Wee1

Wee1
  • 文章类型: Journal Article
    小细胞肺癌(SCLC)患者迫切需要更有效的治疗选择。SCLC细胞中G1检查点的频繁破坏产生对G2/M检查点的依赖性以维持基因组完整性。的确,在临床前模型中,抑制G2/M检查点激酶WEE1显示抑制SCLC生长的前景。然而,毒性和获得性耐药性限制了该策略的临床有效性。这里,在体外和体内使用CRISPR-Cas9基因敲除筛选,我们确定了影响SCLC细胞对WEE1激酶抑制剂AZD1775反应的多种因素,包括GCN2激酶及其信号通路的其他成员.AZD1775处理后GCN2的快速活化触发SCLC细胞中的应激反应。GCN2途径的药理学或遗传学激活增强了AZD1775对癌细胞的杀伤作用。因此,激活GCN2途径代表了增加SCLC中WEE1抑制剂疗效的有希望的策略.
    Patients with small-cell lung cancer (SCLC) are in dire need of more effective therapeutic options. Frequent disruption of the G1 checkpoint in SCLC cells creates a dependency on the G2/M checkpoint to maintain genomic integrity. Indeed, in pre-clinical models, inhibiting the G2/M checkpoint kinase WEE1 shows promise in inhibiting SCLC growth. However, toxicity and acquired resistance limit the clinical effectiveness of this strategy. Here, using CRISPR-Cas9 knockout screens in vitro and in vivo, we identified multiple factors influencing the response of SCLC cells to the WEE1 kinase inhibitor AZD1775, including the GCN2 kinase and other members of its signaling pathway. Rapid activation of GCN2 upon AZD1775 treatment triggers a stress response in SCLC cells. Pharmacological or genetic activation of the GCN2 pathway enhances cancer cell killing by AZD1775. Thus, activation of the GCN2 pathway represents a promising strategy to increase the efficacy of WEE1 inhibitors in SCLC.
    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
    缺乏功能性p53的癌细胞预后不良,需要有效的治疗策略。抑制WEE1,G2/M细胞周期检查点看门人,代表了治疗p53缺陷型NSCLC的有希望的方法。这里,我们阐明p53和WEE1之间的联系,并探索治疗p53缺陷型NSCLC的协同治疗方法.我们的研究表明,p53缺乏通过抑制其SUMO化过程上调WEE1的蛋白质水平和激酶活性,从而增强p53缺陷型NSCLC对WEE1抑制剂的敏感性。此外,我们证明了WEE1抑制剂Adavosertib诱导细胞内脂质过氧化,特别是在缺乏p53的NSCLC细胞中,提示与促氧化剂药物的潜在协同作用。重修双硫仑(DSF),与铜(Cu)联合使用的酒精中毒治疗药物,表现出抗NSCLC的促氧化特性。DSF-Cu处理的p53缺陷型NSCLC细胞显示WEE1蛋白水平的时间依赖性增加。随后对涉及Adavosertib和DSF-Cu的联合疗法的评估显示,在p53缺陷细胞以及异种移植模型中观察到的细胞活力降低,肿瘤体积较小,肿瘤重量较轻,同时与溶质载体家族7成员11(SLC7A11)/谷胱甘肽调节的铁凋亡途径激活相关。总之,我们的研究结果阐明了p53和WEE1之间的分子相互作用,并揭示了治疗p53缺陷型NSCLC的新型协同治疗策略.
    Cancer cells lacking functional p53 exhibit poor prognosis, necessitating effective treatment strategies. Inhibiting WEE1, the G2/M cell cycle checkpoint gatekeeper, represents a promising approach for treating p53-deficient NSCLC. Here, we elucidate the connection between p53 and WEE1 and explore a synergistic therapeutic approach for managing p53-deficient NSCLC. Our study reveals that p53 deficiency upregulates both protein levels and kinase activity of WEE1 by inhibiting its SUMOylation process, thereby enhancing the susceptibility of p53-deficient NSCLC to WEE1 inhibitors. Furthermore, we demonstrate that the WEE1 inhibitor Adavosertib induces intracellular lipid peroxidation, specifically in p53-deficient NSCLC cells, suggesting potential synergy with pro-oxidant drugs. Repurposing Disulfiram (DSF), an alcoholism treatment drug used in combination with copper (Cu), exhibits pro-oxidant properties against NSCLC. DSF-Cu-treated p53-deficient NSCLC cells show a time-dependent increase in WEE1 protein levels. Subsequent evaluation of the combination therapy involving Adavosertib and DSF-Cu reveals reduced cell viability along with smaller tumor volumes and lighter tumor weights observed both in p53-deficient cells as well as in xenograft models while correlating with solute carrier family 7-member 11 (SLC7A11)/glutathione-regulated ferroptosis pathway activation. In conclusion, our findings elucidate the molecular interplay between p53 and WEE1 and unveil a novel synergistic therapeutic strategy for treating p53-deficient NSCLC.
