base excision repair

基底切除修复
  • 文章类型: Journal Article
    三核苷酸重复序列(TNR)扩增是导致40多种神经退行性疾病的原因,包括亨廷顿病和弗里德赖希共济失调(FRDA)。由于对神经系统中体细胞TNR扩张和收缩的分子机制了解不足,因此没有有效的治疗方法。我们和其他人发现DNA碱基切除修复(BER)积极调节TNR的不稳定性,通过DNA修复收缩扩展的重复序列,揭示了有效治疗疾病的发展。在这项研究中,替莫唑胺(TMZ)被用作模型DNA碱基损伤剂,以揭示BER途径在FRDA神经细胞和转基因小鼠中调节共济失调(FXN)基因的GAA重复不稳定性的机制。我们发现TMZ在FRDA小鼠脑组织中诱导大的GAA重复收缩,神经元,和FRDAiPSC分化的神经细胞,增加FRDA小鼠脑和神经细胞中共济失调蛋白的水平。令人惊讶的是,我们发现TMZ还可以抑制H3K9甲基转移酶,导致开放的染色质和增加的ssDNA断裂和关键的BER酶的募集,polβ,FRDA神经细胞的重复。我们进一步证明,H3K9甲基转移酶抑制剂BIX01294还通过打开染色质并增加内源性ssDNA断裂和重复序列上polβ的募集,诱导了FRDA神经细胞中扩增重复序列的收缩和共济失调蛋白的增加。我们的研究提供了新的机制见解,说明H3K9甲基化的抑制可以与BER串扰以诱导FRDA中的GAA重复收缩。我们的结果将为通过靶向组蛋白甲基化和BER途径来开发重复扩增疾病的新型基因治疗开辟一条新途径。
    Trinucleotide repeat (TNR) expansion is the cause of over 40 neurodegenerative diseases, including Huntington\'s disease and Friedreich\'s ataxia (FRDA). There are no effective treatments for these diseases due to the poor understanding of molecular mechanisms underlying somatic TNR expansion and contraction in neural systems. We and others have found that DNA base excision repair (BER) actively modulates TNR instability, shedding light on the development of effective treatments for the diseases by contracting expanded repeats through DNA repair. In this study, temozolomide (TMZ) was employed as a model DNA base damaging agent to reveal the mechanisms of the BER pathway in modulating GAA repeat instability at the frataxin (FXN) gene in FRDA neural cells and transgenic mouse mice. We found that TMZ induced large GAA repeat contraction in FRDA mouse brain tissue, neurons, and FRDA iPSC-differentiated neural cells, increasing frataxin protein levels in FRDA mouse brain and neural cells. Surprisingly, we found that TMZ could also inhibit H3K9 methyltransferases, leading to open chromatin and increasing ssDNA breaks and recruitment of the key BER enzyme, pol β, on the repeats in FRDA neural cells. We further demonstrated that the H3K9 methyltransferase inhibitor BIX01294 also induced the contraction of the expanded repeats and increased frataxin protein in FRDA neural cells by opening the chromatin and increasing the endogenous ssDNA breaks and recruitment of pol β on the repeats. Our study provides new mechanistic insight illustrating that inhibition of H3K9 methylation can crosstalk with BER to induce GAA repeat contraction in FRDA. Our results will open a new avenue for developing novel gene therapy by targeting histone methylation and the BER pathway for repeat expansion diseases.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: English Abstract
    非洲猪瘟病毒(ASFV)作为一种传染性病毒病原体,负责非洲猪瘟(ASF)的发生,一种快速传播和高度致命的疾病。自2018年ASFV引入中国以来,已迅速传播到许多省份,给我国养猪业带来了巨大的挑战。由于对ASFV发病机制的认识有限,既没有疫苗也没有抗病毒药物。我们已经发现ASFV感染可以诱导细胞的氧化应激反应,DNA修复酶在这一过程中起着关键作用。这项研究采用了RNA干扰,RT-qPCR,西方印迹,血液吸附(HAD),和流式细胞术研究DNA修复酶OGG1和MTH1抑制剂对ASFV复制的影响,并评估抑制剂的抗ASFV作用。本研究为抗病毒药物的开发提供了参考。
    African swine fever virus (ASFV), as a contagious viral pathogen, is responsible for the occurrence of African swine fever (ASF), a rapidly spreading and highly lethal disease. Since ASFV was introduced into China in 2018, it has been quickly spread to many provinces, which brought great challenges to the pig industry in China. Due to the limited knowledge about the pathogenesis of ASFV, neither vaccines nor antiviral drugs are available. We have found that ASFV infection can induce oxidative stress responses in cells, and DNA repair enzymes play a key role in this process. This study employed RNA interference, RT-qPCR, Western blotting, Hemadsorption (HAD), and flow cytometry to investigate the effects of the inhibitors of DNA repair enzymes OGG1 and MTH1 on ASFV replication and evaluated the anti-ASFV effects of the inhibitors. This study provides reference for the development of anti-viral drugs.
