Cdc20 Proteins

Cdc20 蛋白质类
  • 文章类型: Journal Article
    靶向CDC20可以增强肿瘤细胞的放射敏感性,但CDC20对DNA毁伤修复反响的感化和机制依然隐约。为了研究这个问题,肿瘤细胞系,包括KYSE200,KYSE450和HCT116,用于检测表达,函数,以及CDC20在放射性化疗耐药中的潜在机制。Westernblot和免疫荧光染色证实CDC20的表达和定位。辐射可以上调细胞核中CDC20的表达。利用同源重组(HR)和非同源末端连接(NHEJ)报告基因系统探索CDC20对DNA损伤修复的影响。表明CDC20可以促进HR修复和放射/化疗抵抗。在DNA损伤的早期阶段,CDC20通过蛋白质-蛋白质相互作用稳定RPA1蛋白,激活ATR介导的信号级联,从而帮助基因组修复。在后期阶段,CDC20通过泛素介导的RPA1降解辅助损伤修复的后续步骤。CCK-8和集落形成实验检测CDC20在细胞活力和增殖中的作用,和靶向CDC20可以加剧由顺铂或依托泊苷引起的DNA损伤水平的增加。在BALB/c-nu/nu小鼠中进行肿瘤异种移植模型以证实CDC20在体内的功能。确认体外结果。总之,本研究通过揭示CDC20在DNA损伤修复过程中调节RPA1的新作用,进一步验证了CDC20作为克服放射化疗耐药的策略的潜在临床意义.
    Targeting CDC20 can enhance the radiosensitivity of tumor cells, but the function and mechanism of CDC20 on DNA damage repair response remains vague. To examine that issue, tumor cell lines, including KYSE200, KYSE450, and HCT116, were utilized to detect the expression, function, and underlying mechanism of CDC20 in radio-chemoresistance. Western blot and immunofluorescence staining were employed to confirm CDC20 expression and location, and radiation could upregulate the expression of CDC20 in the cell nucleus. The homologous recombination (HR) and non-homologous end joining (NHEJ) reporter gene systems were utilized to explore the impact of CDC20 on DNA damage repair, indicating that CDC20 could promote HR repair and radio/chemo-resistance. In the early stages of DNA damage, CDC20 stabilizes the RPA1 protein through protein-protein interactions, activating the ATR-mediated signaling cascade, thereby aiding in genomic repair. In the later stages, CDC20 assists in the subsequent steps of damage repair by the ubiquitin-mediated degradation of RPA1. CCK-8 and colony formation assay were used to detect the function of CDC20 in cell vitality and proliferation, and targeting CDC20 can exacerbate the increase in DNA damage levels caused by cisplatin or etoposide. A tumor xenograft model was conducted in BALB/c-nu/nu mice to confirm the function of CDC20 in vivo, confirming the in vitro results. In conclusion, this study provides further validation of the potential clinical significance of CDC20 as a strategy to overcome radio-chemoresistance via uncovering a novel role of CDC20 in regulating RPA1 during DNA damage repair.
    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
    早期胚胎的有丝分裂通常以快速的速度进行,但是这个速度是如何实现的还不清楚。这里,我们发现细胞周期蛋白B3是秀丽隐杆线虫胚胎中快速胚胎有丝分裂的主要驱动因素。细胞周期蛋白B1和B2支持缓慢的有丝分裂(NEBD至后期~600s),但是细胞周期蛋白B3的存在主要驱动了野生型中观察到的大约三倍更快的有丝分裂。在细胞周期蛋白B1和B2驱动的有丝分裂中,多个有丝分裂事件减慢,细胞周期蛋白B3相关的Cdk1H1激酶活性比细胞周期蛋白B1相关的Cdk1活性高25倍。将细胞周期蛋白B1添加到仅快速细胞周期蛋白B3的有丝分裂中,会在完成染色体排列和后期开始之间引入〜60s延迟;这种延迟,这对隔离保真度很重要,依赖于后期激活剂Cdc20的抑制性磷酸化。因此,细胞周期蛋白B3优势,与通过Cdc20磷酸化起作用的细胞周期蛋白B1依赖性延迟相结合,在秀丽隐杆线虫早期胚胎中设置快速的步伐并确保有丝分裂的保真度。
    Mitosis in early embryos often proceeds at a rapid pace, but how this pace is achieved is not understood. Here, we show that cyclin B3 is the dominant driver of rapid embryonic mitoses in the C. elegans embryo. Cyclins B1 and B2 support slow mitosis (NEBD to anaphase ∼600 s), but the presence of cyclin B3 dominantly drives the approximately threefold faster mitosis observed in wildtype. Multiple mitotic events are slowed down in cyclin B1 and B2-driven mitosis, and cyclin B3-associated Cdk1 H1 kinase activity is ∼25-fold more active than cyclin B1-associated Cdk1. Addition of cyclin B1 to fast cyclin B3-only mitosis introduces an ∼60-s delay between completion of chromosome alignment and anaphase onset; this delay, which is important for segregation fidelity, is dependent on inhibitory phosphorylation of the anaphase activator Cdc20. Thus, cyclin B3 dominance, coupled to a cyclin B1-dependent delay that acts via Cdc20 phosphorylation, sets the rapid pace and ensures mitotic fidelity in the early C. elegans embryo.
