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: 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
    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)

  • 文章类型: Journal Article
    碱基切除修复(BER)途径是由DNA糖基化酶启动的精确且通用的DNA修复机制。核酸内切酶VIII样1(NEIL1)是一种双功能糖基化酶/无碱基位点(AP)裂解酶,可切除受损的碱基并随后裂解磷酸二酯骨架。NEIL1能够识别和水解广泛的氧化诱导的碱基损伤和取代的环状片段化鸟嘌呤,包括黄曲霉毒素诱导的8,9-二氢-8-(2,6-二氨基-4-氧代-3,4-二氢嘧啶-5-基-甲酰氨基)-9-羟基黄曲霉毒素B1(AFB1-FapyGua)。由于NEIL1对这些和其他促诱变病变的保护作用,据推测,天然存在的NEIL1单核苷酸多态性(SNP)变异体可增加人类患黄曲霉毒素诱导的肝细胞癌(HCC)的风险.鉴于南亚人群的饮食黄曲霉毒素暴露和乙型肝炎病毒感染导致氧化应激的高水平,对该区域发生的NEIL1SNP变异的研究可能具有临床意义.在这项研究中,表达了NEIL1的最常见的南亚变体,纯化,功能特征。所有测试的变体在含有一系列氧化诱导的碱基损伤的高分子量DNA上表现出与野生型(wt)-NEIL1相似的活性和底物特异性。在含有位点特异性嘌呤/嘧啶(AP)位点的短寡脱氧核苷酸(17-mer)上,胸腺嘧啶乙二醇(ThyGly),或者AFB1-FapyGua,P206L-NEIL1在催化上与wt-NEIL1相当,而NEIL1变体Q67K和T278I在这些底物上的活性降低了约2倍。变体T103A与17聚体DNA的结合能力大大降低,限制随后的糖基化酶和裂解酶反应。与这一观察一致,无法测量T103A对含有ThyGly或AFB1-FapyGua的17聚体寡脱氧核苷酸的切除率。然而,T103A切除ThyGly的能力在较长的寡脱氧核苷酸(51-mer)上得到改善,与wt-NEIL1相比,活性降低约7倍。我们的研究表明,NEIL1变体T103A可能呈现一种在损伤识别方面受到限制的致病表型,潜在增加人类肝癌的风险。
    The base excision repair (BER) pathway is a precise and versatile mechanism of DNA repair that is initiated by DNA glycosylases. Endonuclease VIII-like 1 (NEIL1) is a bifunctional glycosylase/abasic site (AP) lyase that excises a damaged base and subsequently cleaves the phosphodiester backbone. NEIL1 is able to recognize and hydrolyze a broad range of oxidatively-induced base lesions and substituted ring-fragmented guanines, including aflatoxin-induced 8,9-dihydro-8-(2,6-diamino-4-oxo-3,4-dihydropyrimid-5-yl-formamido)-9-hydroxyaflatoxin B1 (AFB1-FapyGua). Due to NEIL1\'s protective role against these and other pro-mutagenic lesions, it was hypothesized that naturally occurring single nucleotide polymorphic (SNP) variants of NEIL1 could increase human risk for aflatoxin-induced hepatocellular carcinoma (HCC). Given that populations in South Asia experience high levels of dietary aflatoxin exposures and hepatitis B viral infections that induce oxidative stress, investigations on SNP variants of NEIL1 that occur in this region may have clinical implications. In this study, the most common South Asian variants of NEIL1 were expressed, purified, and functionally characterized. All tested variants exhibited activities and substrate specificities similar to wild type (wt)-NEIL1 on high-molecular weight DNA containing an array of oxidatively-induced base lesions. On short oligodeoxynucleotides (17-mers) containing either a site-specific apurinic/apyrimidinic (AP) site, thymine glycol (ThyGly), or AFB1-FapyGua, P206L-NEIL1 was catalytically comparable to wt-NEIL1, while the activities of NEIL1 variants Q67K and T278I on these substrates were ≈2-fold reduced. Variant T103A had a greatly diminished ability to bind to 17-mer DNAs, limiting the subsequent glycosylase and lyase reactions. Consistent with this observation, the rate of excision by T103A on 17-mer oligodeoxynucleotides containing ThyGly or AFB1-FapyGua could not be measured. However, the ability of T103A to excise ThyGly was improved on longer oligodeoxynucleotides (51-mers), with ≈7-fold reduced activity compared to wt-NEIL1. Our studies suggest that NEIL1 variant T103A may present a pathogenic phenotype that is limited in damage recognition, potentially increasing human risk for HCC.
