H3K18ac

H3K18ac
  • 文章类型: Journal Article
    背景:口腔癌的发生是复杂的,受遗传和表观遗传变化的影响。改变的组蛋白修饰是在癌症发展和进展中起作用的表观遗传事件。已显示组蛋白的不同修饰模式会影响选定癌症的患者预后。本研究旨在评估口腔上皮异型增生(OED)和口腔鳞状细胞癌(OSCC)中组蛋白H3修饰的特征与临床病理特征的关系。
    方法:将100例患者分为4组:低度OED,高级OED,OSCC,和正常口腔粘膜(NOM)。3种组蛋白修饰的水平-H3K18ac,免疫组化分析H3K9me3和H3K9ac。比较了它们的表达谱,并将其与预后相关的临床和病理特征相关联。
    结果:在OSCC中H3K18ac和H3K9me3上调,与OED和NOM相比。相比之下,H3K9ac在低品位OED中下调,但在高品位OED和OSCC中升高。H3K18和H3K9的高乙酰化与晚期癌的浸润深度和高T分期显著相关。分别。
    结论:组蛋白H3乙酰化和赖氨酸残基甲基化不同程度地参与多步骤口腔癌变并影响侵袭性癌症表型。H3K9ac的作用出现在OED发展的早期,而H3K18ac和H3K9me3的增加可能对OSCC的出现至关重要。
    BACKGROUND: Oral carcinogenesis is complex and influenced by both genetic and epigenetic changes. Altered histone modification is the epigenetic event that plays a role in cancer development and progression. Distinct modification patterns of histones have been shown to affect patient prognosis in selected cancers. This study aimed to evaluate the profiles of histone H3 modification in oral epithelial dysplasia (OED) and oral squamous cell carcinoma (OSCC) in association with the clinical-pathologic characteristics.
    METHODS: One hundred patients were divided into 4 groups: low-grade OED, high-grade OED, OSCC, and normal oral mucosa (NOM). The levels of 3 types of histone modification-the H3K18ac, H3K9me3, and H3K9ac-were analysed immunohistochemically. Their expression profiles were compared and correlated with prognostically relevant clinical and pathologic features.
    RESULTS: The H3K18ac and H3K9me3 were upregulated in OSCC, compared with OED and NOM. In contrast, the H3K9ac was downregulated in low-grade OED but increased in high-grade OED and OSCC. The hyperacetylations of H3K18 and H3K9 significantly correlated with advanced cancer depth of invasion and high T stage, respectively.
    CONCLUSIONS: Histone H3 acetylation and methylation at lysine residues are differentially involved in the multistep oral carcinogenesis and impact aggressive cancer phenotypes. The effect of H3K9ac appears early in OED development, whilst the increased H3K18ac and H3K9me3 may be vital in the emergence of OSCC.
    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
    目的:临床上推荐对乙酰氨基酚(APAP)作为孕妇的止痛药和解热药。然而,越来越多的实验室证据表明,在怀孕期间使用APAP可能会改变胎儿发育。由于胎儿期是早期卵子发生的易感窗口,我们旨在评估母体给予APAP对胎儿卵母细胞的潜在影响.
