RAD18

Rad18
  • 文章类型: Journal Article
    在过去的几十年里,肿瘤干细胞(CSC)已成为与肿瘤复发和化疗耐药相关的关键肿瘤细胞亚群.了解CSC介导的化学耐药的潜在机制对于改善癌症治疗结果至关重要。这项研究探讨了DNA糖基化酶NEIL1的调节作用,卵巢CSC的化学耐药性。我们首先观察到NEIL1在卵巢CSC中的表达降低,表明其可能参与CSC监管。使用泛癌症分析,我们证实与正常组织相比,卵巢肿瘤中NEIL1的表达减少.此外,NEIL1下调与干性标志物的增加和CSC的富集相关,强调其在调节CSC表型中的作用。进一步的机制研究显示,卵巢CSC中NEIL1和RAD18表达呈负相关。NEIL1耗尽导致RAD18表达升高,可能通过增强TranslesionDNA合成(TLS)介导的DNA损伤旁路来促进化学耐药性。此外,NEIL1的dowregulation导致卵巢癌中DNA损伤积累减少和细胞凋亡抑制。总的来说,我们的发现揭示了NEIL1和RAD18参与调节卵巢CSC化疗耐药的新机制.靶向这个NEIL1-RAD18轴可能为对抗化疗耐药和改善卵巢癌治疗结果提供有希望的治疗策略。
    Over the past decades, cancer stem cells (CSCs) have emerged as a critical subset of tumor cells associated with tumor recurrence and resistance to chemotherapy. Understanding the mechanisms underlying CSC-mediated chemoresistance is imperative for improving cancer therapy outcomes. This study delves into the regulatory role of NEIL1, a DNA glycosylase, in chemoresistance in ovarian CSCs. We first observed a decreased expression of NEIL1 in ovarian CSCs, suggesting its potential involvement in CSC regulation. Using pan-cancer analysis, we confirmed the diminished NEIL1 expression in ovarian tumors compared to normal tissues. Furthermore, NEIL1 downregulation correlated with an increase in stemness markers and enrichment of CSCs, highlighting its role in modulating CSC phenotype. Further mechanistic investigation revealed an inverse correlation between NEIL1 and RAD18 expression in ovarian CSCs. NEIL1 depletion led to heightened RAD18 expression, promoting chemoresistance possibly via enhancing Translesion DNA Synthesis (TLS)-mediated DNA lesion bypass. Moreover, dowregulation of NEIL1 results in reduced DNA damage accumulation and suppressed apoptosis in ovarian cancer. Overall, our findings unveil a novel mechanism involving NEIL1 and RAD18 in regulating chemoresistance in ovarian CSCs. Targeting this NEIL1-RAD18 axis may offer promising therapeutic strategies for combating chemoresistance and improving ovarian cancer treatment outcomes.
    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
    致癌RAS和RAF信号与胰腺癌和甲状腺癌的放射抗性有关。在这项研究中,我们试图更好地阐明导致这种效应的分子机制.我们发现miRNA296-3p(miR-296-3p)与一组胰腺癌细胞的放射敏感性显着相关,miR-296-3p在正常细胞中高表达,但癌细胞系低。miR-296-3p的表达升高会增加放射增敏作用,同时降低胰腺癌和甲状腺癌细胞中DNA修复酶RAD18的表达。与匹配的正常对照相比,RAD18在胰腺和甲状腺肿瘤中过表达,RAD18在肿瘤中的高表达与不良预后特征相关。调节胰腺癌细胞中突变型KRAS或甲状腺癌细胞中突变型BRAF的表达证明了两种癌症类型中RAD18表达的严格调节。RAD18的耗尽导致DNA损伤和辐射诱导的细胞死亡。重要的是,在KRAS突变型胰腺癌原位肿瘤和BRAF突变型甲状腺异位肿瘤中,RAD18耗竭联合放疗可导致明显和持续的肿瘤消退。总的来说,我们的发现在胰腺癌和甲状腺癌细胞中发现了一个新的协同RAS/RAF-miR-296-3p-RAD18信号网络,这导致增强的辐射抗性。
    Oncogenic RAS and RAF signaling has been implicated in contributing to radioresistance in pancreatic and thyroid cancers. In this study, we sought to better clarify molecular mechanisms contributing to this effect. We discovered that miRNA 296-3p (miR-296-3p) is significantly correlated with radiosensitivity in a panel of pancreatic cancer cells, and miR-296-3p is highly expressed in normal cells, but low in cancer cell lines. Elevated expression of miR-296-3p increases radiosensitization while decreasing the expression of the DNA repair enzyme RAD18 in both pancreatic and thyroid cancer cells. RAD18 is overexpressed in both pancreatic and thyroid tumors compared to matched normal controls, and high expression of RAD18 in tumors is associated with poor prognostic features. Modulating the expression of mutant KRAS in pancreatic cancer cells or mutant BRAF in thyroid cancer cells demonstrates a tight regulation of RAD18 expression in both cancer types. Depletion of RAD18 results in DNA damage and radiation-induced cell death. Importantly, RAD18 depletion in combination with radiotherapy results in marked and sustained tumor regression in KRAS mutant pancreatic cancer orthotopic tumors and BRAF mutant thyroid heterotopic tumors. Overall, our findings identify a novel coordinated RAS/RAF-miR-296-3p-RAD18 signaling network in pancreatic and thyroid cancer cells, which leads to enhanced radioresistance.