    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:高迁移率族蛋白2(HMGB2)被认为是各种癌症预后不良的生物标志物。本研究旨在探讨HMGB2在胶质瘤中的作用及其机制。
    方法:使用神经胶质瘤相关的在线数据库和我们当地的医院数据库,对HMGB2表达差异进行Kaplan-Meier生存分析和COX回归分析。对临床病理特征及影像学参数与HMGB2表达的相关性进行分析。然后进行GSEA和PPI网络以找出最重要的途径。该通路抑制剂用于验证HMGB2的参与。CCK8,EDU检测,采用γ-H2AX免疫荧光染色和集落形成实验观察对脑胶质瘤细胞的影响。
    结果:可用的数据集显示HMGB2在神经胶质瘤中高表达,高表达HMGB2的患者预后和分子特征较差。来自我们中心的蛋白质印迹和免疫组织化学的蛋白质水平证据支持上述结论。对影像学特征的分析表明,HMGB2表达水平与ADCmean呈负相关,但与增强边缘的厚度呈正相关。GSEA和PPI网络分析结果显示HMGB2参与碱基切除修复(BER)信号通路。实验证据表明,HMGB2的过表达促进了神经胶质瘤细胞的增殖并增强了放射抗性。
    结论:HMGB2能促进胶质瘤的发展,增强胶质瘤细胞的放射抗性,可能与BER途径有关,提示它可能作为神经胶质瘤患者的潜在生物标志物。
    BACKGROUND: High mobility group box 2 (HMGB2) is considered as a biomarker of poor prognosis in various cancers.This study aims to investigate the effect and mechanism of HMGB2 in gliomas.
    METHODS: With the glioma related on-line and our local hospital databases, the expression differences of HMGB2,Kaplan-Meier survival analysis and COX regression analysis were performed.The correlation analysis between the clinicopathological features and imaging parameters with the HMGB2 expression had been done. Then GSEA and PPI networks were carried out to find out the most significant pathway. The pathway inhibitor was applied to verify HMGB2\'s participation. CCK8,EDU assays,γ-H2AX immunofluorescence staining and colony formation assay were conducted to observe effects on glioma cells.
    RESULTS: Available datasets showed that HMGB2 was highly expressed in glioma and patients with high expression of HMGB2 had poorer prognosis and molecular characteristics. Protein level evidence of western blot and immunohistochemistry from our center supported the conclusions above. Analysis on imaging features suggested that HMGB2 expression level had an inverse association with ADCmean but positively with the thickness of enhancing margin. Results from GSEA and PPI network analysis exhibited that HMGB2 was involved in base excision repair (BER) signaling pathway. Experimental evidence demonstrated that the overexpression of HMGB2 promoted the proliferation of glioma cells and enhanced the radio-resistance.
    CONCLUSIONS: HMGB2 could promote glioma development and enhance the radioresistance of glioma cells, potentially related to the BER pathway, suggesting it may serve as an underlying biomarker for patients with glioma.
