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
    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
    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)

  • 文章类型: Journal Article
    抗癌靶标hRpn13是蛋白酶体底物受体。然而,hRpn13靶向分子不会损害其与蛋白酶体或泛素的相互作用,提示其他关键细胞活动。我们发现hRpn13缺失导致相关的蛋白质组和转录组变化,在骨髓瘤细胞中对细胞骨架和免疫反应蛋白以及骨髓特异性精氨酸脱亚胺酶PADI4具有明显作用。此外,针对hRpn13的PROTAC共消耗PADI4、组蛋白脱乙酰酶HDAC8和DNA甲基转移酶MGMT。PADI4结合并瓜氨酸化hRpn13和蛋白酶体,来自PADI4抑制的骨髓瘤细胞的蛋白酶体表现出降低的肽酶活性。当关闭蛋白酶体时,hRpn13可以结合HDAC8,并且这种相互作用抑制HDAC8活性。进一步将hRpn13与转录联系起来,它的丢失会降低蛋白酶体通过切割其前体蛋白而产生的核因子κB(NF-κB)转录因子p50。NF-κB抑制耗尽hRpn13相互作用因子PADI4和HDAC8。总之,我们发现hRpn13在蛋白质降解和表达中双重作用,反过来,调节因细胞类型而异。
    The anti-cancer target hRpn13 is a proteasome substrate receptor. However, hRpn13-targeting molecules do not impair its interaction with proteasomes or ubiquitin, suggesting other critical cellular activities. We find that hRpn13 depletion causes correlated proteomic and transcriptomic changes, with pronounced effects in myeloma cells for cytoskeletal and immune response proteins and bone-marrow-specific arginine deiminase PADI4. Moreover, a PROTAC against hRpn13 co-depletes PADI4, histone deacetylase HDAC8, and DNA methyltransferase MGMT. PADI4 binds and citrullinates hRpn13 and proteasomes, and proteasomes from PADI4-inhibited myeloma cells exhibit reduced peptidase activity. When off proteasomes, hRpn13 can bind HDAC8, and this interaction inhibits HDAC8 activity. Further linking hRpn13 to transcription, its loss reduces nuclear factor κB (NF-κB) transcription factor p50, which proteasomes generate by cleaving its precursor protein. NF-κB inhibition depletes hRpn13 interactors PADI4 and HDAC8. Altogether, we find that hRpn13 acts dually in protein degradation and expression and that proteasome constituency and, in turn, regulation varies by cell type.
    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
    涎腺肿瘤(SGT)是一种罕见且复杂的肿瘤,其特征是组织学和临床行为异质性以及对全身治疗的抵抗力。目前正在阐明肿瘤的病因,并且已经提出遗传和表观遗传变化的相互作用有助于肿瘤的发展。在这项工作中,我们研究了可能改变基因表达并参与SGT肿瘤发病机制的表观遗传调节因子和组蛋白修饰因子.我们对94个ACC组织的公开RNA-seq数据集进行了详细的生物信息学分析,并补充了临床数据和相应的对照,并生成了染色质和组蛋白修饰因子的蛋白质-蛋白质相互作用(PPI)网络。TP53和组蛋白修饰酶SUV39H1,EZH2,PRMT1,HDAC8和KDM5B的显着上调,随着DNA甲基转移酶DNMT3A和泛素连接酶UHRF1mRNA水平的上调,以及赖氨酸乙酰转移酶KAT2B水平的下调,在ACC组织中检测到。在SGT组织中进一步验证了p53,SUV39H1,EZH2和HDAC8的蛋白表达,以及它们分别对抑制组蛋白标记H3K9me3和H3K27me3的功能沉积。总的来说,这项研究是第一个检测到影响唾液腺肿瘤细胞染色质结构和组蛋白修饰的相互作用蛋白网络,进一步提供对SGT分子谱的机械见解,赋予改变的基因表达程序。
    Salivary gland tumors (SGTs) are rare and complex neoplasms characterized by heterogenous histology and clinical behavior as well as resistance to systemic therapy. Tumor etiology is currently under elucidation and an interplay of genetic and epigenetic changes has been proposed to contribute to tumor development. In this work, we investigated epigenetic regulators and histone-modifying factors that may alter gene expression and participate in the pathogenesis of SGT neoplasms. We performed a detailed bioinformatic analysis on a publicly available RNA-seq dataset of 94 ACC tissues supplemented with clinical data and respective controls and generated a protein-protein