    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
    复制叉失速时,叉后形成的RPA包被的单链DNA(ssDNA)激活共济失调毛细血管扩张突变和Rad3相关(ATR)激酶,同时启动依赖Rad18的PCNA单项化。然而,这两个事件之间是否存在串扰以及这种相互作用的潜在生理意义仍然难以捉摸。在这项研究中,我们证明了在复制应激期间,ATR在Ser403上磷酸化人Rad18,Ser403是Rad18中先前未鉴定的PIP基序(PCNA相互作用肽)的相邻残基。这个磷酸化事件破坏了Rad18和PCNA之间的相互作用,从而限制了Rad18介导的PCNA单尿素化的程度。因此,肿瘤抑制蛋白SLX4的过度积累,现在被描述为泛素化PCNA的新读者,在停滞的叉子被阻止,有助于防止停滞的叉子倒塌。我们进一步确定,ATR通过限制Rad18介导的PCNA单尿素化和端粒上过度的SLX4积累,在端粒(ALT)细胞的替代延长中保持端粒稳定性。这些发现揭示了ATR激活之间复杂的相互作用,Rad18依赖的PCNA单数化,和SLX4相关的停滞叉处理,强调ATR在保持复制叉稳定性和促进端粒酶非依赖性端粒维持方面的关键作用。
    Upon replication fork stalling, the RPA-coated single-stranded DNA (ssDNA) formed behind the fork activates the ataxia telangiectasia-mutated and Rad3-related (ATR) kinase, concomitantly initiating Rad18-dependent monoubiquitination of PCNA. However, whether crosstalk exists between these two events and the underlying physiological implications of this interplay remain elusive. In this study, we demonstrate that during replication stress, ATR phosphorylates human Rad18 at Ser403, an adjacent residue to a previously unidentified PIP motif (PCNA-interacting peptide) within Rad18. This phosphorylation event disrupts the interaction between Rad18 and PCNA, thereby restricting the extent of Rad18-mediated PCNA monoubiquitination. Consequently, excessive accumulation of the tumor suppressor protein SLX4, now characterized as a novel reader of ubiquitinated PCNA, at stalled forks is prevented, contributing to the prevention of stalled fork collapse. We further establish that ATR preserves telomere stability in alternative lengthening of telomere (ALT) cells by restricting Rad18-mediated PCNA monoubiquitination and excessive SLX4 accumulation at telomeres. These findings shed light on the complex interplay between ATR activation, Rad18-dependent PCNA monoubiquitination, and SLX4-associated stalled fork processing, emphasizing the critical role of ATR in preserving replication fork stability and facilitating telomerase-independent telomere maintenance.