    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
    具有内部串联重复的过度活跃的FMS样受体酪氨酸激酶3突变体(FLT3-ITD)是侵袭性急性髓细胞性白血病(AML)的频繁驱动突变。FLT3的抑制剂在合理设计的共处理方案中产生了有希望的结果。由于FLT3-ITD调节DNA复制和DNA修复,有效的抗白血病策略可能依赖于FLT3-ITD的联合抑制以及细胞周期进程和DNA完整性的调节因子.这些包括控制细胞周期进程的WEE1激酶,核苷酸合成,和DNA复制起点激发。我们研究了FLT3和WEE1的药理学抑制如何影响AML细胞系和原代AML细胞的存活和基因组完整性。我们发现,FLT3和WEE1的有希望的临床分级和临床前抑制剂可协同触发表达FLT3-ITD的白血病细胞的凋亡。单链和双链DNA损伤的积累先于此过程。基于质谱的蛋白质组分析显示FLT3-ITD和WEE1维持核糖核苷酸还原酶亚基RRM2的表达,其为DNA复制提供dNTP。与FLT3-ITD对白血病细胞的强烈促凋亡作用不同,FLT3和WEE1的抑制剂不会损害健康的人血细胞和鼠造血干细胞。因此,FLT3-ITD和WEE1的药理学抑制可能会得到改善,合理设计的治疗选择。
    Hyperactive FMS-like receptor tyrosine kinase-3 mutants with internal tandem duplications (FLT3-ITD) are frequent driver mutations of aggressive acute myeloid leukemia (AML). Inhibitors of FLT3 produce promising results in rationally designed cotreatment schemes. Since FLT3-ITD modulates DNA replication and DNA repair, valid anti-leukemia strategies could rely on a combined inhibition of FLT3-ITD and regulators of cell cycle progression and DNA integrity. These include the WEE1 kinase which controls cell cycle progression, nucleotide synthesis, and DNA replication origin firing. We investigated how pharmacological inhibition of FLT3 and WEE1 affected the survival and genomic integrity of AML cell lines and primary AML cells. We reveal that promising clinical grade and preclinical inhibitors of FLT3 and WEE1 synergistically trigger apoptosis in leukemic cells that express FLT3-ITD. An accumulation of single and double strand DNA damage precedes this process. Mass spectrometry-based proteomic analyses show that FLT3-ITD and WEE1 sustain the expression of the ribonucleotide reductase subunit RRM2, which provides dNTPs for DNA replication. Unlike their strong pro-apoptotic effects on leukemia cells with FLT3-ITD, inhibitors of FLT3 and WEE1 do not damage healthy human blood cells and murine hematopoietic stem cells. Thus, pharmacological inhibition of FLT3-ITD and WEE1 might become an improved, rationally designed therapeutic option.