    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
    紫杉醇在临床相关剂量下诱导多极纺锤体,但基本上不增加有丝分裂指数。假设紫杉醇的抗癌作用是通过促进多极纺锤体上的染色体错误分离导致细胞凋亡而发生的。子细胞中的坏死和干扰素基因的环状GMP-AMP合成酶刺激因子(cGAS-STING)途径激活,导致I型干扰素(IFN)分泌和免疫原性细胞死亡。据报道,依瑞布林和长春瑞滨也会导致癌细胞中多极纺锤体的增加。最近,据报道,使用CRISPR/Cas9诱变抑制后期促进复合物/环状体细胞分裂周期20(APC/C-CDC20)活性可增加对Kinesin家族18a(KIF18a)抑制的敏感性,其功能是抑制癌细胞中的多极有丝分裂纺锤体。我们提出了一种增强增加多极纺锤体的抗癌剂有效性的方法是通过抑制APC/C-CDC20来延迟,但不能阻止,后期输入。延迟基因组不稳定细胞的后期进入可能会增强多极纺锤体诱导的细胞死亡。在基因组稳定的健康人类细胞中,延迟的后期进入可能会抑制抗癌药物诱导的多极纺锤体水平,并降低有丝分裂的细胞毒性。我们概述了分子的特定组合,以研究可以实现增强抗癌剂有效性的目标。
    Paclitaxel induces multipolar spindles at clinically relevant doses but does not substantially increase mitotic indices. Paclitaxel\'s anti-cancer effects are hypothesized to occur by promoting chromosome mis-segregation on multipolar spindles leading to apoptosis, necrosis and cyclic-GMP-AMP Synthase-Stimulator of Interferon Genes (cGAS-STING) pathway activation in daughter cells, leading to secretion of type I interferon (IFN) and immunogenic cell death. Eribulin and vinorelbine have also been reported to cause increases in multipolar spindles in cancer cells. Recently, suppression of Anaphase-Promoting Complex/Cyclosome-Cell Division Cycle 20 (APC/C-CDC20) activity using CRISPR/Cas9 mutagenesis has been reported to increase sensitivity to Kinesin Family 18a (KIF18a) inhibition, which functions to suppress multipolar mitotic spindles in cancer cells. We propose that a way to enhance the effectiveness of anti-cancer agents that increase multipolar spindles is by suppressing the APC/C-CDC20 to delay, but not block, anaphase entry. Delaying anaphase entry in genomically unstable cells may enhance multipolar spindle-induced cell death. In genomically stable healthy human cells, delayed anaphase entry may suppress the level of multipolar spindles induced by anti-cancer drugs and lower mitotic cytotoxicity. We outline specific combinations of molecules to investigate that may achieve the goal of enhancing the effectiveness of anti-cancer agents.