    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
    MAGEA4是一种癌症-睾丸抗原,主要在睾丸中表达,但在几种癌症中异常过度表达。MAGEA4与RING泛素连接酶RAD18相互作用并激活跨损伤DNA合成(TLS),可能有利于肿瘤进化。这里,我们使用NMR和AlphaFold2(AF)阐明了RAD18和MAGEA4之间的相互作用模式,并揭示了RAD18的RAD6结合域(R6BD)在MAGEA4的C端翼状螺旋亚结构域中占据了一个凹槽。我们发现MAGEA4部分取代RAD18R6BD的RAD6并抑制降解性RAD18自动尿素化,这可以通过RAD18R6BD的竞争肽来抵消。AlphaFold2和交联质谱(XL-MS)还揭示了催化RING与RAD18的DNA结合SAP结构域之间的进化不变的分子内相互作用,这对于PCNA单泛素化至关重要。利用相互作用蛋白质组学,我们发现另一个I型MAGE,MAGE-C2以类似于MAGEA4/RAD18复合物的方式与RING泛素连接酶TRIM28相互作用,提示MAGEA4肽结合沟在其他I型MAGE中也充当连接酶结合裂隙。我们的数据为RAD18介导的PCNA单泛素化的机制和调控提供了新的见解。
    MAGEA4 is a cancer-testis antigen primarily expressed in the testes but aberrantly overexpressed in several cancers. MAGEA4 interacts with the RING ubiquitin ligase RAD18 and activates trans-lesion DNA synthesis (TLS), potentially favouring tumour evolution. Here, we employed NMR and AlphaFold2 (AF) to elucidate the interaction mode between RAD18 and MAGEA4, and reveal that the RAD6-binding domain (R6BD) of RAD18 occupies a groove in the C-terminal winged-helix subdomain of MAGEA4. We found that MAGEA4 partially displaces RAD6 from the RAD18 R6BD and inhibits degradative RAD18 autoubiquitination, which could be countered by a competing peptide of the RAD18 R6BD. AlphaFold2 and cross-linking mass spectrometry (XL-MS) also revealed an evolutionary invariant intramolecular interaction between the catalytic RING and the DNA-binding SAP domains of RAD18, which is essential for PCNA mono-ubiquitination. Using interaction proteomics, we found that another Type-I MAGE, MAGE-C2, interacts with the RING ubiquitin ligase TRIM28 in a manner similar to the MAGEA4/RAD18 complex, suggesting that the MAGEA4 peptide-binding groove also serves as a ligase-binding cleft in other type-I MAGEs. Our data provide new insights into the mechanism and regulation of RAD18-mediated PCNA mono-ubiquitination.
    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
    作为对抗多种恶性肿瘤的关键防线,自然杀伤(NK)细胞存在于肿瘤微环境(TME)中。据报道,RAD18E3泛素蛋白连接酶(RAD18)可促进多种癌症的恶性进展,但其对NK功能的影响尚未被挖掘。这里,本研究旨在挖掘RAD18调节NK细胞对大肠癌(CRC)细胞杀伤作用的机制.E2F转录因子7(E2F7)和RAD18在CRC组织中的表达,它们的相关性,结合位点,并对RAD18富集途径进行生物信息学分析。通过qRT-PCR和蛋白质印迹测定细胞中E2F7和RAD18的表达。双荧光素酶测定和染色质免疫沉淀(ChIP)测定验证了E2F7和RAD18之间的调控关系。CCK-8测定法用于测定细胞活力,集落形成试验检测细胞增殖,乳酸脱氢酶(LDH)测试以测定NK细胞的细胞毒性,ELISA测定粒细胞-巨噬细胞集落刺激因子(GM-CSF)的水平,肿瘤坏死因子-α(TNF-α)和干扰素-γ(IFN-γ),和免疫荧光检测毒性分子穿孔素和颗粒酶B的表达。在CRC组织和细胞中发现RAD18和E2F7的高表达。沉默RAD18可能会阻碍CRC细胞的增殖,培养NK细胞的活力和细胞毒性,增加GM-CSF的分泌,TNF-α,IFN-γ以及穿孔素和颗粒酶B的表达。ChIP和双荧光素酶报告基因测定确定了RAD18启动子区和E2F7之间的结合关系。E2F7可以激活RAD18的转录,沉默RAD18可以逆转E2F7过表达对NK细胞杀伤的抑制作用。这项工作阐明了E2F7/RAD18轴对CRC中NK细胞杀伤的抑制作用。并为靶向免疫微环境中的CRC免疫治疗提供了新的方向。
    As a pivotal defensive line against multitudinous malignant tumors, natural killer (NK) cells exist in the tumor microenvironment (TME). RAD18 E3 Ubiquitin Protein Ligase (RAD18) has been reported to foster the malignant progression of multiple cancers, but its effect on NK function has not been mined. Here, the study was designed to mine the mechanism by which RAD18 regulates the killing effect of NK cells on colorectal cancer (CRC) cells. Expression of E2F Transcription Factor 7 (E2F7) and RAD18 in CRC tissues, their correlation, binding sites, and RAD18 enrichment pathway were analyzed by bioinformatics. Expression of E2F7 and RAD18 in cells was assayed by qRT-PCR and western blot. Dual-luciferase assay and chromatin immunoprecipitation (ChIP) assay verified the regulatory relationship between E2F7 and RAD18. CCK-8 assay was utilized to assay cell viability, colony formation assay to detect