Histone Deacetylase 1

组蛋白去乙酰化酶 1
  • 文章类型: Journal Article
    背景:食管癌仍然是一种具有挑战性的疾病,死亡率高,治疗选择少。鉴于这些困难,表观遗传药物已成为患者护理的潜在替代品。这项研究的目标是评估Panobinostat治疗的效果和生物学后果,HDAC(组蛋白去乙酰化酶)抑制剂已被批准用于多发性骨髓瘤患者的治疗,在正常和恶性来源的食管细胞系中,后者代表了两种主要的组织学亚型:腺癌和鳞状细胞癌。
    结果:Panobinostat治疗抑制生长并阻碍增殖,食管癌细胞的集落形成和侵袭。考虑HDAC组织表达,与肿瘤组织相比,HDAC1在正常食管上皮中显著上调,而HDAC3与非恶性粘膜相比在食管癌中过度表达。在正常组织和肿瘤组织之间没有观察到HDAC2和HDAC8表达的差异。
    结论:Panobinostat暴露可有效损害食管癌细胞的恶性特征。因为HDAC3在食道肿瘤样本中显示过表达,这种表观遗传药物可能是食管癌患者的替代治疗选择.
    BACKGROUND: Oesophageal cancer remains a challenging disease with high mortality rates and few therapeutic options. In view of these difficulties, epigenetic drugs have emerged as potential alternatives for patient care. The goal of this study was to evaluate the effect and biological consequences of Panobinostat treatment, an HDAC (histone deacetylase) inhibitor already approved for treatment of patients with multiple myeloma, in oesophageal cell lines of normal and malignant origin, with the latter being representative of the two main histological subtypes: adenocarcinoma and squamous cell carcinoma.
    RESULTS: Panobinostat treatment inhibited growth and hindered proliferation, colony formation and invasion of oesophageal cancer cells. Considering HDAC tissue expression, HDAC1 was significantly upregulated in normal oesophageal epithelium in comparison with tumour tissue, whereas HDAC3 was overexpressed in oesophageal cancer compared to non-malignant mucosa. No differences between normal and tumour tissue were observed for HDAC2 and HDAC8 expression.
    CONCLUSIONS: Panobinostat exposure effectively impaired malignant features of oesophageal cancer cells. Because HDAC3 was shown to be overexpressed in oesophageal tumour samples, this epigenetic drug may represent an alternative therapeutic option for oesophageal cancer patients.
    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
    剪接因子RNA结合基序蛋白10(RBM10)在肺腺癌(LUAD)中经常发生突变(9-25%)。大多数RBM10癌症突变是功能丧失的,与增加的肿瘤发生和限制当前LUAD靶向治疗的疗效相关。值得注意的是,利用RBM10缺乏症的治疗策略仍有待探索。这里,我们进行了CRISPR-Cas9合成致死性(SL)筛选,并鉴定了约60个RBM10SL基因,包括WEE1激酶。WEE1抑制在体外和体内使RBM10缺陷型LUAD细胞敏感。机械上,我们确定了RBM10在调节DNA复制叉进程和复制应激反应中的独立剪接作用,支撑RBM10-WEE1SL。此外,RBM10与活跃的DNA复制叉相互作用,依赖于合成冈崎RNA引物的DNA引物亚基1(PRIM1)。功能上,我们证明RBM10作为一个锚募集组蛋白去乙酰化酶1(HDAC1),以促进H4K16去乙酰化和R环稳态,以维持复制叉的稳定性。总的来说,我们的数据揭示了RBM10在微调DNA复制中的作用,并为靶向RBM10缺陷型肿瘤提供了治疗手段.