    结果:母亲给药和胎儿卵巢培养显示APAP(50和150mg/kg。bw/day)通过其代谢物NAPQI引起胎儿卵母细胞减数分裂畸变,包括减数分裂前期I(MPI)进展延迟和同源重组缺陷。与NAM或NRC共同治疗,NAD+补充剂,有效地恢复了MPI的逮捕,而SIRT7抑制剂的添加使NAD+补充剂的效果无效。此外,RNA测序显示用NAPQI处理的胎儿卵巢转录组失真。Further,雌性后代的繁殖力受到影响,表现为延迟的原始卵泡发生和青春期发作,降低卵巢激素水平,MII卵母细胞发育能力受损。
    方法:这些发现提供了第一个已知的证明,即NAPQI,从APAP的孕产妇管理转变为,扰乱胎儿卵母细胞的减数分裂过程,并进一步损害成年女性的繁殖力。伴随的NAM口服给药进一步支持NAD+补充剂对卵子发生的益处。
    结论:妊娠小鼠短期服用APAP会通过其代谢产物NAPQI引起胎儿卵母细胞减数分裂畸变,而与NAD+补充剂共同治疗可通过与SIRT7相互作用有效缓解不良反应。
    Aim: Acetaminophen (APAP) is clinically recommended as analgesic and antipyretic among pregnant women. However, accumulating laboratory evidence shows that the use of APAP during pregnancy may alter fetal development. Since fetal stage is a susceptible window for early oogenesis, we aim to assess the potential effects of maternal administration of APAP on fetal oocytes. Results: Pregnant mice at 14.5 dpc (days post-coitus) were orally administered with APAP (50 and 150mg/kg.bw/day) for 3 days; meanwhile, 14.5 dpc ovaries were collected and cultured with APAP or its metabolite N-acetyl-p-benzoquinone imine (NAPQI; 5 and 15 μM) for 3 days. It showed that APAP caused meiotic aberrations in fetal oocytes through its metabolite NAPQI, including meiotic prophase I (MPI) progression delay and homologous recombination defects. Co-treatment with nicotinamide (NAM) or nicotinamide riboside chloride (NRC), nicotinamide adenine dinucleotide (NAD+) supplements, efficiently restored the MPI arrest, whereas the addition of the inhibitor of sirtuin 7 (SIRT7) invalidated the effect of the NAD+ supplement. In addition, RNA sequencing revealed distorted transcriptomes of fetal ovaries treated with NAPQI. Furthermore, the fecundity of female offspring was affected, exhibiting delayed primordial folliculogenesis and puberty onset, reduced levels of ovarian hormones, and impaired developmental competence of MII oocytes. Innovation: These findings provide the first known demonstration that NAPQI, converted from maternal administration of APAP, disturbs meiotic process of fetal oocytes and further impairs female fecundity in adulthood. The concomitant oral dosing with NAM further supports the benefits of NAD+ supplements on oogenesis. Conclusion: Short-term administration of APAP to pregnant mouse caused meiotic aberrations in fetal oocytes by its metabolite NAPQI, whereas co-treatment with NAD+ supplement efficiently relieves the adverse effects by interacting with SIRT7.
    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
    砷污染是全球环境问题。砷引起的慢性肝损伤及其不可逆转的结果,包括肝硬化和肝癌,威胁着砷污染地区居民的健康。肝纤维化是砷诱导的慢性肝损伤进展为肝硬化和肝癌的可逆病理阶段。这项研究的目的是确定基于肝窦内皮细胞(LSECs)去分化的砷诱导的肝纤维化的表观遗传机制。用0.0、2.5、5.0或10.0mg/kg亚砷酸钠治疗大鼠36周。在大鼠肝脏中观察到明显的纤维化表型,表现为肝星状细胞活化和细胞外基质增加,以及胶原纤维的沉积。细胞表面的开窗减少和去分化标记CD31的表达增加证实了肝组织中的LSECs去分化,这也被发现与纤维化表型显著相关。我们进一步发现,砷暴露可以抑制Fcgr2b和Lyve1启动子中组蛋白H3赖氨酸18乙酰化(H3K18ac)的富集,这两个关键基因负责维持LSECs的分化表型。这种抑制随后抑制了基因的表达,促进LSEC去分化和随后的肝纤维化。总之,砷可通过抑制H3K18ac依赖性维持LSEC分化而引发肝纤维化。这些发现揭示了基于表观遗传依赖性LSEC去分化的新见解的砷诱导肝纤维化的新机制。
    Arsenic pollution is a global environmental concern. Arsenic-induced chronic liver injury and its irreversible outcomes, including liver cirrhosis and liver cancer, threaten the health of residents in arsenic-contaminated areas. Liver fibrosis is a reversible pathological stage in the progression of arsenic-induced chronic liver injury to cirrhosis and liver cancer. The aim of this study is to identify the epigenetic mechanism of arsenic-induced liver fibrosis based on the dedifferentiation of liver sinusoidal endothelial cells (LSECs). Rats were treated with 0.0, 2.5, 5.0, or 10.0 mg/kg sodium arsenite for 36 weeks. Marked fibrotic phenotypes were observed in the rat livers, manifested by hepatic stellate cell activation and an increased extracellular matrix, as well as the deposition of collagen fibers. The reduced fenestrations on the cells\' surface and the increased expression of the dedifferentiation marker CD31 corroborated the LSECs\' dedifferentiation in the liver tissue, which was also found to be significantly associated with fibrotic phenotypes. We further revealed that arsenic exposure could inhibit the enrichment of histone H3 lysine 18 acetylation (H3K18ac) in the promoters of Fcgr2b and Lyve1, two key genes responsible for maintaining the differentiation phenotype of LSECs. This inhibition subsequently suppressed the genes\' expression, promoting LSEC dedifferentiation and subsequent liver fibrosis. In conclusion, arsenic can trigger liver fibrosis by inhibiting H3K18ac-dependent maintenance of LSEC differentiation. These findings uncover a novel mechanism of arsenic-induced liver fibrosis based on a new insight into epigenetically dependent LSEC dedifferentiation.