    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
    NEIL1是识别并启动氧化碱基的碱基切除修复的DNA糖基化酶。普遍存在的ssDNA结合支架蛋白,复制蛋白A(RPA),以依赖于DNA结构的方式调节NEIL1活性。已经报道了NEIL1和RPA之间的相互作用,但是这种相互作用的分子基础还有待研究。使用NMR光谱和等温滴定量热法(ITC)的组合,我们表明NEIL1通过两个接触点与RPA相互作用。将与RPA32C蛋白募集结构域的相互作用映射到NEIL1的共同相互作用结构域(CID)中的基序,并测量200nM的解离常数(Kd)。与串联RPA70ABssDNA结合结构域的明显较弱的二级相互作用也被映射到CID。这两个接触点共同表明,NEIL1对RPA具有很高的总体亲和力(Kd~20nM)。在CID中产生了RPA32C与NEIL1RPA结合基序的复合物的同源模型,并用于设计NEIL1中的一组突变以破坏相互作用,ITC证实了这一点。突变体NEIL1在体外对双链体DNA中的胸腺嘧啶二醇损伤保持催化活性。使用荧光多重宿主细胞再激活(FM-HCR)报告分析测试体内破坏NEIL1-RPA相互作用的功能作用揭示了NEIL1在核苷酸切除修复中的意外作用。在NEIL1在复制相关修复中的作用的背景下讨论了这些发现。
    NEIL1 is a DNA glycosylase that recognizes and initiates base excision repair of oxidized bases. The ubiquitous ssDNA binding scaffolding protein, replication protein A (RPA), modulates NEIL1 activity in a manner that depends on DNA structure. Interaction between NEIL1 and RPA has been reported, but the molecular basis of this interaction has yet to be investigated. Using a combination of NMR spectroscopy and isothermal titration calorimetry (ITC), we show that NEIL1 interacts with RPA through two contact points. An interaction with the RPA32C protein recruitment domain was mapped to a motif in the common interaction domain (CID) of NEIL1 and a dissociation constant (Kd) of 200 nM was measured. A substantially weaker secondary interaction with the tandem RPA70AB ssDNA binding domains was also mapped to the CID. Together these two contact points reveal NEIL1 has a high overall affinity (Kd ∼ 20 nM) for RPA. A homology model of the complex of RPA32C with the NEIL1 RPA binding motif in the CID was generated and used to design a set of mutations in NEIL1 to disrupt the interaction, which was confirmed by ITC. The mutant NEIL1 remains catalytically active against a thymine glycol lesion in duplex DNA in vitro. Testing the functional effect of disrupting the NEIL1-RPA interaction in vivo using a Fluorescence Multiplex-Host Cell Reactivation (FM-HCR) reporter assay revealed an unexpected role for NEIL1 in nucleotide excision repair. These findings are discussed in the context of the role of NEIL1 in replication-associated repair.