interaction (PPI) network of chromatin and histone modification factors. A significant upregulation of TP53 and histone-modifying enzymes SUV39H1, EZH2, PRMT1, HDAC8, and KDM5B, along with the upregulation of DNA methyltransferase DNMT3A and ubiquitin ligase UHRF1 mRNA levels, as well as a downregulation of lysine acetyltransferase KAT2B levels, were detected in ACC tissues. The protein expression of p53, SUV39H1, EZH2, and HDAC8 was further validated in SGT tissues along with their functional deposition of the repressive histone marks H3K9me3 and H3K27me3, respectively. Overall, this study is the first to detect a network of interacting proteins affecting chromatin structure and histone modifications in salivary gland tumor cells, further providing mechanistic insights in the molecular profile of SGTs that confer to altered gene expression programs.
    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
    CorneliadeLange综合征(CdLS)是一种罕见的,主要遗传性多系统发育障碍,其特征是生长和发育迟缓的高度可变表现,上肢受累,多毛症,心脏,胃肠,颅面,和其他系统特征。编码cohesin复杂结构亚基和调节蛋白的基因中的致病变异(NIPBL,SMC1A,SMC3,HDAC8和RAD21)是CdLS的主要致病因素。已发现编码这五种蛋白质的基因中的杂合或半合子变体与CdLS有关,NIPBL变异占大多数(>60%)病例,也是迄今为止发现的唯一一种在突变时会导致严重或经典形式的CdLS的基因。除NIPBL以外的粘附蛋白基因中的致病变体倾向于导致不太严重的表型。其他基因的因果变异,例如ANKRD11、EP300、AFF4、TAF1和BRD4可引起CdLS样表型。这些基因的共同作用,和其他人,发挥发育转录控制的关键调节因子已导致它们引起的疾病被称为转录调节障碍(或“DTR”)。这里,我们报告了一个全面的分子分析的结果,在一个队列的716个先证者与典型和非典型的CdLS,以描绘致病变异的遗传贡献在cohesin复杂基因以及新的候选基因。基因型-表型相关性,以及基因组测序在理解该种群突变景观中的实用性。
    Cornelia de Lange Syndrome (CdLS) is a rare, dominantly inherited multisystem developmental disorder characterized by highly variable manifestations of growth and developmental delays, upper limb involvement, hypertrichosis, cardiac, gastrointestinal, craniofacial, and other systemic features. Pathogenic variants in genes encoding cohesin complex structural subunits and regulatory proteins (NIPBL, SMC1A, SMC3, HDAC8, and RAD21) are the major pathogenic contributors to CdLS. Heterozygous or hemizygous variants in the genes encoding these five proteins have been found to be contributory to CdLS, with variants in NIPBL accounting for the majority (>60%) of cases, and the only gene identified to date that results in the severe or classic form of CdLS when mutated. Pathogenic variants in cohesin genes other than NIPBL tend to result in a less severe phenotype. Causative variants in additional genes, such as ANKRD11, EP300, AFF4, TAF1, and BRD4, can cause a CdLS-like phenotype. The common role that these genes, and others, play as critical regulators of developmental transcriptional control has led to the conditions they cause being referred to as disorders of transcriptional regulation (or \"DTRs\"). Here, we report the results of a comprehensive molecular analysis in a cohort of 716 probands with typical and atypical CdLS in order to delineate the genetic contribution of causative variants in cohesin complex genes as well as novel candidate genes, genotype-phenotype correlations, and the utility of genome sequencing in understanding the mutational landscape in this population.