cell proliferation, lactate dehydrogenase (LDH) test to assay NK cell cytotoxicity, ELISA to assay levels of granulocyte-macrophage colony-stimulating factor (GM-CSF), tumor necrosis factor-α (TNF-α) and interferon-γ (IFN-γ), and immunofluorescence to detect expression of toxic molecules perforin and granzyme B. High expression of RAD18 and E2F7 was found in CRC tissues and cells. Silencing RAD18 could hamper the proliferation of CRC cells, foster viability and cytotoxicity of NK cells, and increase the secretion of GM-CSF, TNF-α, IFN-γ as well as the expression of perforin and granzyme B. Additionally, ChIP and dual-luciferase reporter assay ascertained the binding relationship between RAD18 promoter region and E2F7. E2F7 could activate the transcription of RAD18, and silencing RAD18 reversed the inhibitory effect of E2F7 overexpression on NK cell killing. This work clarified the inhibitory effect of the E2F7/RAD18 axis on NK cell killing in CRC, and proffered a new direction for immunotherapy of CRC in targeted immune microenvironment.
    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损伤耐受性(DDT)途径通过挽救停滞在DNA损伤或其他屏障处的复制叉来减轻复制过程中DNA损伤的影响,并修复新复制的DNA中留下的不连续性。从酵母到哺乳动物细胞,RAD18调节的跨病变合成(TLS)和模板转换(TS)代表DDT的主要途径。通过E2/E3蛋白对HRAD6A-B/RAD18对聚合酶滑动钳PCNA进行单倍体化,能够募集可以插入与受损模板碱基相对的核苷酸的专门TLS聚合酶。或者,随后通过Ubc13-Mms2(E2)和HLTF或SHPRH(E3)对单亲素-PCNA进行多泛素化,可以导致合成从受损的模板转换为未受损的新合成的姐妹链,以促进经过病变的合成。当无法立即发生TLS或TS时,缺口可能保留在新合成的链中,部分是由于PRIMPOL引发酶的再灌注活性,可以在细胞周期的后期填充。本综述的第一部分将总结有关RAD18依赖性滴滴涕途径的当前知识,而第二部分将提供用于鉴定和表征DDT蛋白的细胞功能的分子工具包。特别是,我们将专注于先进的技术,可以揭示单链和双链DNA缺口及其在单细胞水平上的修复,以及监测单复制叉的进展,例如特定版本的DNA纤维和彗星测定。这些方法的集合可以作为一个强大的分子工具包来监测间隙的代谢,检测相关途径和分子参与者的贡献,以及表征潜在抑制剂的有效性。
    DNA damage tolerance (DDT) pathways mitigate the effects of DNA damage during replication by rescuing the replication fork stalled at a DNA lesion or other barriers and also repair discontinuities left in the newly replicated DNA. From yeast to mammalian cells, RAD18-regulated translesion synthesis (TLS) and template switching (TS) represent the dominant pathways of DDT. Monoubiquitylation of the polymerase sliding clamp PCNA by HRAD6A-B/RAD18, an E2/E3 protein pair, enables the recruitment of specialized TLS polymerases that can insert nucleotides opposite damaged template bases. Alternatively, the subsequent polyubiquitylation of monoubiquitin-PCNA by Ubc13-Mms2 (E2) and HLTF or SHPRH (E3) can lead to the switching of the synthesis from the damaged template to the undamaged newly synthesized sister strand to facilitate synthesis past the lesion. When immediate TLS or TS cannot occur, gaps may remain in the newly synthesized strand, partly due to the repriming activity of the PRIMPOL primase, which can be filled during the later phases of the cell cycle. The first part of this review will summarize the current knowledge about RAD18-dependent DDT pathways, while the second part will offer a molecular toolkit for the identification and characterization of the cellular functions of a DDT protein. In particular, we will focus on advanced techniques that can reveal single-stranded and double-stranded DNA gaps and their repair at the single-cell level as well as monitor the progression of single replication forks, such as the specific versions of the DNA fiber and comet assays. This collection of methods may serve as a powerful molecular toolkit to monitor the metabolism of gaps, detect the contribution of relevant pathways and molecular players, as well as characterize the effectiveness of potential inhibitors.