    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
    碱基切除修复(BER)需要通过蛋白质-蛋白质相互作用在修复酶之间进行紧密协调,并涉及通过DNA聚合酶(pol)β填充缺口,然后在下游步骤通过DNA连接酶(LIG)1或LIGIIIα进行缺口密封。核酸内切酶1(APE1),通过它的外切核酸酶活性,校对3'-在BER期间由polβ合并的不匹配。我们先前报道过,polβ和BER连接酶之间的功能相互作用中断会导致错误的修复事件。然而,LIG1和LIGIIIα的蛋白质相互作用如何在切口密封的最后步骤中影响修复途径的协调仍然未知。这里,我们证明LIGIIIα与polβ和APE1的相互作用比LIG1更紧密,LIG1的N端非催化区以及LIGIIIα的催化核心和BRCT结构域介导与两种蛋白质的相互作用。我们的结果表明,在不存在LIGIIIαZnF结构域和LIG1N末端区域的情况下,polβ核苷酸插入产物的缺口密封效率较低。此外,我们显示了APE1和LIG1/LIGIIIα之间的协调,从缺口修复中间体中去除3'-错配,在该缺口修复中间体上,两种BER连接酶都可以密封非规范末端或缺口修复中间体,从而导致单缺失诱变产物。总体结果表明,在APE1校对的存在下,通过polβ与LIG1/LIGIIIα进行缺口密封耦合的缺口填充功能协调的重要性,这主要由蛋白质-蛋白质相互作用和蛋白质-DNA中间通讯控制,在BER通路的下游步骤保持修复效率。
    Base excision repair (BER) requires a tight coordination between the repair enzymes through protein-protein interactions and involves gap filling by DNA polymerase (pol) β and subsequent nick sealing by DNA ligase (LIG) 1 or LIGIIIα at the downstream steps. Apurinic/apyrimidinic-endonuclease 1 (APE1), by its exonuclease activity, proofreads 3\' mismatches incorporated by polβ during BER. We previously reported that the interruptions in the functional interplay between polβ and the BER ligases result in faulty repair events. Yet, how the protein interactions of LIG1 and LIGIIIα could affect the repair pathway coordination during nick sealing at the final steps remains unknown. Here, we demonstrate that LIGIIIα interacts more tightly with polβ and APE1 than LIG1, and the N-terminal noncatalytic region of LIG1 as well as the catalytic core and BRCT domain of LIGIIIα mediate interactions with both proteins. Our results demonstrated less efficient nick sealing of polβ nucleotide insertion products in the absence of LIGIIIα zinc-finger domain and LIG1 N-terminal region. Furthermore, we showed a coordination between APE1 and LIG1/LIGIIIα during the removal of 3\' mismatches from the nick repair intermediate on which both BER ligases can seal noncanonical ends or gap repair intermediate leading to products of single deletion mutagenesis. Overall results demonstrate the importance of functional coordination from gap filling by polβ coupled to nick sealing by LIG1/LIGIIIα in the presence of proofreading by APE1, which is mainly governed by protein-protein interactions and protein-DNA intermediate communications, to maintain repair efficiency at the downstream steps of the BER 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
    头颈部鳞状细胞癌(HNSCC)是由不同的外源性危险因素引起的,包括吸烟,酒精消费,和HPV感染。碱基切除修复(BER)是修复氧化性DNA损伤的前线,它由DNAN-糖基化酶蛋白(OGG1)和其他BER因子(包括DNA聚合酶β(POLB))启动。
    探索HNSCC中的BER基因(OGG1,POLB)过表达是否会改变基因组完整性,免疫原性,及其在预后价值中的作用。
    RNA测序(RNA-Seq)和临床信息(年龄,性别,组织学分级,生存状态,和阶段)从癌症基因组图谱中检索到530例HNSCC患者。将患者数据分类为HPV阳性或阴性,以分析包括肿瘤分期在内的肿瘤数据,POLB,和OGG1基因表达。
    检索的HNSCC数据的RNA-Seq,并使用非配对t检验比较突变计数和非整倍性评分。TIMER算法用于计算6种浸润免疫细胞(CD4+T细胞,CD8+T细胞,B细胞,中性粒细胞,巨噬细胞,和树突状细胞)基于RNA-Seq表达谱数据。POLB之间的相关性,使用TIMER2.0通过Spearman相关性分析计算OGG1和免疫细胞。
    我们的数据分析表明,BER基因在HNSCC肿瘤中经常过表达并增加突变计数。此外,OGG1和POLB过表达与免疫细胞的低浸润有关,低免疫检查点基因表达(PD-1,细胞毒性T淋巴细胞抗原4,程序死亡配体1和程序死亡配体2),和先天免疫信号基因。此外,在人乳头瘤病毒(HPV)阳性肿瘤中,BER因子失调有较好的总生存期.
    我们的分析表明,BER基因组的失调可能是HNSCC癌症中潜在的预后标志物和/或免疫检查点阻断的有吸引力的靶标。然而,我们的观察仍需要进一步的基于实验的科学验证研究.