    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
    结肠腺癌(COAD)是导致癌症死亡的第二大原因,仍然缺乏诊断生物标志物和治疗靶点。在这项研究中,使用TCGA数据库的生物信息学分析来获得RUNX1,这是一个在COAD中具有预后价值的基因。RUNX1在许多恶性肿瘤中起重要作用,及其在COAD中的分子调控机制仍有待充分理解。为了探索RUNX1的生理作用,我们进行了功能分析,如CCK-8,集落形成和迁移测定。此外,我们使用转录组测序和染色质免疫沉淀分析研究了潜在的机制.RUNX1在COAD患者中高表达,与生存率显著相关。沉默RUNX1显著减慢了COAD细胞的增殖和迁移能力。此外,我们证明CDC20和MCM2可能是RUNX1的靶基因,并且RUNX1可能与去泛素化酶USP31物理连接,后者介导RUNX1蛋白的上调以促进转录功能.我们的结果可能为RUNX1在COAD中的作用机制提供新的见解,并揭示该疾病的潜在治疗靶标。
    Colon adenocarcinoma (COAD) is the second leading cause of cancer death, and there is still a lack of diagnostic biomarkers and therapeutic targets. In this study, bioinformatics analysis of the TCGA database was used to obtain RUNX1, a gene with prognostic value in COAD. RUNX1 plays an important role in many malignancies, and its molecular regulatory mechanisms in COAD remain to be fully understood. To explore the physiological role of RUNX1, we performed functional analyses, such as CCK-8, colony formation and migration assays. In addition, we investigated the underlying mechanisms using transcriptome sequencing and chromatin immunoprecipitation assays. RUNX1 is highly expressed in COAD patients and significantly correlates with survival. Silencing of RUNX1 significantly slowed down the proliferation and migratory capacity of COAD cells. Furthermore, we demonstrate that CDC20 and MCM2 may be target genes of RUNX1, and that RUNX1 may be physically linked to the deubiquitinating enzyme USP31, which mediates the upregulation of RUNX1 protein to promote transcriptional function. Our results may provide new insights into the mechanism of action of RUNX1 in COAD and reveal potential therapeutic targets for this disease.
    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:子宫内膜癌(EC)是一种常见的妇科恶性肿瘤,其特点是发病率和死亡率不断增加。这强调了对新型治疗靶标的关键需求。一个这样的潜在目标是细胞分裂周期20(CDC20),这与肿瘤发生有关。这项研究调查了CDC20抑制剂Apcin对EC的作用,并阐明了所涉及的潜在机制。
    方法:Apcin对EC细胞增殖的影响,凋亡,使用CCK8测定和流式细胞术评估细胞周期。随后进行RNA测序(RNA-seq)以探索潜在的分子机制,随后进行Western印迹和免疫共沉淀以验证结果。进行动物研究以评估体内抗肿瘤作用。还进行了生物信息学分析以鉴定CDC20作为EC中的潜在治疗靶标。
    结果:用Apcin处理抑制EC细胞增殖并诱导其凋亡,导致细胞周期停滞。用Apcin处理后,与凋亡和细胞周期相关的途径被激活。值得注意的是,Apcin处理导致细胞周期调节因子p21上调,其被证实与CDC20相互作用并因此降低EC细胞中下游细胞周期蛋白的表达。体内实验证实Apcin治疗显著阻碍肿瘤生长。在EC组织中观察到比在非恶性组织中更高的CDC20表达,EC患者CDC20表达增加与总生存期和无进展间期缩短相关。
    结论:CDC20是一种新型的EC分子靶标,Apcin可作为治疗EC的候选抗肿瘤药物。
    OBJECTIVE: Endometrial carcinoma (EC) is a prevalent gynecological malignancy characterized by increasing incidence and mortality rates. This underscores the critical need for novel therapeutic targets. One such potential target is cell division cycle 20 (CDC20), which has been implicated in oncogenesis. This study investigated the effect of the CDC20 inhibitor Apcin on EC and elucidated the underlying mechanism involved.
    METHODS: The effects of Apcin on EC cell proliferation, apoptosis, and the cell cycle were evaluated using CCK8 assays and flow cytometry. RNA sequencing (RNA-seq) was subsequently conducted to explore the underlying molecular mechanism, and Western blotting and coimmunoprecipitation were subsequently performed to validate the results. Animal studies were performed to evaluate the antitumor effects in vivo. Bioinformatics analysis was also conducted to identify CDC20 as a potential therapeutic target in EC.
    RESULTS: Treatment with Apcin inhibited proliferation and induced apoptosis in EC cells, resulting in cell cycle arrest. Pathways associated with apoptosis and the cell cycle were activated following treatment with Apcin. Notably, Apcin treatment led to the upregulation of the cell cycle regulator p21, which was verified to interact with CDC20 and consequently decrease the expression of downstream cyclins in EC cells. In vivo experiments confirmed that Apcin treatment significantly impeded tumor growth. Higher CDC20 expression was observed in EC tissue than in nonmalignant tissue, and increased CDC20 expression in EC patients was associated with shorter overall survival and progress free interval.
    CONCLUSIONS: CDC20 is a novel molecular target in EC, and Apcin could be developed as a candidate antitumor drug for EC treatment.