    The splicing factor RNA-binding motif protein 10 (RBM10) is frequently mutated in lung adenocarcinoma (LUAD) (9-25%). Most RBM10 cancer mutations are loss-of-function, correlating with increased tumorigenesis and limiting the efficacy of current LUAD targeted therapies. Remarkably, therapeutic strategies leveraging RBM10 deficiency remain unexplored. Here, we conduct a CRISPR-Cas9 synthetic lethality (SL) screen and identify ~60 RBM10 SL genes, including WEE1 kinase. WEE1 inhibition sensitizes RBM10-deficient LUAD cells in-vitro and in-vivo. Mechanistically, we identify a splicing-independent role of RBM10 in regulating DNA replication fork progression and replication stress response, which underpins RBM10-WEE1 SL. Additionally, RBM10 interacts with active DNA replication forks, relying on DNA Primase Subunit 1 (PRIM1) that synthesizes Okazaki RNA primers. Functionally, we demonstrate that RBM10 serves as an anchor for recruiting Histone Deacetylase 1 (HDAC1) to facilitate H4K16 deacetylation and R-loop homeostasis to maintain replication fork stability. Collectively, our data reveal a role of RBM10 in fine-tuning DNA replication and provide therapeutic arsenal for targeting RBM10-deficient tumors.
    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
    背景:肺癌是世界上第二常见的恶性肿瘤。解决推动非小细胞肺癌进展的潜在机制至关重要。在这项研究中,我们已经阐明了PHF12在这方面的关键作用。
    方法:我们利用临床肺癌组织样本和非小细胞肺癌细胞系来辨别PHF12的表达模式。进行探查细胞增殖的体外测定以证实PHF12的功能影响。此外,采用体内异种移植模型来剖析PHF12的作用。采用ChIP分析和qRT-PCR,我们深入研究了PHF12和HDAC1之间复杂的结合动力学。通过RNA-seq和GSEA分析,探讨了PHF12-HDAC1轴在肺癌进展中的机制见解。
    结果:值得注意的是,PHF12在肿瘤组织内表现出实质性的上调,伴随着它与HDAC1的相关性。细胞增殖测定的三部曲,transwell分析,异种移植模型共同强调了PHF12对肺癌增殖的促进作用,在体外和体内。ChIP测定揭示了PHF12在控制HDAC1表达中的转录调节作用。这种相关性延伸到mRNA和蛋白质水平。PHF12通过调节HDCA1表达促进NSCLC进展。有趣的是,PHF12敲低后NSCLC细胞系内的功能拯救是通过HDAC1过表达实现的.此外,我们的发现揭示了PHF12-HDAC1轴激活EGFR/AKT信号通路的能力,从而进一步证实其在肺癌进展中的意义。
    结论:我们的研究首次确定PHF12在肺癌增殖和迁移中具有致癌作用。PHF12转录调控HDAC1并激活EGFR/AKT信号通路在NSCLC进展中的作用.PHF12可能作为肺癌治疗的重要靶点。
    BACKGROUND: Lung cancer stands as the second most prevalent malignant neoplasm worldwide. Addressing the underlying mechanisms propelling the progression of non-small cell lung cancer is of paramount importance. In this study, we have elucidated the pivotal role of PHF12 in this context.
    METHODS: We harnessed clinical lung cancer tissue samples and non-small cell lung cancer cell lines to discern the expression pattern of PHF12. In vitro assays probing cell proliferation were conducted to substantiate the functional impact of PHF12. Furthermore, an in vivo Xenograft model was employed to dissect the role of PHF12. Employing ChIP assays and qRT-PCR, we delved into the intricate binding dynamics between PHF12 and HDAC1. Mechanistic insights into the PHF12-HDAC1 axis in lung cancer progression were pursued via RNA-seq and GSEA analyses.
    RESULTS: Notably, PHF12 exhibited a substantial upregulation within tumor tissue, concomitant with its correlation to HDAC1. The trilogy of cell proliferation assays, transwell assays, and the Xenograft model collectively underscored the promoting influence of PHF12 on lung cancer proliferation, both in vitro and in vivo. The ChIP assay unveiled the transcriptional regulatory role of PHF12 in governing HDAC1 expression. This correlation extended to both mRNA and protein levels. PHF12 promotes NSCLC progression through regulating HDCA1 expression. Intriguingly, the rescue of function within NSCLC cell lines post PHF12 knockdown was achievable through HDAC1 overexpression. Additionally, our findings unveiled the capacity of the PHF12-HDAC1 axis to activate the EGFR/AKT signaling pathway, thereby further corroborating its significance in lung cancer progression.