    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
    肾小球足细胞丢失是慢性肾脏病发病机制中的基本事件。目前,有丝分裂突变(MC)已成为足细胞损失的主要原因。然而,足细胞中MC的调节尚未阐明。本研究旨在研究p53在阿霉素(ADR)诱导的肾病足细胞MC中的作用及其机制。体外足细胞刺激与ADR引发MC的发生,伴有p53和细胞周期蛋白依赖性激酶(CDK1)/细胞周期蛋白B1的过度激活。p53的抑制逆转了足细胞中ADR诱发的MC,并防止了足细胞的损伤和损失。进一步研究表明,p53通过调节Wee1的表达介导CDK1/cyclinB1的激活。抑制Wee1消除了p53抑制对CDK1/cyclinB1的调节作用,并通过p53抑制在ADR刺激的足细胞中重新启动MC。在ADR肾病的小鼠模型中,抑制p53可改善蛋白尿和足细胞损伤。此外,p53的抑制通过调节Wee1/CDK1/cyclinB1轴阻断ADR肾病小鼠足细胞中MC的进展。我们的发现证实,p53通过调节Wee1/CDK1/CyclinB1轴促进足细胞中的MC,这可能代表了慢性肾脏疾病进展过程中足细胞损伤和丢失的新机制。
    Podocyte loss in glomeruli is a fundamental event in the pathogenesis of chronic kidney diseases. Currently, mitotic catastrophe (MC) has emerged as the main cause of podocyte loss. However, the regulation of MC in podocytes has yet to be elucidated. The current work aimed to study the role and mechanism of p53 in regulating the MC of podocytes using adriamycin (ADR)-induced nephropathy. In vitro podocyte stimulation with ADR triggered the occurrence of MC, which was accompanied by hyperactivation of p53 and cyclin-dependent kinase (CDK1)/cyclin B1. The inhibition of p53 reversed ADR-evoked MC in podocytes and protected against podocyte injury and loss. Further investigation showed that p53 mediated the activation of CDK1/cyclin B1 by regulating the expression of Wee1. Restraining Wee1 abolished the regulatory effect of p53 inhibition on CDK1/cyclin B1 and rebooted MC in ADR-stimulated podocytes via p53 inhibition. In a mouse model of ADR nephropathy, the inhibition of p53 ameliorated proteinuria and podocyte injury. Moreover, the inhibition of p53 blocked the progression of MC in podocytes in ADR nephropathy mice through the regulation of the Wee1/CDK1/cyclin B1 axis. Our findings confirm that p53 contributes to MC in podocytes through regulation of the Wee1/CDK1/Cyclin B1 axis, which may represent a novel mechanism underlying podocyte injury and loss during the progression of chronic kidney disorder.
    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:利用癌细胞脆弱性的靶向治疗有望改善患者预后并减少化疗的副作用。然而,精准治疗的疗效有限,部分原因是肿瘤细胞异质性。更好地理解药物作用如何与癌细胞状态多样性相关联对于确定可以预防疾病复发的有效组合疗法至关重要。
    结果:这里,我们描述了G2/M检查点抑制在急性淋巴细胞白血病(ALL)中的作用,并证明了WEE1靶向治疗对细胞命运决定调节回路的影响.我们发现KMT2A重排对ALL细胞增殖恢复的抑制作用最高。RS4的单细胞RNA-seq和ATAC-seq;11个带有KMT2A::AFF1的细胞,用WEE1抑制剂AZD1775处理,揭示细胞状态的多样化,部分细胞表现出与细胞凋亡和衰老相关的p53驱动过程的强烈激活,以及核心KMT2A-RUNX1-MYC监管网络的中断。在这种由WEE1抑制诱导的细胞状态多样化中,亚群过渡到药物耐受性细胞状态,其特征是转录因子的激活调节前B细胞命运,脂质代谢,和以可逆方式的前BCR信号传导。BCR信号抑制剂达沙替尼的序贯治疗,伊布替尼,或通过fatostatin或AZD2014干扰代谢通过诱导细胞死亡和抑制干性标志物有效抵消药物耐受性。
    结论:总的来说,我们的研究结果为与细胞周期和细胞命运调控相关的基因调控程序的紧密连接提供了新的见解,和顺序给药WEE1抑制剂与前BCR信号或代谢的低毒性抑制剂的基本原理。
    Targeted therapies exploiting vulnerabilities of cancer cells hold promise for improving patient outcome and reducing side-effects of chemotherapy. However, efficacy of precision therapies is limited in part because of tumor cell heterogeneity. A better mechanistic understanding of how drug effect is linked to cancer cell state diversity is crucial for identifying effective combination therapies that can prevent disease recurrence.
    Here, we characterize the effect of G2/M checkpoint inhibition in acute lymphoblastic leukemia (ALL) and demonstrate that WEE1 targeted therapy impinges on cell fate decision regulatory circuits. We find the highest inhibition of recovery of proliferation in ALL cells with KMT2A-rearrangements. Single-cell RNA-seq and ATAC-seq of RS4;11 cells harboring KMT2A::AFF1, treated with the WEE1 inhibitor AZD1775, reveal diversification of cell states, with a fraction of cells exhibiting strong activation of p53-driven processes linked to apoptosis and senescence, and disruption of a core KMT2A-RUNX1-MYC regulatory network. In this cell state diversification induced by WEE1 inhibition, a subpopulation transitions to a drug tolerant cell state characterized by activation of transcription factors regulating pre-B cell fate, lipid metabolism, and pre-BCR signaling in a reversible manner. Sequential treatment with BCR-signaling inhibitors dasatinib, ibrutinib, or perturbing metabolism by fatostatin or AZD2014 effectively counteracts drug tolerance by inducing cell death and repressing stemness markers.
