PCNA monoubiquitination

  • 文章类型: Journal Article
    范可尼贫血(FA)修复途径控制着高度基因毒性的DNA链间交联(ICL)的修复,并依赖于跨损伤合成(TLS)。通过REV1或在赖氨酸164(K164)处的增殖细胞核抗原(PCNA)(PCNA-Ub)的位点特异性单泛素化来促进TLS。PcnaK164R/K164R而不是Rev1-/-突变使哺乳动物对ICL过敏。除了FA途径,替代途径与ICL修复相关(1,2),尽管两者之间的决策仍然难以捉摸。为了研究PCNA-Ub在FA修复中的依赖性和相关性,我们交叉了PcnaK164R/+;Fancg-/+小鼠。发现组合突变(PcnaK164R/K164R;Fancg-/-)是胚胎致死的。原代双突变(DM)小鼠胚胎成纤维细胞(MEF)的RNA-seq显示复制应激诱导的检查点水平升高。为了排除压力诱发的混杂因素,我们利用Trp53敲除法获得了一个模型来深入研究ICL修复.关于ICL诱导的细胞毒性,细胞周期停滞,和复制叉进展,发现单突变型和DMMEFs同样敏感,建立PCNA-Ub对FA-ICL修复至关重要。免疫沉淀和基于光谱的分析揭示了PCNA-Ub在排除错配识别复合物MSH2/MSH6被募集到ICL中的未知作用。总之,我们的结果揭示了PCNA-Ub在ICL修复中的双重功能,即排除MSH2/MSH6募集,将ICL引导至规范FA修复,除了在协调与未连接的ICL相对的TLS方面的既定作用。
    The Fanconi anemia (FA) repair pathway governs repair of highly genotoxic DNA interstrand crosslinks (ICLs) and relies on translesion synthesis (TLS). TLS is facilitated by REV1 or site-specific monoubiquitination of proliferating cell nuclear antigen (PCNA) (PCNA-Ub) at lysine 164 (K164). A PcnaK164R/K164R but not Rev1-/- mutation renders mammals hypersensitive to ICLs. Besides the FA pathway, alternative pathways have been associated with ICL repair (1, 2), though the decision making between those remains elusive. To study the dependence and relevance of PCNA-Ub in FA repair, we intercrossed PcnaK164R/+; Fancg-/+ mice. A combined mutation (PcnaK164R/K164R; Fancg-/- ) was found embryonically lethal. RNA-seq of primary double-mutant (DM) mouse embryonic fibroblasts (MEFs) revealed elevated levels of replication stress-induced checkpoints. To exclude stress-induced confounders, we utilized a Trp53 knock-down to obtain a model to study ICL repair in depth. Regarding ICL-induced cell toxicity, cell cycle arrest, and replication fork progression, single-mutant and DM MEFs were found equally sensitive, establishing PCNA-Ub to be critical for FA-ICL repair. Immunoprecipitation and spectrometry-based analysis revealed an unknown role of PCNA-Ub in excluding mismatch recognition complex MSH2/MSH6 from being recruited to ICLs. In conclusion, our results uncovered a dual function of PCNA-Ub in ICL repair, i.e. exclude MSH2/MSH6 recruitment to channel the ICL toward canonical FA repair, in addition to its established role in coordinating TLS opposite the unhooked ICL.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Published Erratum
    [这更正了文章DOI:10.3389/fimmu.202.871766。].
    [This corrects the article DOI: 10.3389/fimmu.2022.871766.].