    CONCLUSIONS: Our study identified PHF12 as an oncogenic role in lung cancer proliferation and migration for the first time. PHF12 transcriptionally regulate HDAC1 and activate EGFR/AKT signaling pathway in NSCLC progression. PHF12 may serve as an important target in lung cancer therapy.
    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
    组蛋白精氨酸残基甲基化对个体发育和基因调控至关重要。然而,组蛋白精氨酸甲基化响应细胞应激的动力学仍未被研究。此外,这种组蛋白修饰与其他组蛋白修饰之间的相互作用和调节机制是重要的科学问题,需要进一步研究.本研究旨在探讨组蛋白精氨酸甲基化在DNA损伤中的变化。我们报告了响应DNA损伤,组蛋白H3R26对称二甲基化(H3R26me2s)和H3K27位点的低乙酰化总体减少。值得注意的是,H3R26me2s在整个基因组中表现出与H3K27ac相似的分布模式,两者都对H3K27me3具有拮抗作用。此外,组蛋白脱乙酰酶1(HDAC1)可以募集到H3R26me2s去甲基化区域以介导H3K27脱乙酰。这些发现表明H3R26me2s和H3K27ac在调节基因表达中的串扰。
    Histone arginine residue methylation is crucial for individual development and gene regulation. However, the dynamics of histone arginine methylation in response to cellular stress remains largely unexplored. In addition, the interplay and regulatory mechanisms between this and other histone modifications are important scientific questions that require further investigation. This study aimed to investigate the changes in histone arginine methylation in response to DNA damage. We report a global decrease in histone H3R26 symmetric dimethylation (H3R26me2s) and hypoacetylation at the H3K27 site in response to DNA damage. Notably, H3R26me2s exhibits a distribution pattern similar to that of H3K27ac across the genome, both of which are antagonistic to H3K27me3. Additionally, histone deacetylase 1 (HDAC1) may be recruited to the H3R26me2s demethylation region to mediate H3K27 deacetylation. These findings suggest crosstalk between H3R26me2s and H3K27ac in regulating gene 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
    目的:动脉粥样硬化(AS)是心血管疾病的常见发病机制。葛根素(Pue)是一种用于预防和治疗AS的中草药。这里,这项研究调查了Pue对AS进展的影响。
    方法:用丙烯醛诱导ApoE-/-小鼠。体重,血脂指标,炎症因子,线粒体氧化应激,并检测到脂质沉积。ELISA法检测IL-6和TNF-α。采用油红染色和H&E染色观察主动脉窦斑块病变。血清炎症因子IL-6、TNF-α的表达,SOD,ELISA法检测GSH和MDA,RT-qPCR检测主动脉中HDAC1的mRNA表达水平,免疫组化法检测主动脉中IL-6和TNF-α的表达。JNK,p-JNK,通过蛋白质印迹法检测OPA-1和HDAC1。
    结果:Pue给药可有效减少丙烯醛诱导的AS小鼠脂质蓄积。Pue促进了SOD的活性,GSH和MDA,并抑制动脉粥样硬化斑块的形成和主动脉组织学改变的过程。Pue降低IL-6和TNF-α。HDAC1表达下调,p-JNK-1和JNK蛋白表达上调。
    结论:Pue通过介导JNK通路抑制HDAC1介导的氧化应激紊乱,减轻炎症和减轻丙烯醛诱导的AS。
    Atherosclerosis (AS) is a common pathogenesis of cardiovascular diseases. Puerarin (Pue) is a Chinese herbal remedy used to prevent and treat AS. Here, this research investigated the effect of Pue on AS progression.