    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
    口腔粘膜下纤维化(OSF)是一种与槟榔咀嚼有关的癌前疾病。该研究的目的是确定组蛋白脱乙酰酶(HDAC)8的作用,在纤维化条件的调节中,为OSF提供治疗潜力。
    首先,我们检测了HDAC8在纤维化颊粘膜成纤维细胞(fBMF)和OSF组织中的表达。在具有HDAC8敲低的fBMF中进行肌成纤维细胞和TGF-β信号传导的标记。此外,上皮间质转化(EMT)标志物,还在用sh-HDAC8转染的fBMF中检查了胶原蛋白凝胶的收缩和迁移能力。HDAC8抑制剂用于分析fBMF中胶原蛋白凝胶收缩和伤口愈合能力。
    我们观察到fBMF中HDAC8的mRNA表达显著增加。与正常组织相比,OSF中HDAC8蛋白水平上调。接下来,HDAC8的mRNA和卵白表达显著下降,在感染sh-HDAC8的fBMF中伴随α-SMA和COL1A1的下调。为了确定HDAC8在OSF纤维化中的关键作用,结果表明,在HDAC8敲低的fBMF中,TGF-β的分泌以及EMT转录因子SNAIL和p-Smad的表达显着降低。我们进一步证明,在sh-HDAC8转染的fBMF中,胶原蛋白凝胶的收缩和迁移能力显着降低。最后,结果表明,HDAC8抑制剂治疗可显著降低fBMF的胶原凝胶收缩和伤口愈合能力。
    我们得出结论,HDAC8的下调减轻了OSF中肌成纤维细胞和TGF-β/Smad信号通路的活性。
    UNASSIGNED: Oral submucous fibrosis (OSF) is a premalignant disorder that is associated with betel nut chewing. The purpose of the study was to establish the role of histone deacetylase (HDAC) 8, one of histone deacetylases, in the regulation of fibrotic conditions to provide a therapeutic potential for OSF.
    UNASSIGNED: First, we examined the expression of HDAC8 in fibrotic buccal mucosal fibroblasts (fBMFs) and OSF tissues. Markers of myofibroblasts and TGF-β signaling were conducted in fBMFs with HDAC8 knockdown were examined. Furthermore, epithelial-mesenchymal transition (EMT) markers, collagen gel contraction and migration ability were also examined in fBMFs transfected with sh-HDAC8. HDAC8 inhibitor was used to analyze the collagen gel contraction and wound healing ability in fBMFs.
    UNASSIGNED: We observed the mRNA expression of HDAC8 was significantly increased in fBMFs. Compared to normal tissues, the protein level of HDAC8 was upregulated in OSF. Next, mRNA and protein expression of HDAC8 was significantly decreased, accompanying downregulation of α-SMA and COL1A1 in fBMFs infected with sh-HDAC8. To determine the critical role of HDAC8 in OSF fibrogenesis, results revealed that TGF-β secretion and the expression of EMT transcription factor SNAIL and p-Smad were significantly decreased in HDAC8-knockdown fBMFs. We further demonstrated that collagen gel contraction and migration ability were significantly decreased in fBMFs transfected with sh-HDAC8. Last, results revealed that significantly reduced collagen gel contraction and wound healing ability in fBMFs with HDAC8 inhibitor treatment.
    UNASSIGNED: We concluded that downregulation of HDAC8 alleviated the activities of myofibroblasts and TGF-β/Smad signaling pathway in OSF.