    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
    我们调查了氧化DNA损伤和修复之间的相互作用,编码主要碱基切除修复(BER)酶和旁路DNA聚合酶的基因的表达,和哺乳动物细胞中的诱变。原代小鼠胚胎成纤维细胞被亚甲蓝和可见光诱导的氧化应激攻击,以及DNA损伤的形成和修复,基因表达的变化,并且在处理后(0-192小时)以增加的间隔测定诱变。氧化DNA损伤的显着形成以及Ogg1,Polβ的上调,和波尔κ,在处理的细胞中没有发现Mutyh和Nudt1表达的变化。Ogg1和Polβ表达与DNA损伤形成和修复之间存在明显的相互联系,这两个基因的表达变化与氧化性DNA损伤水平成正比。超过3小时的滞后时间(P<0.05)。Polκ表达与氧化性DNA损伤和修复动力学的匹配模式同样显著(P<0.05)。DNA损伤和基因表达数据与处理细胞中的诱变性数据非常一致;诱导的突变谱指示在基因组DNA复制期间氧化的DNA碱基的错误旁路和氧化的脱氧核苷三磷酸的掺入。我们的发现支持后续功能研究,以阐明DNA碱基和核苷酸池的氧化,Polk过表达,Ogg1和Polβ的延迟上调,Nudt1和Mutyh的表达不足共同影响氧化应激后的诱变。
    We investigated the interplay among oxidative DNA damage and repair, expression of genes encoding major base excision repair (BER) enzymes and bypass DNA polymerases, and mutagenesis in mammalian cells. Primary mouse embryonic fibroblasts were challenged with oxidative stress induced by methylene blue plus visible light, and formation and repair of DNA damage, changes in gene expression, and mutagenesis were determined at increasing intervals post-treatment (0 - 192 hours). Significant formation of oxidative DNA damage together with upregulation of Ogg1, Polβ, and Polκ, and no changes in Mutyh and Nudt1 expression were found in treated cells. There was a distinct interconnection between Ogg1 and Polβ expression and DNA damage formation and repair whereby changes in expression of these two genes were proportionate to the levels of oxidative DNA damage, once a 3-plus hour lag time passed (P < 0.05). Equally notable was the matching pattern of Polκ expression and kinetics of oxidative DNA damage and repair (P < 0.05). The DNA damage and gene expression data were remarkably consistent with mutagenicity data in the treated cells; the induced mutation spectrum is indicative of erroneous bypass of oxidized DNA bases and incorporation of oxidized deoxynucleoside triphosphates during replication of the genomic DNA. Our findings support follow-up functional studies to elucidate how oxidation of DNA bases and the nucleotide pool, overexpression of Polκ, delayed upregulation of Ogg1 and Polβ, and inadequate expression of Nudt1 and Mutyh collectively affect mutagenesis consequent to oxidative stress.
    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
    非膜区室或生物分子缩合物在调节包括DNA修复的细胞过程中起重要作用。这里,XRCC1是一种参与DNA碱基切除修复(BER)和单链断裂修复的支架蛋白,发现在DNA双链体存在下形成富含蛋白质的微相。我们还表明,XRCC1的存在显着增加了BER相关的DNA聚合酶λ(Polλ)的填补活性。仅在微摩尔XRCC1浓度下观察到Polλ活性的刺激,远高于XRCC1-Polλ复合物确定的纳摩尔解离常数,并指出除蛋白质-蛋白质相互作用外还存在辅助刺激因子。的确,根据动态光散射测量,XRCC1对Polλ活性的刺激与蛋白质-DNA混合物中的微相分离相结合。荧光显微镜显示Polλ,XRCC1和微相内的缺口DNA。因此,Polλ活性的刺激是由其与XRCC1的相互作用以及微相分离的特定条件引起的;这种现象首次显示。
    Non-membrane compartments or biomolecular condensates play an important role in the regulation of cellular processes including DNA repair. Here, an ability of XRCC1, a scaffold protein involved in DNA base excision repair (BER) and single-strand break repair, to form protein-rich microphases in the presence of DNA duplexes was discovered. We also showed that the gap-filling activity of BER-related DNA polymerase λ (Pol λ) is significantly increased by the presence of XRCC1. The stimulation of the Pol λ activity was observed only at micromolar XRCC1 concentrations, which were well above the nanomolar dissociation constant determined for the XRCC1-Pol λ complex and pointed to the presence of an auxiliary stimulatory factor in addition to protein-protein interactions. Indeed, according to dynamic light scattering measurements, the stimulation of the Pol λ activity by XRCC1 was coupled with microphase separation in a protein-DNA mixture. Fluorescence microscopy revealed colocalization of Pol λ, XRCC1, and gapped DNA within the microphases. Thus, stimulation of Pol λ activity is caused both by its interaction with XRCC1 and by specific conditions of microphase separation; this phenomenon is shown for the first time.