    ApoE-/- mice were induced with acrolein. Body weight, blood lipid index, inflammatory factors, mitochondrial oxidative stress, and lipid deposition were detected. IL-6 and TNF-α were detected by ELISA. Oil red staining and H&E staining were used to observe the aortic sinus plaque lesions. Serum expressions of inflammatory factors IL-6, TNF-a, SOD, GSH and MDA were detected by ELISA, the mRNA expression levels of HDAC1 in the aorta were detected by RT-qPCR, and IL-6 and TNF-α in the aorta were detected by immunohistochemistry. JNK, p-JNK, OPA-1, and HDAC1 were detected by Western blotting.
    Pue administration can effectively reduce lipid accumulation in AS mice induced by acrolein. Pue promoted the activity of SOD, GSH and MDA, and inhibited the formation of atherosclerotic plaques and the process of aortic histological changes. Pue reduced IL-6 and TNF-α. HDAC1 expression was down-regulated and p-JNK-1 and JNK protein expression was up-regulated.
    Pue reduces inflammation and alleviates AS induced by acrolein by mediating the JNK pathway to inhibit HDAC1-mediated oxidative stress disorder.
    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
    背景:乳腺癌是女性中最致命的癌症之一。尽管在乳腺癌的诊断和治疗方面取得了重大进展,许多患者仍然屈服于这种疾病,因此,迫切需要新的有效治疗方法。天然产物香豆素已被广泛研究,因为它揭示了医药领域的各种生物学特性。越来越多的证据表明,组蛋白去乙酰化酶抑制剂(HDACIs)是有前途的新型抗乳腺癌药物。然而,目前的大多数HDACIs对实体肿瘤仅表现出中等效果,并伴有严重的副作用。因此,开发更有效的乳腺癌治疗HDACIs,HDACIs的异羟肟酸盐与香豆素核心有关,并设计合成了香豆素-异羟肟酸盐杂种。
    方法:通过药效团融合策略将取代的香豆素部分掺入到经典的异羟肟酸盐HDACIs中。通过使用HDACI筛选试剂盒和细胞活力测定鉴定ZN444B。进行分子对接以探索ZN444B与HDAC1的结合模式。蛋白质印迹,免疫荧光染色,细胞活力,使用集落形成和细胞迁移以及流式细胞术测定来分析ZN444B的体外抗乳腺癌作用。将小鼠模型中的原位研究用于体内功效和毒性的临床前评估。蛋白质组学分析,双荧光素酶报告分析,染色质免疫沉淀,免疫共沉淀,免疫荧光染色分析和免疫组织化学(IHC)分析用于阐明ZN444B作用的分子基础。
    结果:我们合成并鉴定了一种新的香豆素-异羟肟酸盐偶联物,ZN444B在体外和体内均具有有希望的抗乳腺癌活性。分子对接模型显示ZN444B以高亲和力结合HDAC1。进一步的机理研究表明,ZN444B通过抑制HDAC1在K703对Sp1的脱乙酰酶活性而特异性降低FOS样抗原2(FOSL2)mRNA水平,并消除Sp1与FOSL2启动子的结合能力。此外,FOSL2表达与乳腺癌进展和转移呈正相关。沉默FOSL2表达降低了乳腺癌细胞对ZN444B治疗的敏感性。此外,ZN444B在小鼠中没有显示全身毒性。
    结论:我们的发现强调了FOSL2作为乳腺癌新的生物标志物和治疗靶点的潜力,并且用ZN444B靶向HDAC1-Sp1-FOSL2信号轴可能是一种有希望的乳腺癌治疗策略。
    BACKGROUND: Breast cancer is one of the most lethal cancers in women. Despite significant advances in the diagnosis and treatment of breast cancer, many patients still succumb to this disease, and thus, novel effective treatments are urgently needed. Natural product coumarin has been broadly investigated since it reveals various biological properties in the medicinal field. Accumulating evidence indicates that histone deacetylase inhibitors (HDACIs) are promising novel anti-breast cancer agents. However, most current HDACIs exhibit only moderate effects against solid tumors and are associated with severe side effects. Thus, to develop more effective HDACIs for breast cancer therapy, hydroxamate of HDACIs was linked to coumarin core, and coumarin-hydroxamate hybrids were designed and synthesized.