    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
    O-连接的β-N-乙酰葡糖胺(O-GlcNAc)转移酶(OGT)是催化细胞内所有O-GlcNAc酰化反应的唯一酶。先前的研究发现,OGT水平在细胞分裂过程中会振荡。具体来说,OGT丰度在有丝分裂期间下调,但是缺乏潜在的机制。在这里,我们证明OGT被泛素E3连接酶泛素化,后期促进复合物/环小体(APC/C)-细胞分裂周期20(Cdc20)。我们表明APC/CCdc20通过保守的破坏盒(D盒)与OGT相互作用:Arg-351/Leu-354,其废除使OGT稳定。由于APC/CCdc20-底物结合通常在引发泛素化事件之前,我们还使用了质谱,并将OGTLys-352映射为泛素化位点,这是OGT与APC/C亚基关联的先决条件。有趣的是,在癌症基因组图谱中,R351C是子宫癌突变体,表明D-box的突变与肿瘤发生有关。矛盾的是,我们发现R351C和D-box突变体(R351A/L354A)在小鼠异种移植模型中抑制子宫癌,可能是由于细胞分裂和增殖受损。总之,我们提出了一个模型,其中OGTLys-352泛素化引发其与APC/C的结合,然后APC/CCdc20通过D-box与OGT合作进行有丝分裂破坏。我们的工作不仅突出了细胞周期中调节OGT的关键机制,而且还揭示了糖基化与细胞分裂机制之间的相互协调。
    O-linked β-N-acetylglucosamine (O-GlcNAc) transferase (OGT) is the sole enzyme that catalyzes all O-GlcNAcylation reactions intracellularly. Previous investigations have found that OGT levels oscillate during the cell division process. Specifically, OGT abundance is downregulated during mitosis, but the underlying mechanism is lacking. Here we demonstrate that OGT is ubiquitinated by the ubiquitin E3 ligase, anaphase promoting complex/cyclosome (APC/C)-cell division cycle 20 (Cdc20). We show that APC/CCdc20 interacts with OGT through a conserved destruction box (D-box): Arg-351/Leu-354, the abrogation of which stabilizes OGT. As APC/CCdc20-substrate binding is often preceded by a priming ubiquitination event, we also used mass spectrometry and mapped OGT Lys-352 to be a ubiquitination site, which is a prerequisite for OGT association with APC/C subunits. Interestingly, in The Cancer Genome Atlas, R351C is a uterine carcinoma mutant, suggesting that mutations of the D-box are linked with tumorigenesis. Paradoxically, we found that both R351C and the D-box mutants (R351A/L354A) inhibit uterine carcinoma in mouse xenograft models, probably due to impaired cell division and proliferation. In sum, we propose a model where OGT Lys-352 ubiquitination primes its binding with APC/C, and then APC/CCdc20 partners with OGT through the D-box for its mitotic destruction. Our work not only highlights the key mechanism that regulates OGT during the cell cycle, but also reveals the mutual coordination between glycosylation and the cell division machinery.
    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
    干扰有丝分裂中的微管动力学会激活纺锤体组装检查点(SAC),以防止染色体分离错误。SAC通过经由有丝分裂检查点复合物(MCC)抑制后期促进复合物(APC)来诱导有丝分裂阻滞。MCC成分MAD2中和关键的APC辅因子,CDC20,防止退出有丝分裂。延长有丝分裂阻滞可促进线粒体凋亡和caspase激活。然而,有丝分裂细胞死亡对体内组织稳态的影响尚不明确。通过条件性MAD2过表达,我们观察到慢性SAC激活引发小鼠骨髓发育不全和肠萎缩。虽然骨髓抑制可以得到补偿,胃肠萎缩是有害的。值得注意的是,促凋亡Bim/Bcl2l11的缺失可预防胃肠道综合征,虽然失去Noxa/Pmaip或共同删除Bid和Puma/Bbc3都没有这种保护作用,鉴定BIM作为胃肠上皮有丝分裂细胞死亡的限速凋亡效应。相比之下,仅过度表达抗凋亡BCL2,但没有上述仅BH3蛋白缺陷,可以减轻骨髓抑制。我们的发现强调了体内对SAC扰动的组织和细胞类型特异性存活依赖性。
    Interference with microtubule dynamics in mitosis activates the spindle assembly checkpoint (SAC) to prevent chromosome segregation errors. The SAC induces mitotic arrest by inhibiting the anaphase-promoting complex (APC) via the mitotic checkpoint complex (MCC). The MCC component MAD2 neutralizes the critical APC cofactor, CDC20, preventing exit from mitosis. Extended mitotic arrest can promote mitochondrial apoptosis and caspase activation. However, the impact of mitotic cell death on tissue homeostasis in vivo is ill-defined. By conditional MAD2 overexpression, we observe that chronic SAC activation triggers bone marrow aplasia and intestinal atrophy in mice. While myelosuppression can be compensated for, gastrointestinal atrophy is detrimental. Remarkably, deletion of pro-apoptotic Bim/Bcl2l11 prevents gastrointestinal syndrome, while neither loss of Noxa/Pmaip or co-deletion of Bid and Puma/Bbc3 has such a protective effect, identifying BIM as rate-limiting apoptosis effector in mitotic cell death of the gastrointestinal epithelium. In contrast, only overexpression of anti-apoptotic BCL2, but none of the BH3-only protein deficiencies mentioned above, can mitigate myelosuppression. Our findings highlight tissue and cell-type-specific survival dependencies in response to SAC perturbation in vivo.