    Collectively, our findings provide new insights into the tight connectivity of gene regulatory programs associated with cell cycle and cell fate regulation, and a rationale for sequential administration of WEE1 inhibitors with low toxicity inhibitors of pre-BCR signaling or metabolism.
    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
    Kirsten大鼠肉瘤病毒(KRAS)-G12C抑制剂治疗KRAS突变肺癌的临床发展受到共突变的限制,固有电阻,以及后天抵抗力的出现。因此,在KRAS突变的非小细胞肺癌(NSCLC)中,我们迫切需要提高细胞凋亡的创新策略.通过使用746个crRNA文库的CRISPR-Cas9敲除筛选和使用432个化合物的定制文库的药物筛选,我们发现WEE1激酶抑制剂是有效的细胞凋亡促进剂,特别是在携带TP53突变的KRAS突变型NSCLC细胞中。机械上,WEE1抑制促进G2/M转换并减少DNA损伤反应(DDR)途径中的检查点激酶2(CHK2)和Rad51表达,这与细胞凋亡和DNA双链断裂的修复有关,导致有丝分裂灾难。值得注意的是,KRAS-G12C和WEE1的联合抑制持续抑制肿瘤生长。我们的结果表明靶向WEE1是一种有希望的治疗具有TP53突变的KRAS突变的NSCLC的策略。
    The clinical development of Kirsten rat sarcoma virus (KRAS)-G12C inhibitors for the treatment of KRAS-mutant lung cancer is limited by the presence of co-mutations, intrinsic resistance, and the emergence of acquired resistance. Therefore, innovative strategies for enhancing apoptosis in KRAS-mutated non-small cell lung cancer (NSCLC) are urgently needed. Through CRISPR-Cas9 knockout screening using a library of 746 crRNAs and drug screening with a custom library of 432 compounds, we discover that WEE1 kinase inhibitors are potent enhancers of apoptosis, particularly in KRAS-mutant NSCLC cells harboring TP53 mutations. Mechanistically, WEE1 inhibition promotes G2/M transition and reduces checkpoint kinase 2 (CHK2) and Rad51 expression in the DNA damage response (DDR) pathway, which is associated with apoptosis and the repair of DNA double-strand breaks, leading to mitotic catastrophe. Notably, the combined inhibition of KRAS-G12C and WEE1 consistently suppresses tumor growth. Our results suggest targeting WEE1 as a promising therapeutic strategy for KRAS-mutated NSCLC with TP53 mutations.
    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:GL-V9对各种类型的肿瘤具有抗肿瘤作用。本研究旨在验证GL-V9与奥沙利铂在抑制结直肠癌(CRC)中的协同作用,并探讨协同作用机制。
    方法:用MTT法检测协同作用,用彗星法检测机制,蛋白质印迹和免疫组织化学(IHC)。构建异种移植模型以证实其体内协同作用及其机制。
    结果:GL-V9可增强奥沙利铂的DNA损伤作用,从而在体外和体内协同抑制结肠癌细胞。在HCT-116细胞中,GL-V9加速了Wee1的降解,并导致细胞周期停滞和错误进入有丝分裂的废除,绕过奥沙利铂引起的DNA损伤反应。我们的发现表明,GL-V9与HSP90的结合是Wee1降解和结肠癌细胞对奥沙利铂的脆弱性的原因。功能上,HSP90或WEE1的过表达取消了GL-V9和奥沙利铂的协同作用。
    结论:总的来说,我们的研究结果表明,GL-V9与奥沙利铂协同作用抑制CRC,并显示出提高奥沙利铂疗效的有希望的策略.
    OBJECTIVE: GL-V9 exhibited anti-tumour effects on various types of tumours. This study aimed to verify if GL-V9 synergized with oxaliplatin in suppressing colorectal cancer (CRC) and to explore the synergistic mechanism.
    METHODS: The synergy effect was tested by MTT assays and the mechanism was examined by comet assay, western blotting and immunohistochemistry (IHC). Xenograft model was constructed to substantiated the synergy effect and its mechanism in vivo.
    RESULTS: GL-V9 was verified to enhance the DNA damage effect of oxaliplatin, so as to synergistically suppress colon cancer cells in vitro and in vivo. In HCT-116 cells, GL-V9 accelerated the degradation of Wee1 and induced the abrogation of cell cycle arrest and mis-entry into mitosis, bypassing the DNA damage response caused by oxaliplatin. Our findings suggested that GL-V9 binding to HSP90 was responsible for the degradation of Wee1 and the vulnerability of colon cancer cells to oxaliplatin. Functionally, overexpression of either HSP90 or WEE1 annulled the synergistic effect of GL-V9 and oxaliplatin.