    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
    Rad18与SMC5/6定位因子1(SLF1)相互作用,将SMC5/6复合物募集到DNA损伤位点进行修复。SLF1对Rad18特异性识别的机制尚不清楚。这里,我们介绍了SLF1(SLF1tBRCT)中与相互作用的Rad18肽结合的串联BRCT重复序列(tBRCT)的晶体结构。我们的结构和生化研究表明,SLF1tBRCT与Rad18中的两个磷酸丝氨酸和相邻残基相互作用,以实现高亲和力和特异性的Rad18识别。我们发现SLF1tBRCT利用tBRCT之间常见的机制以及Rad18结合的独特机制,后者包括与Rad18中的α-螺旋结构的相互作用,这在其他tBRCT结合的配体蛋白中未观察到。我们的工作提供了对SLF1靶向Rad18的结构见解,并扩展了对BRCT介导的复合物组装的理解。
    Rad18 interacts with the SMC5/6 localization factor 1 (SLF1) to recruit the SMC5/6 complex to DNA damage sites for repair. The mechanism of the specific Rad18 recognition by SLF1 is unclear. Here, we present the crystal structure of the tandem BRCT repeat (tBRCT) in SLF1 (SLF1tBRCT) bound with the interacting Rad18 peptide. Our structure and biochemical studies demonstrate that SLF1tBRCT interacts with two phosphoserines and adjacent residues in Rad18 for high-affinity and specificity Rad18 recognition. We found that SLF1tBRCT utilizes mechanisms common among tBRCTs as well as unique ones for Rad18 binding, the latter include interactions with an α-helical structure in Rad18 that has not been observed in other tBRCT-bound ligand proteins. Our work provides structural insights into Rad18 targeting by SLF1 and expands the understanding of BRCT-mediated complex assembly.
    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
    丝裂霉素C(MMC)治疗后,范可尼贫血(FA)途径的激活对于防止称为“放射状”的染色体易位至关重要。“当复制叉在MMC诱导的链间交联(ICL)处停滞时,FA途径被激活以协调ICL解钩和DNA断裂中间体的修复。然而,在缺乏FA的细胞中,与ICL相关的中断如何以导致放射状的方式解决尚不清楚。这里,我们证明MMC诱导的放射依赖于DNA聚合酶θ(POLθ)介导的替代末端连接(A-EJ)。具体来说,我们表明,在FANCD2-/-细胞中观察到的放射状依赖于POLθ和DNA连接酶III,并且独立于经典的非同源末端连接而发生。此外,用POLθ抑制剂处理FANCD2-/-细胞可以消除放射状,并导致与常见脆性位点共定位的断裂积累。均匀,这些观察结果暗示A-EJ参与了放射状形成,并为使用POLθ抑制剂治疗FA途径缺陷型癌症提供了机制见解。
    Activation of the Fanconi anemia (FA) pathway after treatment with mitomycin C (MMC) is essential for preventing chromosome translocations termed \"radials.\" When replication forks stall at MMC-induced interstrand crosslinks (ICLs), the FA pathway is activated to orchestrate ICL unhooking and repair of the DNA break intermediates. However, in FA-deficient cells, how ICL-associated breaks are resolved in a manner that leads to radials is unclear. Here, we demonstrate that MMC-induced radials are dependent on DNA polymerase theta (POLθ)-mediated alternative end joining (A-EJ). Specifically, we show that radials observed in FANCD2-/- cells are dependent on POLθ and DNA ligase III and occur independently of classical non-homologous end joining. Furthermore, treatment of FANCD2-/- cells with POLθ inhibitors abolishes radials and leads to the accumulation of breaks co-localizing with common fragile sites. Uniformly, these observations implicate A-EJ in radial formation and provide mechanistic insights into the treatment of FA pathway-deficient cancers with POLθ inhibitors.