    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
    后期促进复合物/环小体(APC/C)是一种关键且受严格调节的E3连接酶,可通过控制细胞周期调节剂的降解来协调细胞生命周期。这个复合物的一个有趣的特征是自动抑制机制:一个内在无序的环域,Apc1-300L,阻断Cdc20共激活因子结合,然而Apc1-300L的磷酸化抵消了这种自动抑制。APC/C内的许多此类无序回路仍未被探索。我们对缺乏环的APC/C突变体的系统分析揭示了Apc8的C末端环(Apc8-L)在有丝分裂激活中的关键作用。Apc8-L直接募集CDK衔接蛋白,Xe-p9/Cks2,将Xe-p9-CDK-CycB复合物定位在Apc1-300L附近。这刺激了Apc1-300L的磷酸化和去除,促使形成活性APC/CCdc20。引人注目的是,没有Apc8-L和Apc3-L,APC/C在有丝分裂期间变得不活跃,强调Apc8-L与其他环和激酶的协同作用。这项研究拓宽了我们对APC/C调节中复杂动力学的理解,并提供了对大分子复合物调节的见解。
    The anaphase-promoting complex/cyclosome (APC/C) is a critical and tightly regulated E3 ligase that orchestrates the cellular life cycle by controlling the degradation of cell cycle regulators. An intriguing feature of this complex is an autoinhibition mechanism: an intrinsically disordered loop domain, Apc1-300L, blocks Cdc20 coactivator binding, yet phosphorylation of Apc1-300L counteracts this autoinhibition. Many such disordered loops within APC/C remain unexplored. Our systematic analysis of loop-deficient APC/C mutants uncovered a pivotal role for Apc8\'s C-terminal loop (Apc8-L) in mitotic activation. Apc8-L directly recruits the CDK adaptor protein, Xe-p9/Cks2, positioning the Xe-p9-CDK-CycB complex near Apc1-300L. This stimulates the phosphorylation and removal of Apc1-300L, prompting the formation of active APC/CCdc20. Strikingly, without both Apc8-L and Apc3-L, the APC/C is rendered inactive during mitosis, highlighting Apc8-L\'s synergistic role with other loops and kinases. This study broadens our understanding of the intricate dynamics in APC/C regulation and provides insights on the regulation of macromolecular complexes.
    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
    细胞周期控制依赖于CDK1和磷酸酶如PP1和PP2A-B55的磷酸化的微妙平衡。然而,确定导致细胞周期振荡的主要底物仍然是一个挑战。我们揭示了磷酸调节在后期促进复合物/环小体(APC/C)中的关键作用,特别是通过Apc1-loop300域(Apc1-300L),由CDK1和PP2A-B55协调。PP2A-B55在有丝分裂过程中过早激活,由Greatwall激酶耗竭诱导,导致Apc1-300L去磷酸化,停止APC/C活性和延迟细胞周期蛋白B降解。可以使用B55特异性抑制剂pEnsa或通过去除Apcl-300L来抵消这种作用。我们还显示了Cdc20在细胞周期阶段的动态APC/C相互作用,但是Apc1-300L的去磷酸化特异性地抑制了Cdc20的进一步募集。我们的研究强调了APC/C在细胞周期振荡中的核心作用,将其鉴定为受CDK-PP2A伙伴关系调节的主要底物。
    Cell cycle control relies on a delicate balance of phosphorylation with CDK1 and phosphatases like PP1 and PP2A-B55. Yet, identifying the primary substrate responsible for cell cycle oscillations remains a challenge. We uncover the pivotal role of phospho-regulation in the anaphase-promoting complex/cyclosome (APC/C), particularly through the Apc1-loop300 domain (Apc1-300L), orchestrated by CDK1 and PP2A-B55. Premature activation of PP2A-B55 during mitosis, induced by Greatwall kinase depletion, leads to Apc1-300L dephosphorylation, stalling APC/C activity and delaying Cyclin B degradation. This effect can be counteracted using the B55-specific inhibitor pEnsa or by removing Apc1-300L. We also show Cdc20\'s dynamic APC/C interaction across cell cycle stages, but dephosphorylation of Apc1-300L specifically inhibits further Cdc20 recruitment. Our study underscores APC/C\'s central role in cell cycle oscillation, identifying it as a primary substrate regulated by the CDK-PP2A partnership.