    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
    TFIIIC是RNA聚合酶III转录tRNA所需的多亚基复合物。人TFIIIC完整复合物具有赖氨酸乙酰转移酶活性,其有助于减轻RNA聚合酶III介导的转录和染色质组装的色谱介导的抑制。在这里,我们已经表征了TFIIIC复合物的最大和DNA结合亚基的乙酰转移酶活性,TFIIC220.体外纯化的重组人TFIIIC220乙酰化核心组蛋白H3、H4和H2A。此外,我们已经确定了TFIIIC220的推定催化结构域,该结构域在体外有效地乙酰化核心组蛋白。推定的乙酰辅酶A结合“P环”的关键残基突变会大大降低乙酰转移酶结构域的催化活性。进一步的分析表明,在哺乳动物细胞系中TFIIIC220的敲低显著降低了整体H3K18乙酰化水平,通过过度表达人TFIIIC220的推定乙酰转移酶结构域来挽救。我们的发现表明TFIIIC220在维持细胞乙酰化稳态中可能发挥关键作用。
    TFIIIC is a multi-subunit complex required for tRNA transcription by RNA polymerase III. Human TFIIIC holo-complex possesses lysine acetyltransferase activity that aids in relieving chromatin-mediated repression for RNA polymerase III-mediated transcription and chromatin assembly. Here we have characterized the acetyltransferase activity of the largest and DNA-binding subunit of TFIIIC complex, TFIIIC220. Purified recombinant human TFIIIC220 acetylated core histones H3, H4 and H2A in vitro. Moreover, we have identified the putative catalytic domain of TFIIIC220 that efficiently acetylates core histones in vitro. Mutating critical residues of the putative acetyl-CoA binding \'P loop\' drastically reduced the catalytic activity of the acetyltransferase domain. Further analysis showed that the knockdown of TFIIIC220 in mammalian cell lines dramatically reduces global H3K18 acetylation level, which was rescued by overexpression of the putative acetyltransferase domain of human TFIIIC220. Our findings indicated a possibility of a crucial role for TFIIIC220 in maintaining acetylation homeostasis in the cell.