    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
    目标:在东亚,乳腺癌的发病率一直在快速增长,尤其是绝经前妇女。雌激素-DNA加合物的比例升高与乳腺癌的风险更高有关。本研究探讨了碱基切除修复(BER)基因多态性与雌激素-DNA加合物之间的相互作用对乳腺癌风险的影响。
    方法:我们进行了一项病例对照研究,包括健康志愿者和良性乳腺疾病患者(对照组,n=176)和浸润性癌或原位癌患者(病例臂,n=177)。BER相关基因的基因分型,包括SMUG1,OGG1,ERCC5和APEX1。逻辑回归模型,整合基因多态性之间的相互作用,雌激素-DNA加合物比,和临床变量,用于确定乳腺癌的危险因素。
    结果:单变量分析显示乳腺癌风险与APEX1rs1130409T>G(P=0.057)和APEX1rs1760944T>G(P=0.065)之间存在边缘关联。多变量回归分析显示,APEX1_rs1130409(GT/GG与TT)与雌激素-DNA加合物比的自然对数值(估计OR1.164,P=0.023)和绝经前状态与雌激素-DNA加合物比>2.93(估计OR2.433,P=0.001)的乳腺癌风险增加显着相关。
    结论:APEX1_rs1130409(GT/GG与TT)多态性,这与BER活动减少有关,加上雌激素-DNA加合物的比例增加,增加东亚女性患乳腺癌的风险。
    OBJECTIVE: In East Asia, the incidence of breast cancer has been increasing rapidly, particularly among premenopausal women. An elevated ratio of estrogen-DNA adducts was linked to a higher risk of breast cancer. The present study explored the influence of the interaction between base excision repair (BER) gene polymorphisms and estrogen-DNA adducts on breast cancer risk.
    METHODS: We conducted a case-control study comprising healthy volunteers and individuals with benign breast disease (control arm, n = 176) and patients with invasive carcinoma or carcinoma in situ (case arm, n = 177). Genotyping for BER-related genes, including SMUG1, OGG1, ERCC5, and APEX1, was performed. A logistic regression model, incorporating interactions between gene polymorphisms, estrogen-DNA adduct ratio, and clinical variables, was used to identify the risk factors for breast cancer.
    RESULTS: Univariate analysis indicated marginal associations between breast cancer risk and APEX1 rs1130409 T > G (P = 0.057) and APEX1 rs1760944 T > G (P = 0.065). Multivariate regression analysis revealed significant associations with increased breast cancer risk for APEX1_rs1130409 (GT/GG versus TT) combined with a natural logarithmic value of the estrogen-DNA adduct ratio (estimated OR 1.164, P = 0.023) and premenopausal status with an estrogen-DNA adduct ratio > 2.93 (estimated OR 2.433, P = 0.001).
    CONCLUSIONS: APEX1_rs1130409 (GT/GG versus TT) polymorphisms, which are related to decreased BER activity, combined with an increased ratio of estrogen-DNA adducts, increase the risk of breast cancer in East Asian women.