    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损伤耐受性(DDT)是真核生物绕过复制阻断损伤以恢复DNA合成并维持细胞活力的机制。在酿酒酵母中,DDT是通过增殖细胞核抗原(PCNA,由POL30编码)在K164残基处。删除RAD5或RAD18,编码PCNA泛素化所需的两个泛素连接酶,导致严重的DNA损伤敏感性,其可以通过编码抑制不期望的同源重组的DNA解旋酶的SRS2的失活来拯救。在这项研究中,我们从rad5Δ细胞中分离出DNA损伤抗性突变体,发现其中一个含有pol30-A171D突变,它可以以srs2依赖性和PCNASumoylation非依赖性方式挽救rad5Δ和rad18ΔDNA损伤敏感性。Pol30-A171D消除了与Srs2的物理相互作用,但没有另一种PCNA相互作用蛋白Rad30;然而,Pol30-A171不位于PCNA-Srs2界面中。分析了PCNA-Srs2结构,以设计并在复杂界面中产生突变,其中之一,pol30-I128A,导致表型让人想起pol30-A171D。这项研究使我们得出结论,与其他PCNA结合蛋白不同,Srs2通过部分保守的基序与PCNA相互作用,并且可以通过PCNASumoylation加强相互作用,这将Srs2招募变成一个受监管的过程。重要性众所周知,出芽酵母PCNASumoylation可作为配体,通过其串联受体基序招募DNA解旋酶Srs2,以防止复制叉上不需要的同源重组(HR),一个被称为打捞HR的过程。这项研究揭示了详细的分子机制,其中组成型PCNA-PIP相互作用已适应调节事件。由于PCNA和Srs2在真核生物中高度保守,从酵母到人类,这项研究可能有助于研究类似的调节机制。
    DNA-damage tolerance (DDT) is a mechanism by which eukaryotes bypass replication-blocking lesions to resume DNA synthesis and maintain cell viability. In Saccharomyces cerevisiae, DDT is mediated by sequential ubiquitination and sumoylation of proliferating cell nuclear antigen (PCNA, encoded by POL30) at the K164 residue. Deletion of RAD5 or RAD18, encoding two ubiquitin ligases required for PCNA ubiquitination, results in severe DNA-damage sensitivity, which can be rescued by inactivation of SRS2 encoding a DNA helicase that inhibits undesired homologous recombination. In this study, we isolated DNA-damage resistant mutants from rad5Δ cells and found that one of them contained a pol30-A171D mutation, which could rescue both rad5Δ and rad18Δ DNA-damage sensitivity in a srs2-dependent and PCNA sumoylation-independent manner. Pol30-A171D abolished physical interaction with Srs2 but not another PCNA-interacting protein Rad30; however, Pol30-A171 is not located in the PCNA-Srs2 interface. The PCNA-Srs2 structure was analyzed to design and create mutations in the complex interface, one of which, pol30-I128A, resulted in phenotypes reminiscent of pol30-A171D. This study allows us to conclude that, unlike other PCNA-binding proteins, Srs2 interacts with PCNA through a partially conserved motif, and the interaction can be strengthened by PCNA sumoylation, which turns Srs2 recruitment into a regulated process. IMPORTANCE It is known that budding yeast PCNA sumoylation serves as a ligand to recruit a DNA helicase Srs2 through its tandem receptor motifs that prevent unwanted homologous recombination (HR) at replication forks, a process known as salvage HR. This study reveals detailed molecular mechanisms, in which constitutive PCNA-PIP interaction has been adapted to a regulatory event. Since both PCNA and Srs2 are highly conserved in eukaryotes, from yeast to human, this study may shed light to investigation of similar regulatory mechanisms.
    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
    Pluripotent stem cells (PSCs) have the potential to differentiate to any of the other organs. The genome DNA integrity of PSCs is maintained by a high level of transcription for a number of genes involved in DNA repair, cell cycle and apoptosis. However, it remains unclear how high the frequency of genetic mutation is and how these DNA repair factors function in PSCs. In this study, we employed Sup F assay for the measurement of mutation frequency after UV-C irradiation in induced pluripotent stem cells (iPSCs) as PSC models and neural progenitor cells (NPCs) were derived from iPSCs as differentiated cells. iPSCs and NPCs exhibited a lower mutation frequency compared with the original skin fibroblasts. In RNA-seq analysis, iPSCs and NPCs showed a high expression of RAD18, which is involved in trans-lesion synthesis (TLS) for the emergency tolerance system during the replication process of DNA. Although RAD18 is involved in both error free and error prone TLS in somatic cells, it still remains unknown the function of RAD18 in PSCs. In this study we depleted of the RAD18 by siRNA knockdown resulted in decreased frequency of mutation in iPSCs and NPCs. Our results will provide information on the genome maintenance machinery in PSCs.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号