    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
    砷是一种常见的环境毒物。长期砷暴露可引起各种类型的肝损伤,但是潜在的机制仍然不清楚,因此,有效的预防和治疗措施是未知的。本研究旨在基于组蛋白H3K18乙酰化依赖性抗氧化途径探讨砷诱导大鼠肝损伤的机制,罗莎刺梨特拉特果汁,在打击它。通过组织病理学测量,在暴露于不同剂量的NaAsO2的大鼠中观察到肝脂肪变性和炎性细胞浸润。肝组织中8-OHdG和MDA的增加证实了肝氧化损伤。我们进一步发现,肝脏中H3K18ac的减少显示出剂量-反应关系,随着NaAsO2治疗剂量的增加,并且与8-OHdG和MDA的增加显着相关。ChIP-qPCR的结果确定,H3K18ac在Hspa1a和Hspb8基因启动子中的富集减少导致基因表达的抑制,发现这与砷引起的肝氧化损伤加重有关。值得注意的是,发现刺梨汁可降低肝脏中的8-OHdG和MDA,从而减轻砷引起的组织病理学损伤,通过恢复Hspa1a和Hspb8基因的H3K18ac依赖性转录激活来调节。一起来看,我们提供了一种新的表观遗传学见解,以阐明砷诱导的肝损伤的机制及其通过刺梨汁的挽救。
    Arsenic is a common environmental toxicant. Long-term arsenic exposure can induce various types of liver injury, but the underlying mechanism remains unclear, so effective prevention and treatment measures are unknown. This study aims to explore the mechanism of arsenic-induced rat liver injury based on the histone H3K18 acetylation-dependent antioxidant pathway and to identify the role of a medicinal and edible resource, Rosa roxburghii Tratt juice, in combating it. Hepatic steatosis and inflammatory cell infiltration were observed in rats exposed to different doses of NaAsO2 using histopathological measurement. Increased 8-OHdG and MDA in liver tissue corroborated hepatic oxidative damage. We further found that a reduction in H3K18ac in the liver showed a dose-response relationship, with an increase in the NaAsO2 treatment dose, and it was remarkably associated with increased 8-OHdG and MDA. The results of ChIP-qPCR identified that the decreased enrichment of H3K18ac in promoters of the Hspa1a and Hspb8 genes culminated in the inhibition of the genes\' expression, which was found to be involved in the aggravation of hepatic oxidative damage induced by arsenic. Notably, Rosa roxburghii Tratt juice was found to reduce 8-OHdG and MDA in the liver, thereby alleviating the histopathological lesions induced by arsenic, which was modulated by recovering the H3K18ac-dependent transcriptional activation of the Hspa1a and Hspb8 genes. Taken together, we provide a novel epigenetics insight into clarifying the mechanism of arsenic-induced liver injury and its rescue by Rosa roxburghii Tratt juice.
    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
    组蛋白乙酰化的调节决定了基因表达的模式,从而决定了细胞的同一性。由于它们在癌症生物学中的临床相关性,了解人类胚胎干细胞(hESCs)如何调节其组蛋白乙酰化的基因组模式至关重要,但在很大程度上仍有待调查。这里,我们提供的证据表明,组蛋白H3赖氨酸-18(H3K18ac)和赖氨酸-27(H3K27ac)的乙酰化仅部分通过p300在干细胞中建立,而它代表了体细胞中这些标记的主要组蛋白乙酰转移酶(HAT)。我们的分析表明,尽管p300在hESC中与H3K18ac和H3K27ac略有相关,它在分化时与这些组蛋白标记大部分重叠。有趣的是,我们显示H3K18ac在hESCs中的RNA聚合酶III转录因子C(TFIIIC)富集的“干性”基因中发现,缺少P300此外,在涉及神经元生物学的基因附近也发现了TFIIC,虽然没有H3K18ac。我们的数据表明,与以前认为的相比,在hESC中负责组蛋白乙酰化的HAT模式更为复杂,提示H3K18ac和TFIIIC在调节“干性”基因以及与hESCs神经元分化相关的基因中的推定作用。结果为hESC基因组乙酰化的可能新范例开辟了基础,这可能导致癌症和发育疾病的治疗干预的新途径。
    Regulation of histone acetylation dictates patterns of gene expression and hence cell identity. Due to their clinical relevance in cancer biology, understanding how human embryonic stem cells (hESCs) regulate their genomic patterns of histone acetylation is critical, but it remains largely to be investigated. Here, we provide evidence that acetylation of histone H3 lysine-18 (H3K18ac) and lysine-27 (H3K27ac) is only partially established by p300 in stem cells, while it represents the main histone acetyltransferase (HAT) for these marks in somatic cells. Our analysis reveals that whereas p300 marginally associated with H3K18ac and H3K27ac in hESCs, it largely overlapped with these histone marks upon differentiation. Interestingly, we show that H3K18ac is found at \"stemness\" genes enriched in RNA polymerase III transcription factor C (TFIIIC) in hESCs, whilst lacking p300. Moreover, TFIIIC was also found in the vicinity of genes involved in neuronal biology, although devoid of H3K18ac. Our data suggest a more complex pattern of HATs responsible for histone acetylations in hESCs than previously considered, suggesting a putative role for H3K18ac and TFIIIC in regulating \"stemness\" genes as well as genes associated with neuronal differentiation of hESCs. The results break ground for possible new paradigms for genome acetylation in hESCs that could lead to new avenues for therapeutic intervention in cancer and developmental diseases.
