HDAC8

HDAC8
  • 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
    传统的亲和方法在体内捕获酶-翻译后修饰(PTM)底物的瞬时相互作用是一个挑战。在这里,我们提出了一种称为基于邻近标记的正交陷阱方法(ProLORT)的策略,依靠APEX2催化的邻近标记和正交陷阱管道以及定量蛋白质组学直接研究活细胞中酶-PTM底物的瞬时相互作用。作为概念的证明,ProLORT允许对已知的HDAC8基板进行稳健评估,组蛋白H3K9ac.通过利用这种方法,我们鉴定出大量HDAC8靶向的推定乙酰化蛋白,并进一步证实CTTN是体内真正的底物.接下来,我们证明HDAC8通过CTTN在赖氨酸144处的脱乙酰作用促进细胞运动,从而减弱其与F-肌动蛋白的相互作用,扩大HDAC8的基本监管机制。我们开发了一种通用策略来描述PTM介导的瞬时酶-底物相互作用,为识别活细胞中酶调节的时空PTM网络提供了强大的工具。
    It is a challenge for the traditional affinity methods to capture transient interactions of enzyme-post-translational modification (PTM) substrates in vivo. Herein we presented a strategy termed proximity labeling-based orthogonal trap approach (ProLORT), relying upon APEX2-catalysed proximity labeling and an orthogonal trap pipeline as well as quantitative proteomics to directly investigate the transient interactome of enzyme-PTM substrates in living cells. As a proof of concept, ProLORT allows for robust evaluation of a known HDAC8 substrate, histone H3K9ac. By leveraging this approach, we identified numerous of putative acetylated proteins targeted by HDAC8, and further confirmed CTTN as a bona fide substrate in vivo. Next, we demonstrated that HDAC8 facilitates cell motility via deacetylation of CTTN at lysine 144 that attenuates its interaction with F-actin, expanding the underlying regulatory mechanisms of HDAC8. We developed a general strategy to profile the transient enzyme-substrate interactions mediated by PTMs, providing a powerful tool for identifying the spatiotemporal PTM-network regulated by enzymes in living cells.
    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
    在这项研究中,我们从一名年轻男性患者中分离出外周血单个核细胞,该患者携带组蛋白脱乙酰酶8(HDAC8)突变,患有经临床和基因诊断证实的CorneliadeLange综合征.通过非整合性方法建立诱导多能干细胞(iPSCs),使用携带OCT4、SOX2、KLF4、BCL-XL和C-MYC的质粒。已建立的iPSCs呈现典型的多能细胞形态,并在mRNA和蛋白质水平表达多能干细胞标志物。iPSC在体外也显示出分化能力,和正常的核型。此外,已建立的iPSC仍携带在供体组织中观察到的HDAC8突变.
    In this study, peripheral blood mononuclear cells were isolated from a young male patient bearing a histone deacetyl-lase 8 (HDAC8) mutation and suffering from Cornelia de Lange Syndrome verified by clinical and genetic diagnosis. Induced pluripotent stem cells (iPSCs) were established by a non-integrative method, using plasmids carrying OCT4, SOX2, KLF4, BCL-XL and C-MYC. The established iPSCs presented typical pluripotent cells morphology, and expressed pluripotent stem cell markers at the mRNA and protein level. The iPSCs also showed differentiative capacity in vitro, and a normal karyotype. In addition, the established iPSCs still carried the HDAC8 mutation observed in the donor tissue.
    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
    在这里,通过将HDAC抑制剂的关键药效团引入2,6-二芳基吡啶和2'-芳基查耳酮的骨架中,合成了两个系列的HDAC/微管蛋白双重抑制剂。其中,2,6-基于二芳基吡啶的异羟肟酸10a对HDAC8表现出良好的抑制活性(IC50=117nM),相对于HDAC1和HDAC6分别具有50倍和42倍的高选择性。同时,10a有效地破坏了微管蛋白聚合,并对BE-(2)-C细胞系表现出有效的抗增殖活性,IC50值为17nM。机制研究表明,10a阻断细胞周期,诱导细胞凋亡和抑制集落形成。此外,10a具有良好的理化性质和代谢稳定性。重要的是,10a在人神经母细胞瘤异种移植小鼠模型中表现出比临床HDAC抑制剂和微管蛋白抑制剂更好的抗肿瘤作用,无论是单独使用还是组合使用。这些结果突出了HDAC8/微管蛋白双重抑制剂10a作为杰出的抗肿瘤剂的优点。
    Herein, two series of HDAC/tubulin dual inhibitors via introducing the key pharmacophore of HDAC inhibitor into the skeletons of 2,6-diarylpyridine and 2\'-arylchalcone were synthesized. Among them, 2,6-diarylpyridine-based hydroxamic acid 10a exhibited good inhibitory activity against HDAC8 (IC50 = 117 nM) with 50-fold and 42-fold high selectivity relative to HDAC1 and HDAC6, respectively. Meanwhile, 10a disrupted tubulin polymerization effectively and exhibited potent antiproliferative activity against BE-(2)-C cell line, with IC50 value of 17 nM. Mechanism studies revealed that 10a blocked cell cycle, induced cellular apoptosis and suppressed colony formation. Moreover, 10a possessed good physicochemical properties and metabolic stability. Importantly, 10a exhibited better antitumor effects in human neuroblastoma xenograft mice model than those of clinical HDAC inhibitor and tubulin inhibitor, whether used alone or in combination. These results highlighted the advantages of the HDAC8/tubulin dual inhibitor 10a as an outstanding antitumor agent.