    METHODS: A substituted coumarin moiety was incorporated into the classic hydroxamate HDACIs by the pharmacophore fusion strategy. ZN444B was identified by using the HDACI screening kit and cell viability assay. Molecular docking was performed to explore the binding mode of ZN444B with HDAC1. Western blot, immunofluorescent staining, cell viability, colony formation and cell migration and flow cytometry assays were used to analyze the anti-breast cancer effects of ZN444B in vitro. Orthotopic studies in mouse models were applied for preclinical evaluation of efficacy and toxicity in vivo. Proteomic analysis, dual-luciferase reporter assay, chromatin immunoprecipitation, co-immunoprecipitation, immunofluorescent staining assays along with immunohistochemical (IHC) analysis were used to elucidate the molecular basis of the actions of ZN444B.
    RESULTS: We synthesized and identified a novel coumarin-hydroxamate conjugate, ZN444B which possesses promising anti-breast cancer activity both in vitro and in vivo. A molecular docking model showed that ZN444B binds to HDAC1 with high affinity. Further mechanistic studies revealed that ZN444B specifically decreases FOS-like antigen 2 (FOSL2) mRNA levels by inhibiting the deacetylase activity of HDAC1 on Sp1 at K703 and abrogates the binding ability of Sp1 to the FOSL2 promoter. Furthermore, FOSL2 expression positively correlates with breast cancer progression and metastasis. Silencing FOSL2 expression decreases the sensitivity of breast cancer cells to ZN444B treatment. In addition, ZN444B shows no systemic toxicity in mice.
    CONCLUSIONS: Our findings highlight the potential of FOSL2 as a new biomarker and therapeutic target for breast cancer and that targeting the HDAC1-Sp1-FOSL2 signaling axis with ZN444B may be a promising therapeutic strategy for breast cancer.
    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
    双重靶向染色质调节和DNA损伤修复信号为癌症治疗提供了有希望的途径。应用合理的药物设计,我们合成了一种有效的双靶向小分子,SP-1-303.这里,我们报道了SP-1-303作为I类同工型选择性组蛋白去乙酰化酶(HDAC)抑制剂和共济失调-毛细血管扩张突变蛋白(ATM)的激活剂.体外酶分析显示HDAC1和HDAC3的选择性抑制。细胞生长抑制研究表明,SP-1-303对MCF-7和T47D细胞的有效生长抑制浓度(EC50)为0.32至0.34μM,差异抑制雌激素受体阳性乳腺癌(ERBC)细胞的生长。与三阴性乳腺癌细胞的1.2-2.5μM相比,和约12μM的正常乳腺上皮细胞。Western分析显示,SP-1-303降低雌激素受体α(ER-α)表达并增加p53蛋白表达,同时诱导ATM及其底物的磷酸化,BRCA1和p53在ER+BC细胞中呈时间依赖性。药代动力学评价表明,在大鼠模型中静脉内给药后,曲线下面积(AUC)为5227.55ng/ml×h,消除半衰期为1.26h。总的来说,SP-1-303作为一种新型第二代I类(HDAC1和HDAC3)选择性HDAC抑制剂,和ATM激活器,能够调节ER表达,并抑制ER+BC细胞的生长。SP-1-303对I类HDAC和ATM的联合靶向为治疗ER+乳腺癌提供了有希望的治疗方法,并支持进一步的临床前评估。
    