    CONCLUSIONS: Collectively, our findings revealed that GL-V9 synergized with oxaliplatin to suppress CRC and displayed a promising strategy to improve the efficacy of oxaliplatin.
    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
    口腔鳞状细胞癌(OSCC)是一种常见且高致死性的上皮癌。本研究旨在证实METTL3在促进OSCC中的作用,并探讨其具体的潜在机制。METTL3,含YTH结构域的家族2(YTHDF2)的表达,在正常口腔上皮细胞和OSCC细胞中检测WEE1。在OSCC细胞中过表达WEE1后检查细胞功能。MeRIP-qPCR分析用于检测HOK中的WEE1m6A水平,SCC25和CAL27细胞。通过敲低METTL3/YTHDF2在OSCC细胞中评估WEE1及其m6A水平,评估METTL3/YTHDF2和WEE1之间的相互作用。还研究了METTL3和YTHDF2下调对WEE1mRNA稳定性的影响。测量过表达WEE1和YTHDF2后的OSCC裸鼠模型中的肿瘤重量和体积。免疫组化法检测OSCC组织中Ki-67和WEE1的表达。与正常口腔上皮细胞相比,METTL3和YTHDF2在OSCC细胞中上调,当WEE1被下调时,METTL3/YTHDF2表达与WEE1呈负相关。WEE1过表达抑制增殖,入侵,和迁移,同时促进OSCC细胞凋亡。METTL3和YTHDF2与WEE1mRNA结合。METTL3/YTHDF2敲低可增加WEE1水平和WEE1mRNA稳定性。METTL3抑制降低了WEE1m6A水平。抑制METTL3削弱了YTHDF2和WEE1mRNA之间的相互作用。在体内,WEE1的过表达抑制了OSCC的发展,通过YTHDF2的过表达而逆转。METTL3通过m6A-YTHDF2依赖性下调WEE1促进OSCC的进展。
    Oral squamous cell carcinoma (OSCC) is a common and highly lethal epithelial cancer. This study aimed to confirm the role of METTL3 in promoting OSCC and investigate its specific underlying mechanisms. Expression of the METTL3, YTH domain-containing family 2 (YTHDF2), and WEE1 were examined in normal oral epithelial cells and OSCC cells. Cell functions were examined after overexpressing WEE1 in OSCC cells. MeRIP-qPCR analysis was used to detect WEE1 m6A levels in HOK, SCC25, and CAL27 cells. WEE1 and its m6A levels were evaluated in OSCC cells by knocking down METTL3/YTHDF2, assessing the interaction between METTL3/YTHDF2 and WEE1. The impact of METTL3 and YTHDF2 downregulation on WEE1 mRNA stability was also investigated. The tumor weight and volume in a nude mouse model of OSCC after overexpression of WEE1 and YTHDF2 were measured. Expression of Ki-67 and WEE1 in OSCC tissue was detected using immunohistochemistry. Compared to normal oral epithelial cells, METTL3 and YTHDF2 were upregulated in OSCC cells, while WEE1 was downregulated, and there was a negative correlation between WEE1 and METTL3/YTHDF2 expression. WEE1 overexpression inhibited proliferation, invasion, and migration while promoting apoptosis in OSCC cells. METTL3 and YTHDF2 bound to WEE1 mRNA. METTL3/YTHDF2 knockdown increased WEE1 levels and WEE1 mRNA stability. METTL3 inhibition reduced WEE1 m6A levels. Inhibition of METTL3 weakened the interaction between YTHDF2 and WEE1 mRNA. In vivo, overexpression of WEE1 suppressed OSCC development, which was reversed by overexpression of YTHDF2. METTL3 facilitates the progression of OSCC through m6A-YTHDF2-dependent downregulation of WEE1.