    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
    噬菌体辅助,活性位点定向配体进化(PADLE)是一种最近开发的技术,它使用琥珀密码子编码的非规范氨基酸(ncAA)作为锚将噬菌体展示的肽定向到靶标,以增强配体鉴定过程。2-氨基-8-氧代癸酸(Aoda)是大环肽天然产物Apicidin中的含酮ncAA残基,它是Zn2依赖性组蛋白脱乙酰酶(HDAC)的泛抑制剂。它的酮用作锚定点以协调HDAC中的催化锌离子。使用先前进化的Nβ-乙酰基-赖氨酰-tRNA合成酶与tRNAPyl的组合,我们表明,通过琥珀色抑制,Aoda被有效地掺入大肠杆菌的蛋白质中。通过在编码Aoda的大肠杆菌中繁殖琥珀密码子专性噬菌粒文库,我们产生了含有Aoda的噬菌体展示肽库。使用该文库针对HDAC8进行PADLE揭示了7聚体肽GH8P01F1,其具有与鉴定的HDAC8底物中的现有肽序列匹配的Aoda侧翼氨基酸残基。将GH8P01F1中的Aoda转换为更能螯合Zn2的ncAAS-2-氨基-8-羟基氨基-8-氧辛酸(Asuha)导致了一种非常有效的化合物GH8HA01,其HDAC8抑制Ki值为0.67nM。GH8HA01及其5-mer截短类似物Ac-GH8HA01Δ1Δ1Δ7具有0.31nM的HDAC8抑制Ki值,是已开发的两种最有效的HDAC8抑制剂。此外,与其他测试的HDAC相比,两者都对HDAC8具有高度选择性,证明了使用PADLE鉴定具有高度有效和选择性的配体的靶标的巨大潜力,这些配体在同源物中具有保守的活性位点。
    Phage-assisted, active site-directed ligand evolution (PADLE) is a recently developed technique that uses an amber codon-encoded noncanonical amino acid (ncAA) as an anchor to direct phage-displayed peptides to a target for an enhanced ligand identification process. 2-Amino-8-oxodecanoic acid (Aoda) is a ketone-containing ncAA residue in the macrocyclic peptide natural product apicidin that is a pan-inhibitor of Zn2+ -dependent histone deacetylases (HDACs). Its ketone serves as an anchoring point to coordinate the catalytic zinc ion in HDACs. Using a previously evolved N𝜀 -acetyl-lysyl-tRNA synthetase in combination with tRNAPyl , we showed that Aoda was efficiently incorporated into proteins in Escherichia coli by amber suppression. By propagating an amber codon-obligate phagemid library in E. coli encoding Aoda, we generated an Aoda-containing phage-displayed peptide library. Using this library to conduct PADLE against HDAC8 revealed a 7-mer peptide GH8P01F1 with Aoda-flanking amino acid residues that matched existing peptide sequences in identified HDAC8 substrates. Switching Aoda in GH8P01F1 to a more Zn2+ -chelating ncAA S-2-amino-8-hydroxyamino-8-oxooctanoic acid (Asuha) led to an extremely potent compound GH8HA01, which has an HDAC8-inhibition Ki value of 0.67 nM. GH8HA01 and its 5-mer truncation analogue Ac-GH8HA01Δ1Δ7 that has an HDAC8-inhibition Ki value of 0.31 nM are two of the most potent HDAC8 inhibitors that have been developed. Furthermore, both are highly selective against HDAC8 compared with other HDACs tested, demonstrating the great potential of using PADLE to identify highly potent and selective ligands for targets with conserved active sites among homologues.
    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
    Histone deacetylases (HDACs) catalyze the removal of Ɛ-acetyl-lysine residues of histones via hydrolysis. Removal of acetyl groups results in condensation of chromatin structure and alteration of gene expression by repression. HDACs are considered targets for the treatment of cancer due to their role in regulating transcription. HDAC8 inhibition may be an important anti-proliferative factor for histone deacetylase inhibitors on cancer cells and may give rise to the progression of apoptosis. HDAC8 activity was analyzed with various peptides where the target lysine is modified with medium-chain fatty acyl group. Kinetic data were determined for each p53 peptide substrate. The results suggest that there was HDAC8 deacetylase activity on peptide substrate as well as deacylase activity with acylated peptide substrate variants. HDAC8 inhibition by hexanoic and decanoic acid was also examined. The Ki for hexanoic and decanoic acid were determined to be 2.35 ± 0.341 and 4.48 ± 0.221 mM, respectively.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号