Dual-targeting chromatin regulation and DNA damage repair signaling presents a promising avenue for cancer therapy. Applying rational drug design, we synthesized a potent dual-targeting small molecule, SP-1-303. Here, we report SP-1-303 as a class I isoform selective histone deacetylase (HDAC) inhibitor and an activator of the ataxia-telangiectasia mutated protein (ATM). In vitro enzymatic assays demonstrated selective inhibition of HDAC1 and HDAC3. Cellular growth inhibition studies show that SP-1-303 differentially inhibits growth of estrogen receptor positive breast cancer (ER+ BC) cells with effective growth inhibition concentrations (EC50) for MCF-7 and T47D cells ranging from 0.32 to 0.34 μM, compared to 1.2-2.5 μM for triple negative breast cancer cells, and ~12 μM for normal breast epithelial cells. Western analysis reveals that SP-1-303 decreases estrogen receptor alpha (ER-α) expression and increases p53 protein expression, while inducing the phosphorylation of ATM and its substrates, BRCA1 and p53, in a time-dependent manner in ER+ BC cells. Pharmacokinetic evaluation demonstrates an area under the curve (AUC) of 5227.55 ng/ml × h with an elimination half-life of 1.26 h following intravenous administration in a rat model. Collectively, SP-1-303 emerges as a novel second generation class I (HDAC1 and HDAC3) selective HDAC inhibitor, and ATM activator, capable of modulating ER expression, and inhibiting growth of ER+ BC cells. Combined targeting of class I HDACs and ATM by SP-1-303 offers a promising therapeutic approach for treating ER+ breast cancers and supports further preclinical evaluation.
    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
    急性髓细胞性白血病(AML)是一种以血液和骨髓浸润为特征的血液恶性肿瘤,缓解率低,复发率高。目前的研究表明,I类HDAC抑制剂可以下调抗凋亡蛋白,导致AML细胞凋亡。在目前的调查中,我们对海洋细胞毒素SantacruzamateA(SCA)进行了结构修饰,一种以其对HDAC的抑制活性而闻名的化合物,导致开发了一系列新型有效的I类HDACs酰肼抑制剂。代表性的基于酰肼的化合物25c表现出作为单一药剂的AML细胞中的细胞凋亡的浓度依赖性诱导。此外,25c与维奈托克联用时表现出协同抗AML作用,用于AML治疗的临床Bcl-2抑制剂。这种组合导致抗凋亡蛋白Mcl-1和Bcl-xL更明显的下调,与单一疗法相比,促凋亡蛋白caspase3和DNA双链断裂生物标志物γ-H2AX的显着上调。这些结果强调了25c作为AML治疗的有希望的先导化合物的潜力。特别是与维奈托克联合使用时。
    Acute myeloid leukemia (AML) is a hematologic malignancy characterized by infiltration of the blood and bone marrow, exhibiting a low remission rate and high recurrence rate. Current research has demonstrated that class I HDAC inhibitors can downregulate anti-apoptotic proteins, leading to apoptosis of AML cells. In the present investigation, we conducted structural modifications of marine cytotoxin Santacruzamate A (SCA), a compound known for its inhibitory activity towards HDACs, resulting in the development of a novel series of potent class I HDACs hydrazide inhibitors. Representative hydrazide-based compound 25c exhibited concentration-dependent induction of apoptosis in AML cells as a single agent. Moreover, 25c exhibited a synergistic anti-AML effect when combined with Venetoclax, a clinical Bcl-2 inhibitor employed in AML therapy. This combination resulted in a more pronounced downregulation of anti-apoptotic proteins Mcl-1 and Bcl-xL, along with a significant upregulation of the pro-apoptotic protein cleaved-caspase3 and the DNA double-strand break biomarker γ-H2AX compared to monotherapy. These results highlighted the potential of 25c as a promising lead compound for AML treatment, particularly when used in combination with Venetoclax.