    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
    疟疾寄生虫的两个关键细胞器-每个细胞中的原生质体和线粒体-的基因组的完整性必须通过靶向这些区室的蛋白质介导的DNA修复来维持。我们探索了恶性疟原虫碱基切除修复(BER)DNAN-糖基化酶同源物PfEndoIII和PfOgg1的定位和功能。这些N-糖基化酶将在无嘌呤/无嘧啶(AP)核酸内切酶作用之前推定识别DNA损伤。Ape1和Apn1核酸内切酶先前已显示仅在寄生虫线粒体中起作用。免疫荧光定位显示PfEndoIII仅是线粒体。PfOgg1表达为PfOgg1前导-GFP融合蛋白时,在线粒体中没有清楚地看到,尽管染色质免疫沉淀实验表明它可以与线粒体和细胞质膜DNA相互作用。重组PfEndoIII在胸腺嘧啶乙二醇(Tg)病变上充当DNAN-糖基化酶和AP-裂解酶。我们使用伯氏疟原虫的反向遗传方法进一步研究了Ogg1在疟疾生命周期中的重要性。PbOgg1的靶向破坏导致8-氧代-G特异性DNA糖基化酶/裂解酶活性的丧失。PbOgg1基因敲除并不影响血液,蚊子或肝脏阶段发育,但在小鼠接种子孢子后导致血液阶段感染减少。还观察到PbOgg1敲除肝裂殖子的红细胞感染性显着降低,因此表明PbOgg1确保从肝脏到血液阶段感染的平稳过渡。我们的结果加强了疟原虫线粒体基因组是BER途径修复DNA的重要位点的观点。
    The integrity of genomes of the two crucial organelles of the malaria parasite - an apicoplast and mitochondrion in each cell - must be maintained by DNA repair mediated by proteins targeted to these compartments. We explored the localisation and function of Plasmodium falciparum base excision repair (BER) DNA N-glycosylase homologs PfEndoIII and PfOgg1. These N-glycosylases would putatively recognise DNA lesions prior to the action of apurinic/apyrimidinic (AP)-endonucleases. Both Ape1 and Apn1 endonucleases have earlier been shown to function solely in the parasite mitochondrion. Immunofluorescence localisation showed that PfEndoIII was exclusively mitochondrial. PfOgg1 was not seen clearly in mitochondria when expressed as a PfOgg1leader-GFP fusion, although chromatin immunoprecipitation assays showed that it could interact with both mitochondrial and apicoplast DNA. Recombinant PfEndoIII functioned as a DNA N-glycosylase as well as an AP-lyase on thymine glycol (Tg) lesions. We further studied the importance of Ogg1 in the malaria life cycle using reverse genetic approaches in Plasmodium berghei. Targeted disruption of PbOgg1 resulted in loss of 8-oxo-G specific DNA glycosylase/lyase activity. PbOgg1 knockout did not affect blood, mosquito or liver stage development but caused reduced blood stage infection after inoculation of sporozoites in mice. A significant reduction in erythrocyte infectivity by PbOgg1 knockout hepatic merozoites was also observed, thus showing that PbOgg1 ensures smooth transition from liver to blood stage infection. Our results strengthen the view that the Plasmodium mitochondrial genome is an important site for DNA repair by the BER pathway.
    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
    DNA糖基化酶通过催化从DNA中去除受损或错配的碱基来启动碱基切除修复(BER)途径。拟南芥DNA糖基化酶甲基-CpG结合域蛋白4样(MBD4L)是响应基因毒性剂5-氟尿嘧啶和5-溴尿嘧啶而触发BER的核酶。迄今为止,尚未分析MBD4L在植物生理过程中的参与。为了解决这个问题,我们研究了种子中的酶功能。我们发现吸胀诱导MBD4L基因表达通过产生两个替代转录本,MBD4L.3和MBD4L.4.老化种子的基因激活强于未老化种子。mbd4l-1突变体的种子在控制或老化条件下保持发芽失败,35S:MBD4L.3/mbd4l-1和35S:MBD4L.4/mbd4l-1种子逆转了这些表型。种子核DNA修复,通过彗星试验评估,在吸入后24小时以MBD4L依赖性方式加剧。在这种情况下,BER基因ARP,APE1L,LIG1在35S:MBD4L.3/mbd4l-1和35S:MBD4L.4/mbd4l-1种子中的表达高于mbd4l-1种子中的表达,表明这些成分可以与MBD4L协调以修复种子中受损的DNA碱基。有趣的是,ATM,ATR,与DNA损伤反应(DDR)途径相关的BRCA1,RAD51和WEE1基因在mbd4l-1中被激活,但在35S:MBD4L.3/mbd4l-1或35S:MBD4L.4/mbd4l-1种子中未被激活。这些结果表明,MBD4L是在种子吸胀过程中运作的BER级联的关键酶,其缺陷会导致DDR检测到的基因组损伤,产生发芽延迟或减少。