    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
    尽管最近在诊断和治疗方面取得了进展,胰腺导管腺癌(PDAC),胰腺癌最常见的类型,仍然是最致命的癌症,五年生存率很低。迫切需要开发新的疗法来解决这个问题。在这项研究中,我们通过修饰肿瘤抑制miRNA开发了一种治疗策略,miR-15a和miR-194,与化疗吉西他滨(Gem)产生宝石修饰的模拟物,Gem-miR-15a和Gem-miR-194。在一组PDAC细胞系中,我们发现Gem-miR-15a和Gem-miR-194诱导细胞周期停滞和凋亡,并且这些模拟物是有效的抑制剂,其IC50值比它们的天然对应物或单独的Gem低几百倍。此外,我们发现Gem-miR-15a和Gem-miR-194通过下调Gem-miR-15a的几个关键靶标(包括WEE1,CHK1,BMI1和YAP1)的表达来保留miRNA功能,和FOXA1代表Gem-miR-194。我们还发现,与Gem相比,我们的Gem修饰的miRNA模拟物在患者来源的PDAC类器官中表现出增强的功效。此外,我们观察到Gem-miR-15a在体内显著抑制PDAC肿瘤生长,而未观察到任何明显的毒性迹象.总的来说,我们的结果证明了宝石修饰的miRNA作为PDAC治疗策略的治疗潜力.
    Despite the recent advancement in diagnosis and therapy, pancreatic ductal adenocarcinoma (PDAC), the most common type of pancreatic cancer, is still the most lethal cancer with a low five-year survival rate. There is an urgent need to develop new therapies to address this issue. In this study, we developed a treatment strategy by modifying tumor suppressor miRNAs, miR-15a and miR-194, with the chemotherapeutic gemcitabine (Gem) to create Gem-modified mimics, Gem-miR-15a and Gem-miR-194, respectively. In a panel of PDAC cell lines, we found that Gem-miR-15a and Gem-miR-194 induce cell-cycle arrest and apoptosis, and these mimics are potent inhibitors with IC50 values up to several hundred fold less than their native counterparts or Gem alone. Furthermore, we found that Gem-miR-15a and Gem-miR-194 retained miRNA function by downregulating the expression of several key targets including WEE1, CHK1, BMI1, and YAP1 for Gem-miR-15a, and FOXA1 for Gem-miR-194. We also found that our Gem-modified miRNA mimics exhibit an enhanced efficacy compared to Gem in patient-derived PDAC organoids. Furthermore, we observed that Gem-miR-15a significantly inhibits PDAC tumor growth in vivo without observing any noticeable signs of toxicity. Overall, our results demonstrate the therapeutic potential of Gem-modified miRNAs as a treatment strategy for PDAC.
    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
    复制应激(RS)是癌细胞的特征状态,因为它们倾向于将复制的精确性交换为快速增殖和增加的基因组不稳定性。为了克服复制控制不当的后果,恶性细胞经常使其DNA损伤反应(DDR)途径(ATM-CHK2-p53途径)的部分失活,同时依赖于其他有助于保持复制叉稳定性的途径(ATR-CHK1)。这产生了对剩余DDR路径的依赖,DDR抑制剂的复制和合成致死性进一步不稳定,常见的致癌改变,如TP53,RB1,ATM的突变,MYC的扩增,CCNE1等。对RS的反应通常受到细胞周期协调的限制,转录和复制。抑制WEE1和PKMYT1激酶,防止计划外的有丝分裂进入,导致复制不足的网站的脆弱性。最近的证据还表明,抑制细胞周期蛋白依赖性激酶(CDKs),例如CDK4/6、CDK2、CDK8/19和CDK12/13可以通过破坏DNA修复和复制控制来促成RS。这里,我们回顾了RS在癌症中的主要原因以及主要的治疗靶点-ATR,CHK1、PARP及其抑制剂。
    Replication stress (RS) is a characteristic state of cancer cells as they tend to exchange precision of replication for fast proliferation and increased genomic instability. To overcome the consequences of improper replication control, malignant cells frequently inactivate parts of their DNA damage response (DDR) pathways (the ATM-CHK2-p53 pathway), while relying on other pathways which help to maintain replication fork stability (ATR-CHK1). This creates a dependency on the remaining DDR pathways, vulnerability to further destabilization of replication and synthetic lethality of DDR inhibitors with common oncogenic alterations such as mutations of TP53, RB1, ATM, amplifications of MYC, CCNE1 and others. The response to RS is normally limited by coordination of cell cycle, transcription and replication. Inhibition of WEE1 and PKMYT1 kinases, which prevent unscheduled mitosis entry, leads to fragility of under-replicated sites. Recent evidence also shows that inhibition of Cyclin-dependent kinases (CDKs), such as CDK4/6, CDK2, CDK8/19 and CDK12/13 can contribute to RS through disruption of DNA repair and replication control. Here, we review the main causes of RS in cancers as well as main therapeutic targets-ATR, CHK1, PARP and their inhibitors.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号