    UNASSIGNED: Head and neck squamous carcinoma (HNSCC) is caused by different exogenous risk factors including smoking cigarettes, alcohol consumption, and HPV infection. Base excision repair (BER) is the frontline to repair oxidative DNA damage, which is initiated by the DNA N-glycosylase proteins (OGG1) and other BER factors including DNA polymerase β (POLB).
    UNASSIGNED: Explore whether BER genes\' (OGG1, POLB) overexpression in HNSCC alters genomic integrity, immunogenicity, and its role in prognostic value.
    UNASSIGNED: RNA sequencing (RNA-Seq) and clinical information (age, gender, histological grade, survival status, and stage) of 530 patients of HNSCC were retrieved from the Cancer Genome Atlas. Patients\' data are categorized HPV positive or negative to analyze the tumor data including the tumor stage, POLB, and OGG1 gene expression.
    UNASSIGNED: RNA-Seq of HNSCC data retrieved and mutation count and aneuploidy score were compared using an unpaired t-test. The TIMER algorithm was used to calculate the tumor abundance of six infiltrating immune cells (CD4+ T cells, CD8+ T cells, B cells, neutrophils, macrophages, and dendritic cells) based on RNA-Seq expression profile data. The correlation between the POLB, OGG1, and immune cells was calculated by Spearman correlation analysis using TIMER 2.0.
    UNASSIGNED: Our data analysis reveals that BER genes frequently overexpressed in HNSCC tumors and increase mutation count. In addition, OGG1 and POLB overexpression are associated with low infiltration of immune cells, low immune checkpoint gene expression (PD-1, cytotoxic T-lymphocyte antigen 4, program death ligand 1, and program death ligand 2), and innate immune signaling genes. Furthermore, dysregulated BER factors in Human papillomavirus (HPV) positive tumors had better overall survival.
    UNASSIGNED: Our analysis suggests that dysregulation of the BER genes panel might be a potential prognosis marker and/or an attractive target for an immune checkpoint blockade in HNSCC cancers. However, our observation still requires further experimental-based scientific validation studies.
    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
    DNA修复途径在基因组稳定性中起关键作用,但是在真核细胞中,它们必须在染色质的紧凑和纠结的环境中修复DNA损伤。以前的研究表明,将DNA包装成核小体,它们构成了染色质的基本组成部分,对DNA修复有着深远的影响。在这次审查中,我们讨论了染色质中DNA修复的原理和机制。我们专注于组蛋白翻译后修饰(PTM)在修复中的作用,以及组蛋白突变体影响细胞对DNA损伤剂的敏感性和染色质修复活性的分子机制。重要的是,这些机制被认为会显著影响人类癌症的体细胞突变率,并可能导致癌症发生和其他人类疾病。例如,许多主要在酵母中研究的组蛋白突变体已被确定为不同癌症中的候选癌组蛋白突变。这篇综述强调了这些联系,并讨论了染色质中DNA修复对人类健康的潜在重要性。
    DNA repair pathways play a critical role in genome stability, but in eukaryotic cells, they must operate to repair DNA lesions in the compact and tangled environment of chromatin. Previous studies have shown that the packaging of DNA into nucleosomes, which form the basic building block of chromatin, has a profound impact on DNA repair. In this review, we discuss the principles and mechanisms governing DNA repair in chromatin. We focus on the role of histone post-translational modifications (PTMs) in repair, as well as the molecular mechanisms by which histone mutants affect cellular sensitivity to DNA damage agents and repair activity in chromatin. Importantly, these mechanisms are thought to significantly impact somatic mutation rates in human cancers and potentially contribute to carcinogenesis and other human diseases. For example, a number of the histone mutants studied primarily in yeast have been identified as candidate oncohistone mutations in different cancers. This review highlights these connections and discusses the potential importance of DNA repair in chromatin to human health.