    DNA glycosylases initiate the base excision repair (BER) pathway by catalyzing the removal of damaged or mismatched bases from DNA. The Arabidopsis DNA glycosylase methyl-CpG-binding domain protein 4 like (MBD4L) is a nuclear enzyme triggering BER in response to the genotoxic agents 5-fluorouracil and 5-bromouracil. To date, the involvement of MBD4L in plant physiological processes has not been analyzed. To address this, we studied the enzyme functions in seeds. We found that imbibition induced the MBD4L gene expression by generating two alternative transcripts, MBD4L.3 and MBD4L.4. Gene activation was stronger in aged than in non-aged seeds. Seeds from mbd4l-1 mutants displayed germination failures when maintained under control or ageing conditions, while 35S:MBD4L.3/mbd4l-1 and 35S:MBD4L.4/mbd4l-1 seeds reversed these phenotypes. Seed nuclear DNA repair, assessed by comet assays, was exacerbated in an MBD4L-dependent manner at 24 h post-imbibition. Under this condition, the BER genes ARP, APE1L, and LIG1 showed higher expression in 35S:MBD4L.3/mbd4l-1 and 35S:MBD4L.4/mbd4l-1 than in mbd4l-1 seeds, suggesting that these components could coordinate with MBD4L to repair damaged DNA bases in seeds. Interestingly, the ATM, ATR, BRCA1, RAD51, and WEE1 genes associated with the DNA damage response (DDR) pathway were activated in mbd4l-1, but not in 35S:MBD4L.3/mbd4l-1 or 35S:MBD4L.4/mbd4l-1 seeds. These results indicate that MBD4L is a key enzyme of a BER cascade that operates during seed imbibition, whose deficiency would cause genomic damage detected by DDR, generating a delay or reduction in germination.
    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
    线粒体DNA(mtDNA)位于线粒体基质中,靠近细胞中活性氧(ROS)的主要来源。这使得mtDNA成为细胞中最容易受到损伤的成分之一。核因子E2相关因子2/抗氧化反应元件(Nrf2/ARE)信号通路是重要的细胞保护机制。研究表明,Nrf2可以调节细胞中线粒体靶向抗氧化系统的表达,间接保护mtDNA免受损伤。然而,Nrf2/ARE途径也可以直接影响mtDNA修复过程。在这次审查中,我们总结了Nrf2对mtDNA修复影响的现有数据,主要是碱基切除修复(BER),因为它被认为是线粒体基因组的主要修复途径。我们探索Nrf2/ARE之间的串扰,BRCA1和p53信号通路参与维持mtDNA完整性。讨论了其他修复机制在纠正错配碱基和双链断裂中的作用。此外,这篇综述讨论了Nrf2在修复非规范碱基中的作用,这导致mtDNA突变数量增加,并可能污染核苷酸库。
    Mitochondrial DNA (mtDNA) is located in the mitochondrial matrix, in close proximity to major sources of reactive oxygen species (ROS) in the cell. This makes mtDNA one of the most susceptible components to damage in the cell. The nuclear factor E2-related factor 2/antioxidant response element (Nrf2/ARE) signaling pathway is an important cytoprotective mechanism. It is well-studied and described that Nrf2 can regulate the expression of mitochondrial-targeted antioxidant systems in the cell, indirectly protecting mtDNA from damage. However, the Nrf2/ARE pathway can also directly impact on the mtDNA repair processes. In this review, we summarize the existing data on the impact of Nrf2 on mtDNA repair, primarily base excision repair (BER), as it is considered the main repair pathway for the mitochondrial genome. We explore the crosstalk between Nrf2/ARE, BRCA1, and p53 signaling pathways in their involvement in maintaining mtDNA integrity. The role of other repair mechanisms in correcting mismatched bases and double-strand breaks is discussed. Additionally, the review addresses the role of Nrf2 in the repair of noncanonical bases, which contribute to an increased number of mutations in mtDNA and can contaminate the nucleotide pool.