    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
    丹参酮IIA(TanIIA)对脑缺血再灌注损伤(CIRI)具有神经保护作用,但由于水溶性差和强大的首过消除性能,其临床应用受到限制。在这项研究中,我们开发了载有TanIIA(TanIIAME)的微乳液来突破这些限制,并探讨了TanIIAME对CIRI的神经保护作用以及这种神经保护的表观遗传调控机制。在体内,用TanIIAME和TanIIA溶液或丙戊酸钠治疗大脑中动脉闭塞(MCAO)模型。通过ELISA测定法测定TanIIAME对HDAC活性的影响。此外,我们使用原代海马神经元建立氧糖剥夺和复氧(OGD/R)模型。进行乳酸脱氢酶(LDH)测定和末端脱氧核苷酸转移酶dUTP缺口末端标记(TUNEL)测定以研究TanIIAME的神经保护功效。随后,H3K18ac的表达,H4K8ac,在用TanIIAME处理的MCAO或OGD/R模型中研究了NMDAR1,caspase-3和MAP-2,TanIIA溶液或丙戊酸钠。体内实验结果表明,TanIIAME显著降低了神经评分,梗死体积,和HDAC活性与TanIIA溶液和MCAO组相比,伴随着H3K18ac的上调,H4K8ac,MAP-2的表达和NMDAR1和caspase-3的表达下调。此外,在OGD/R模型中,结果表明,随着H3K18ac的增加,TanIIAME治疗具有更好的神经保护作用,H4K8ac,和MAP-2表达和减少NMDAR1和caspase-3表达,与除丙戊酸钠外的其他治疗方法相比。总的来说,TanIIAME治疗通过可能涉及抑制NMDAR1和caspase-3表达以及通过调节组蛋白H3K18和H4K8乙酰化增强MAP-2表达的机制,在保护抗CIRI方面表现出优异的功效。因此,TanIIAME可能用于开发一种有前途的治疗缺血性中风的药物。
    Tanshinone IIA (TanIIA) has neuroprotective effects against cerebral ischemia reperfusion injury (CIRI), but its clinical application is limited due to poor water solubility and robust first pass elimination property. In this study, we developed microemulsion loaded with TanIIA (TanIIA ME) to break through these limitations, and explored the neuroprotective effect of TanIIA ME against CIRI and the epigenetic regulation mechanism of this neuroprotection. In vivo, middle cerebral artery occlusion (MCAO) models were treated with TanIIA ME and TanIIA solution or sodium valproate as a control. The effect of TanIIA ME on HDAC activity was determined by ELISA assay. In addition, we used primary hippocampal neurons to establish oxygen-glucose deprivation and reoxygenation (OGD/R) models. Lactate dehydrogenase (LDH) assay and terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay were performed to investigate the neuroprotective efficacy of TanIIA ME. Subsequently, the expression of H3K18ac, H4K8ac, NMDAR1, caspase-3, and MAP-2 were investigated in MCAO or OGD/R models treated with TanIIA ME, TanIIA solution or sodium valproate. In vivo experimental results indicated that TanIIA ME significantly reduced neurological scores, infarction volume, and HDAC activity compared with TanIIA solution and MCAO group, accompanied by upregulation of H3K18ac, H4K8ac, and MAP-2 expression and downregulation of NMDAR1 and caspase-3 expression. Additionally, in OGD/R models, the results demonstrated that TanIIA ME treatment had a better neuroprotective effect along with increased H3K18ac, H4K8ac, and MAP-2 expression and decreased NMDAR1 and caspase-3 expression, compared with the other treatments except sodium valproate. Overall, TanIIA ME treatment exhibited superior efficacy in protecting against CIRI through mechanisms that might involve the inhibition of NMDAR1 and caspase-3 expression and the enhancement of MAP-2 expression by regulating histone H3K18 and H4K8 acetylation. Thus, TanIIA ME could be potentially used to develop a promising drug for the treatment of ischemic stroke.