    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8(HDAC8)的过表达与多种疾病如癌症相关。因此,可以调节HDAC8水平的化合物对这些疾病具有治疗潜力。基于蛋白水解靶向嵌合体(PROTAC)策略,我们通过将HDAC6/8双重抑制剂与泊马度胺(一种小脑配体)连接,设计并合成了一系列HDAC8降解剂。其中,化合物ZQ-23显示HDAC8的显著和选择性降解,DC50为147nM,Dmax为93%,对HDAC1和HDAC3无影响。有趣的是,我们发现,在用ZQ-23处理后2h开始降解靶蛋白,在10h达到最大降解效果。HDAC8水平在24h内部分恢复。此外,ZQ-23在所有浓度下对HDAC1和HDAC3均无降解作用,但可以剂量依赖性地增加乙酰化SMC-3(HDAC8底物)的水平。机制研究表明,ZQ-23通过泛素-蛋白酶途径降解HDAC8,而不是溶酶体系统。总的来说,这些结果表明,ZQ-23代表了一种新型的基于PROTAC的HDAC8降解剂,值得进一步研究。
    Overexpression of histone deacetylase 8 (HDAC8) is associated with various diseases such as cancer. Thus, compounds that can modulate HDAC8 levels have therapeutic potential for these diseases. Based on the proteolysis targeting chimera (PROTAC) strategy, we designed and synthesized a series of HDAC8 degraders by tethering an HDAC6/8 dual inhibitor with pomalidomide (a cereblon ligand). Among them, compound ZQ-23 exhibited significant and selective degradation of HDAC8 with DC50 of 147 nM and Dmax of 93%, and exhibited no effects on HDAC1 and HDAC3. Interestingly, we found that the degradation of target protein started at ∼2 h after treatment with ZQ-23 and the maximal degradation effect was achieved at 10 h. The HDAC8 level was partially recovered within 24 h. In addition, ZQ-23 had no degrading effects on HDAC1 and HDAC3 at all concentrations, but could dose-dependently increase the levels of acetylated SMC-3 (HDAC8 substrate). Mechanism study demonstrated that ZQ-23 degraded HDAC8 through the ubiquitin-protease pathway, rather than lysosome system. Collectively, these results suggest that ZQ-23 represents a novel PROTAC-based HDAC8 degrader worthy of further investigation.
    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
    OBJECTIVE: Histone deacetylase 8 (HDAC8) is one of the class I HDAC family proteins, which participates in the neuronal disorders, parasitic/viral infections, tumorigenesis and many other biological processes. However, its potential function during female germ cell development has not yet been fully understood.
    METHODS: HDAC8-targeting siRNA was microinjected into GV oocytes to deplete HDAC8. PCI-34051 was used to inhibit the enzyme activity of HDAC8. Immunostaining, immunoblotting and fluorescence intensity quantification were applied to assess the effects of HDAC8 depletion or inhibition on the oocyte meiotic maturation, spindle/chromosome structure, γ-tubulin dynamics and acetylation level of α-tubulin.
    RESULTS: We observed that HDAC8 was localized in the nucleus at GV stage and then translocated to the spindle apparatus from GVBD to M II stages in porcine oocytes. Depletion of HDAC8 led to the oocyte meiotic failure by showing the reduced polar body extrusion rate. In addition, depletion of HDAC8 resulted in aberrant spindle morphologies and misaligned chromosomes due to the defective recruitment of γ-tubulin to the spindle poles. Notably, these meiotic defects were photocopied by inhibition of HDAC8 activity using its specific inhibitor PCI-34051. However, inhibition of HDAC8 did not affect microtubule stability as assessed by the acetylation level of α-tubulin.
    CONCLUSIONS: Collectively, our findings demonstrate that HDAC8 acts as a regulator of spindle assembly during porcine oocyte meiotic maturation.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    化疗耐药是急性髓系白血病(AML)治疗的主要挑战之一。我们发现组蛋白去乙酰化酶8(HDAC8)在柔红霉素(DNR)抗性AML细胞中的表达增加,而通过其特异性siRNA或抑制剂靶向抑制HDAC8可以恢复DNR治疗的敏感性。Further,靶向抑制HDAC8可以抑制白细胞介素6(IL-6)和IL-8的表达。而重组IL-6(rIL-6)和rIL-8可以逆转AML细胞的si-HDAC8-reoredDNR敏感性。机制研究表明,HDAC8增加了NF-κB复合物的关键成分之一p65的表达,促进IL-6和IL-8的表达。这可能是由于HDAC8可以直接与p65的启动子结合以增加其转录和表达。总的来说,我们的数据表明,HDAC8通过调节IL-6和IL-8促进人AML细胞的DNR抵抗.
    The chemoresistance is one of the major challenges for acute myeloid leukemia (AML) treatment. We found that the expression of histone deacetylase 8 (HDAC8) was increased in daunorubicin (DNR) resistant AML cells, while targeted inhibition of HDAC8 by its specific siRNA or inhibitor can restore sensitivity of DNR treatment . Further, targeted inhibition of HDAC8 can suppress expression of interleukin 6 (IL-6) and IL-8. While recombinant IL-6 (rIL-6) and rIL-8 can reverse si-HDAC8-resored DNR sensitivity of AML cells. Mechanistical study revealed that HDAC8 increased the expression of p65, one of key components of NF-κB complex, to promote the expression of IL-6 and IL-8. It might be due to that HDAC8 can directly bind with the promoter of p65 to increase its transcription and expression. Collectively, our data suggested that HDAC8 promotes DNR resistance of human AML cells via regulation of IL-6 and IL-8.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

公众号