    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
    免疫球蛋白(Ig)基因的体细胞超突变(SHM)是在针对外源抗原的免疫应答成熟期间产生特异性和高亲和力抗体所需的B细胞特异性过程。此过程取决于活化诱导的胞苷脱氨酶(AID)和几种DNA修复因子的活性。AID依赖性SHM在G/C和A/T碱基上均匀分布的Ig可变(V)区中产生突变的全谱。在大多数哺乳动物细胞中,S期脱氧胞苷脱氨基成尿嘧啶诱导靶向G/C诱变,使用尿嘧啶的直接复制或TLS介导的旁路,然而,只有激活的B淋巴细胞的机制才能在AID产生的尿嘧啶周围产生A/T诱变。后者背后的分子机制迄今仍未完全理解。然而,缺乏能够再现G/C和A/T突变谱的细胞模型构成了阐明它的主要障碍。到目前为止,用于研究IgSHM的少数可用B细胞系确实主要经历G/C突变,使它们不合适或用途有限。在这份报告中,我们表明,在Ramos细胞系中,在培养中经历了组成型G/C偏向SHM,A/T突变率低是由于PCNA的泛素化/去泛素化反应失衡,去泛素化反应占主导地位。去泛素酶复合物USP1-UAF1的抑制或泛素与PCNA的组成型融合的表达提供了DNA聚合酶η募集以及此后在IgV基因多样化过程中引入A/T碱基对(bp)突变所需的缺失线索。本研究报道了第一个修饰的人B细胞系的建立,该细胞系在体外概括了Ig基因的SHM机制。
    Somatic hypermutation (SHM) of immunoglobulin (Ig) genes is a B cell specific process required for the generation of specific and high affinity antibodies during the maturation of the immune response against foreign antigens. This process depends on the activity of both activation-induced cytidine deaminase (AID) and several DNA repair factors. AID-dependent SHM creates the full spectrum of mutations in Ig variable (V) regions equally distributed at G/C and A/T bases. In most mammalian cells, deamination of deoxycytidine into uracil during S phase induces targeted G/C mutagenesis using either direct replication of uracils or TLS mediated bypass, however only the machinery of activated B lymphocytes can generate A/T mutagenesis around AID-created uracils. The molecular mechanism behind the latter remains incompletely understood to date. However, the lack of a cellular model that reproduces both G/C and A/T mutation spectra constitutes the major hurdle to elucidating it. The few available B cell lines used thus far to study Ig SHM indeed undergo mainly G/C mutations, that make them inappropriate or of limited use. In this report, we show that in the Ramos cell line that undergoes constitutive G/C-biased SHM in culture, the low rate of A/T mutations is due to an imbalance in the ubiquitination/deubiquitination reaction of PCNA, with the deubiquitination reaction being predominant. The inhibition of the deubiquitinase complex USP1-UAF1 or the expression of constitutive fusion of ubiquitin to PCNA provides the missing clue required for DNA polymerase η recruitment and thereafter the introduction of A/T base pair (bp) mutations during the process of IgV gene diversification. This study reports the establishment of the first modified human B cell line that recapitulates the mechanism of SHM of Ig genes in vitro.
    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 damage tolerance permits bypass of DNA lesions encountered during S-phase and may be carried out by translesion DNA synthesis (TLS). Human TLS requires selective monoubiquitination of proliferating cell nuclear antigen (PCNA) sliding clamps encircling damaged DNA. This posttranslational modification (PTM) is catalyzed by Rad6/Rad18. Recent studies revealed that replication protein A (RPA), the major ssDNA-binding protein, is involved in the regulation of PCNA monoubiquitination and interacts directly with Rad18 on chromatin and in the nucleoplasm. However, it is unclear how RPA regulates this critical PTM and what functional role(s) these interactions serve. Here, we developed an in vitro assay to quantitatively monitor PCNA monoubiquitination under in vivo scenarios. Results from extensive experiments revealed that RPA regulates Rad6/Rad18 activity in an ssDNA-dependent manner. We found that \"DNA-free\" RPA inhibits monoubiquitination of free PCNA by directly interacting with Rad18. This interaction is promoted under native conditions when there is an overabundance of free RPA in the nucleoplasm where Rad6/Rad18 and a significant fraction of PCNA reside. During DNA replication stress, RPA binds the ssDNA exposed downstream of stalled primer/template (P/T) junctions, releasing Rad6/Rad18. RPA restricted the resident PCNAs to the upstream duplex regions by physically blocking diffusion of PCNA along ssDNA, and this activity was required for efficient monoubiquitination of PCNA on DNA. Furthermore, upon binding ssDNA, RPA underwent a conformational change that increased its affinity for Rad18. Rad6/Rad18 complexed with ssDNA-bound RPA was active, and this interaction may selectively promote monoubiquitination of PCNA on long RPA-coated ssDNA.
    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
    Translesion DNA synthesis (TLS) during S-phase uses specialized TLS DNA polymerases to replicate a DNA lesion, allowing stringent DNA synthesis to resume beyond the offending damage. Human TLS involves the conjugation of ubiquitin to PCNA clamps encircling damaged DNA and the role of this post-translational modification is under scrutiny. A widely-accepted model purports that ubiquitinated PCNA recruits TLS polymerases such as pol η to sites of DNA damage where they may also displace a blocked replicative polymerase. We provide extensive quantitative evidence that the binding of pol η to PCNA and the ensuing TLS are both independent of PCNA ubiquitination. Rather, the unique properties of pols η and δ are attuned to promote an efficient and passive exchange of polymerases during TLS on the lagging strand.
    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