘤的干预提供了有针对性的方法。
    Cervical cancer, a leading global cause of female mortality, exhibits diverse molecular aberrations influencing gene expression and signaling pathways. Epigenetic factors, including histone deacetylases (HDACs) such as HDAC8 and HDAC6, along with microRNAs (miRNAs), play pivotal roles in cervical cancer progression. Recent investigations have unveiled miRNAs as potential regulators of HDACs, offering a promising therapeutic avenue. This study employed in-silico miRNA prediction, qRT-PCR co-expression studies, and Dual-Luciferase reporter assays to identify miRNAs governing HDAC8 and HDAC6 in HeLa, cervical cancer cells. Results pinpointed miR-497-3p and miR-324-3p as novel negative regulators of HDAC8 and HDAC6, respectively. Functional assays demonstrated that miR-497-3p overexpression in HeLa cells suppressed HDAC8, leading to increased acetylation of downstream targets p53 and α-tubulin. Similarly, miR-324-3p overexpression inhibited HDAC6 mRNA and protein expression, enhancing acetylation of Hsp90 and α-tubulin. Notably, inhibiting HDAC8 via miRNA overexpression correlated with reduced cell viability, diminished epithelial-to-mesenchymal transition (EMT), and increased microtubule bundle formation in HeLa cells. In conclusion, miR-497-3p and miR-324-3p emerge as novel negative regulators of HDAC8 and HDAC6, respectively, with potential therapeutic implications. Elevated expression of these miRNAs in cervical cancer cells holds promise for inhibiting metastasis, offering a targeted approach for intervention in cervical malignancy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号