    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
    Hexavalent chromium (Cr(VI)) is a well-established human carcinogen with DNA damaging effects. Recently we established a Cr(VI)-induced malignant transformation model from a human bronchial epithelial (16HBE) cell line, and in the transformed (16HBE-T) cells reduced levels of 53BP1 (critical for DNA repair) and the acetylated histone H3K18/27 (H3K18/27ac) were observed. In 16HBE-T cells SET (a multifunctional protein) was elevated by Cr(VI) through quantitative proteomics analysis. In the present study, we further explore the involvement of SET in the H3K18/27ac/53BP1 cascade in the 16HBE-T model, primarily by knockdown of SET. Bioinformatic analysis of the differentially expressed proteins indicated enrichment in histone modifications, in which SET was a major regulator. In 16HBE cells SET expression was enhanced by Cr(VI) in a concentration- and exposure duration-dependent manner. In 16HBE-T cells, SET knockdown showed the following effects: reversal of H3K18/27ac and 53BP1 levels, enhanced enrichment H3K18/27ac in 53BP1\'s promotor region, increase rate of apoptosis and cell cycle G0/G1 arrest (with or without Cr(VI) treatment), and reduced colony-forming efficiency. Finally, In comparison with benzo(a)pyrene-transformed (malignant, 16HBE-B) cells from 16HBE where no changes in H3K18/27ac, 53BP1 or SET were observed, while the H3K18/27ac/53BP1 cascade was downregulated and SET upregulated in 16HBE-T cells, as compared with the parental 16HBE cells; thus the changes in 16HBE-T might be a specific effect of Cr(VI). In conclusion, our results suggest that SET may be involved in the malignant cell transformation, through inhibiting the H3K18/27ac/53BP1 cascade, at least in the 16HBE cell model.
    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
    如何重复元素,表观遗传修饰,和建筑蛋白相互作用,确保正确的基因组表达仍然知之甚少。这里,我们报告的调节机制揭示了Alu元件(AEs)和RNA聚合酶III转录因子C(TFIIIC)在通过染色质循环和组蛋白乙酰化进行基因组结构和功能调节中的重要作用.血清剥夺后,由活性依赖性神经保护者同源异型盒蛋白(ADNP)预先标记并位于细胞周期基因附近的AE子集招募TFIIIC,通过组蛋白H3赖氨酸-18(H3K18)的直接乙酰化改变其染色质可及性。这促进了AEs与其他细胞周期基因启动子附近的远处CTCF位点的接触,在H3K18也被高度乙酰化。这些变化确保了细胞周期基因的基础转录,并且对于血清再暴露后的再激活至关重要。我们的研究揭示了一般转录因子对AE的表观遗传状态的直接操纵如何调节3D基因组折叠和表达。
    How repetitive elements, epigenetic modifications, and architectural proteins interact ensuring proper genome expression remains poorly understood. Here, we report regulatory mechanisms unveiling a central role of Alu elements (AEs) and RNA polymerase III transcription factor C (TFIIIC) in structurally and functionally modulating the genome via chromatin looping and histone acetylation. Upon serum deprivation, a subset of AEs pre-marked by the activity-dependent neuroprotector homeobox Protein (ADNP) and located near cell-cycle genes recruits TFIIIC, which alters their chromatin accessibility by direct acetylation of histone H3 lysine-18 (H3K18). This facilitates the contacts of AEs with distant CTCF sites near promoter of other cell-cycle genes, which also become hyperacetylated at H3K18. These changes ensure basal transcription of cell-cycle genes and are critical for their re-activation upon serum re-exposure. Our study reveals how direct manipulation of the epigenetic state of AEs by a general transcription factor regulates 3D genome folding and expression.
    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
    Acetylation and deacetylation are posttranslational modifications (PTMs) which affect the regulation of chromatin structure and its remodeling. Acetylation of histone 3 at lysine placed on position 18 (H3K18Ac) plays an important role in driving progression of many types of cancer, including breast, colon, lung, hepatocellular, pancreatic, prostate, and thyroid cancer. The aim of this review is to analyze and discuss the newest findings regarding the role of H3K18Ac and acetylation of other histones in carcinogenesis. We summarize the level of H3K18Ac in different cancer cell lines and analyze its association with patients\' outcomes, including overall survival (OS), progression-free survival (PFS), and disease-free survival (DFS). Finally, we describe future perspectives of cancer therapeutic strategies based on H3K18 modifications.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

公众号