    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
    获取DNA是调节基因转录的第一控制水平,对于维持DNA完整性也至关重要的控制。细胞衰老的特征在于深度转录重排和DNA损伤的积累。这里,我们发现HDAC4和HDAC1/HDAC2之间的表观遗传复合物参与H2BK120乙酰化的擦除。HDAC4/HDAC1/HDAC2复合物通过同源重组调节DNA修复效率,通过H2BK120的动态脱乙酰。HDAC4的缺乏导致H2BK120ac的积累,损伤部位的BRCA1和CtIP募集受损,受损DNA的积累和衰老。在衰老细胞中,由于HDAC4的蛋白酶体降解增加,该复合物被分解。在RAS诱导的衰老过程中HDAC4的强制表达减少了γH2AX的基因组扩散。它也影响H2BK120ac水平,在RAS诱导的衰老过程中积累的DNA损伤区域增加。总之,衰老过程中HDAC4的降解导致受损DNA的积累,并有助于激活由维持衰老的超增强子控制的转录程序。
    Access to DNA is the first level of control in regulating gene transcription, a control that is also critical for maintaining DNA integrity. Cellular senescence is characterized by profound transcriptional rearrangements and accumulation of DNA lesions. Here, we discovered an epigenetic complex between HDAC4 and HDAC1/HDAC2 that is involved in the erase of H2BK120 acetylation. The HDAC4/HDAC1/HDAC2 complex modulates the efficiency of DNA repair by homologous recombination, through dynamic deacetylation of H2BK120. Deficiency of HDAC4 leads to accumulation of H2BK120ac, impaired recruitment of BRCA1 and CtIP to the site of lesions, accumulation of damaged DNA and senescence. In senescent cells this complex is disassembled because of increased proteasomal degradation of HDAC4. Forced expression of HDAC4 during RAS-induced senescence reduces the genomic spread of γH2AX. It also affects H2BK120ac levels, which are increased in DNA-damaged regions that accumulate during RAS-induced senescence. In summary, degradation of HDAC4 during senescence causes the accumulation of damaged DNA and contributes to the activation of the transcriptional program controlled by super-enhancers that maintains senescence.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • DOI:
    文章类型: Journal Article
    PRAME(黑色素瘤优先表达的抗原)的上调与多种癌症的进展有关。包括黑色素瘤.肿瘤抑制因子p53是一种转录调节因子,其响应于应激信号而介导细胞周期停滞和细胞凋亡。这里,我们报道PRAME是一种新的p53抑制靶点。这得到了对携带野生型p53的黑素瘤细胞系和人黑素瘤数据库的分析的支持。PRAME的mRNA表达因p53过表达和使用DNA损伤剂的激活而下调,但通过p53耗竭上调。我们在PRAME的启动子区域中鉴定了p53反应元件(p53RE)。荧光素酶和ChIP分析显示p53抑制PRAME启动子的转录活性,并在依托泊苷处理后与HDAC1一起募集到p53RE。通过测量黑色素瘤细胞中的集落形成和p27表达证明了p53激活介导的PRAME下调的功能意义。这些数据表明p53激活,导致PRAME下调,可能是黑色素瘤细胞的治疗策略。
    Upregulation of PRAME (preferentially expressed antigen of melanoma) has been implicated in the progression of a variety of cancers, including melanoma. The tumor suppressor p53 is a transcriptional regulator that mediates cell cycle arrest and apoptosis in response to stress signals. Here, we report that PRAME is a novel repressive target of p53. This was supported by analysis of melanoma cell lines carrying wild-type p53 and human melanoma databases. mRNA expression of PRAME was downregulated by p53 overexpression and activation using DNA-damaging agents, but upregulated by p53 depletion. We identified a p53-responsive element (p53RE) in the promoter region of PRAME. Luciferase and ChIP assays showed that p53 represses the transcriptional activity of the PRAME promoter and is recruited to the p53RE together with HDAC1 upon etoposide treatment. The functional significance of p53 activationmediated PRAME downregulation was demonstrated by measuring colony formation and p27 expression in melanoma cells. These data suggest that p53 activation, which leads to PRAME downregulation, could be a therapeutic strategy in melanoma cells. [BMB Reports 2024; 57(6): 299-304].
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号