    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
    异柠檬酸脱氢酶亚型1(IDH1)的突变主要在继发性胶质母细胞瘤(GBM)和低度神经胶质瘤中发现,但在原发性GBM中很少见。GBM的标准治疗包括放疗联合替莫唑胺,烷化剂。幸运的是,IDH1突变胶质瘤对这种治疗敏感,导致更有利的预后。然而,据估计,随着时间的推移,高达75%的IDH1突变神经胶质瘤将进展至WHOIV级,并对烷化剂产生耐药性.因此,了解IDH1突变神经胶质瘤赋予烷化剂敏感性的机制对于开发靶向化疗方法至关重要.碱基切除修复(BER)途径负责修复由烷化剂诱导的大多数碱基损伤。由于未解决的DNA损伤,该途径中的缺陷可导致对这些试剂的超敏反应。BER蛋白质复合物的协调组装和分解对于细胞存活和在烷化剂暴露后维持基因组完整性至关重要。这些复合物依赖于聚-ADP-核糖的形成,在BER过程中由PARP1和PARP2合成的NAD+依赖性翻译后修饰。在病变部位,聚ADP-核糖促进XRCC1的募集。这种支架蛋白有助于组装BER蛋白,如DNA聚合酶β(Polβ),含有DNA合成和5'-脱氧核糖-磷酸裂解酶(5'dRP裂解酶)活性的双功能DNA聚合酶。这里,我们证实IDH1突变的胶质瘤细胞有缺陷的NAD+代谢,但仍产生足够的核NAD,以响应DNA损伤而进行强大的PARP1激活和BER复合物形成。然而,2-羟基戊二酸的过量生产,由IDH1R132H突变蛋白产生的代谢产物,通过降低Polβ蛋白水平来抑制BER容量。这定义了一种新的机制,通过该机制,神经胶质瘤中的IDH1突变赋予细胞对烷化剂和聚ADP-核糖糖水解酶抑制剂的敏感性。帕格。
    Mutations in isocitrate dehydrogenase isoform 1 (IDH1) are primarily found in secondary glioblastoma (GBM) and low-grade glioma but are rare in primary GBM. The standard treatment for GBM includes radiation combined with temozolomide, an alkylating agent. Fortunately, IDH1 mutant gliomas are sensitive to this treatment, resulting in a more favorable prognosis. However, it\'s estimated that up to 75 % of IDH1 mutant gliomas will progress to WHO grade IV over time and develop resistance to alkylating agents. Therefore, understanding the mechanism(s) by which IDH1 mutant gliomas confer sensitivity to alkylating agents is crucial for developing targeted chemotherapeutic approaches. The base excision repair (BER) pathway is responsible for repairing most base damage induced by alkylating agents. Defects in this pathway can lead to hypersensitivity to these agents due to unresolved DNA damage. The coordinated assembly and disassembly of BER protein complexes are essential for cell survival and for maintaining genomic integrity following alkylating agent exposure. These complexes rely on poly-ADP-ribose formation, an NAD+-dependent post-translational modification synthesized by PARP1 and PARP2 during the BER process. At the lesion site, poly-ADP-ribose facilitates the recruitment of XRCC1. This scaffold protein helps assemble BER proteins like DNA polymerase beta (Polβ), a bifunctional DNA polymerase containing both DNA synthesis and 5\'-deoxyribose-phosphate lyase (5\'dRP lyase) activity. Here, we confirm that IDH1 mutant glioma cells have defective NAD+ metabolism, but still produce sufficient nuclear NAD+ for robust PARP1 activation and BER complex formation in response to DNA damage. However, the overproduction of 2-hydroxyglutarate, an oncometabolite produced by the IDH1 R132H mutant protein, suppresses BER capacity by reducing Polβ protein levels. This defines a novel mechanism by which the IDH1 mutation in gliomas confers cellular sensitivity to alkylating agents and to inhibitors of the poly-ADP-ribose glycohydrolase, PARG.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号