    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
    核酸内切酶VIII样3(NEIL3)是一种多功能的DNA糖基化酶,可修复对DNA的各种化学修饰。与其他糖基化酶不同,NEIL3对单链DNA和单/双链DNA连接处的损伤具有偏好。除了其在氧化DNA碱基切除修复中的典型作用外,NEIL3启动复制依赖性链间DNA交联修复作为范可尼贫血途径的替代方法。这篇综述概述了我们目前对NEIL3生物学功能的理解,在疾病中的作用,和三维结构,因为它涉及底物特异性和催化机理。
    Endonuclease VIII-like 3 (NEIL3) is a versatile DNA glycosylase that repairs a diverse array of chemical modifications to DNA. Unlike other glycosylases, NEIL3 has a preference for lesions within single-strand DNA and at single/double-strand DNA junctions. Beyond its canonical role in base excision repair of oxidized DNA, NEIL3 initiates replication-dependent interstrand DNA crosslink repair as an alternative to the Fanconi Anemia pathway. This review outlines our current understanding of NEIL3\'s biological functions, role in disease, and three-dimensional structure as it pertains to substrate specificity and catalytic mechanism.
    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
    DNA聚合酶β(Polβ)的基因敲除(KO)小鼠模型显示,Polβ的丢失会导致新生儿死亡,强调了这种DNA聚合酶的关键有机作用。虽然生化分析和基因KO细胞系已经证实了其在碱基切除修复和TET介导的去甲基化中的生化作用,继续开发更长寿的小鼠模型,以进一步定义其生物作用。Polb-KO小鼠是Cre介导的组织特异性KO小鼠模型中的第一个。该技术用于研究Polβ在V(D)J重组(可变多样性连接重排)中的作用,DNA去甲基化,基因互补,SPO11诱导的DNA双链断裂修复,生殖细胞基因组稳定性,以及神经元分化,对基因毒素诱导的DNA损伤的敏感性,和癌症发作。通过直接在C57BL/6受精卵中进行CRISPR/cas9介导的基因编辑,使敲入(KI)小鼠模型的革命成为可能。该技术有助于鉴定与Pol3的种系或体细胞突变体相关的表型。这种KI小鼠模型有助于揭示关键Polβ活性位点残基或特定Polβ酶活性的重要性。例如出现狼疮症状的PolbY265C小鼠。最近,我们已经使用这种KI技术突变了具有两个密码子变化的Polb基因,生成PolbL301R/V303R鼠标。在这个KI小鼠模型中,表达的Polβ蛋白不能与其专性异二聚体伴侣结合,Xrcc1。尽管表达的突变体Polβ蛋白在蛋白水解上不稳定,并且在募集到DNA损伤位点时存在缺陷,纯合子PolbL301R/V303R小鼠是有活力和有生育能力的,但身材矮小。我们期望正在开发的这种和其他靶向小鼠模型有望揭示Polβ的新生物学和生物作用。
    Gene knock-out (KO) mouse models for DNA polymerase beta (Polβ) revealed that loss of Polβ leads to neonatal lethality, highlighting the critical organismic role for this DNA polymerase. While biochemical analysis and gene KO cell lines have confirmed its biochemical role in base excision repair and in TET-mediated demethylation, more long-lived mouse models continue to be developed to further define its organismic role. The Polb-KO mouse was the first of the Cre-mediated tissue-specific KO mouse models. This technology was exploited to investigate roles for Polβ in V(D)J recombination (variable-diversity-joining rearrangement), DNA demethylation, gene complementation, SPO11-induced DNA double-strand break repair, germ cell genome stability, as well as neuronal differentiation, susceptibility to genotoxin-induced DNA damage, and cancer onset. The revolution in knock-in (KI) mouse models was made possible by CRISPR/cas9-mediated gene editing directly in C57BL/6 zygotes. This technology has helped identify phenotypes associated with germline or somatic mutants of Polβ. Such KI mouse models have helped uncover the importance of key Polβ active site residues or specific Polβ enzyme activities, such as the PolbY265C mouse that develops lupus symptoms. More recently, we have used this KI technology to mutate the Polb gene with two codon changes, yielding the PolbL301R/V303R mouse. In this KI mouse model, the expressed Polβ protein cannot bind to its obligate heterodimer partner, Xrcc1. Although the expressed mutant Polβ protein is proteolytically unstable and defective in recruitment to sites of DNA damage, the homozygous PolbL301R/V303R mouse is viable and fertile, yet small in stature. We expect that this and additional targeted mouse models under development are poised to reveal new biological and organismic roles for Polβ.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号