    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
    背景:丝裂原激活的蛋白激酶(MAPK)通过将环境的物理化学波动转化为多种适应性反应来维持细胞稳态。这些反应涉及转录重新连接和细胞周期转换的调节,在其他人中。然而,应激条件如何影响有丝分裂的进展在很大程度上是未知的。有丝分裂检查点是一种监视机制,可以在染色体捕获缺陷的情况下抑制有丝分裂的退出,从而防止非整倍体的产生。在这项研究中,我们研究了MAPKPmk1在应激时有丝分裂退出的调节中的作用。
    结果:我们表明,裂殖酵母细胞缺乏Pmk1,细胞完整性途径(CIP)的MAP激酶效应,对微管损伤过敏,并且在维持中期停滞方面存在缺陷。Epistasis分析表明,Pmk1参与维持主轴装配检查点(SAC)信号,并且它的删除是由于缺少Mad2和Mad3等核心SAC组件。引人注目的是,在不受干扰的生长过程中,pmk1Δ细胞的后期促进复合物(APC/C)激活剂Cdc20Slp1的水平增加了两倍。我们证明了Pmk1通过规范的MAPK对接位点与Cdc20Slp1N端物理相互作用。最重要的是,Cdc20Slp1池通过需要MAPK活性的机制在经历有丝分裂的应激细胞中快速降解,Mad3和蛋白酶体,从而导致延迟的有丝分裂退出。
    结论:我们的数据揭示了MAPK在防止有丝分裂退出和胞质分裂激活方面的新功能。MAPKPmk1对Cdc20Slp1周转的调节提供了一个关键机制,通过该机制可以相对于环境条件调整有丝分裂退出的时间。
    BACKGROUND: Mitogen-activated protein kinases (MAPKs) preserve cell homeostasis by transducing physicochemical fluctuations of the environment into multiple adaptive responses. These responses involve transcriptional rewiring and the regulation of cell cycle transitions, among others. However, how stress conditions impinge mitotic progression is largely unknown. The mitotic checkpoint is a surveillance mechanism that inhibits mitotic exit in situations of defective chromosome capture, thus preventing the generation of aneuploidies. In this study, we investigate the role of MAPK Pmk1 in the regulation of mitotic exit upon stress.
    RESULTS: We show that Schizosaccharomyces pombe cells lacking Pmk1, the MAP kinase effector of the cell integrity pathway (CIP), are hypersensitive to microtubule damage and defective in maintaining a metaphase arrest. Epistasis analysis suggests that Pmk1 is involved in maintaining spindle assembly checkpoint (SAC) signaling, and its deletion is additive to the lack of core SAC components such as Mad2 and Mad3. Strikingly, pmk1Δ cells show up to twofold increased levels of the anaphase-promoting complex (APC/C) activator Cdc20Slp1 during unperturbed growth. We demonstrate that Pmk1 physically interacts with Cdc20Slp1 N-terminus through a canonical MAPK docking site. Most important, the Cdc20Slp1 pool is rapidly degraded in stressed cells undergoing mitosis through a mechanism that requires MAPK activity, Mad3, and the proteasome, thus resulting in a delayed mitotic exit.
    CONCLUSIONS: Our data reveal a novel function of MAPK in preventing mitotic exit and activation of cytokinesis in response to stress. The regulation of Cdc20Slp1 turnover by MAPK Pmk1 provides a key mechanism by which the timing of mitotic exit can be adjusted relative to environmental conditions.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号