Mitotic catastrophe

有丝分裂突变
  • 文章类型: Journal Article
    肺类癌(LCs)是一种起源于支气管肺系统的神经内分泌肿瘤(NET)。LC占所有NET的20-25%,约占肺癌的1-2%。鉴于NETs的高度血管化性质及其过度表达血管生长因子受体(VEGFR)的趋势,抑制血管生成似乎是减缓肿瘤生长和扩散的潜在治疗靶点。本研究评估了阿西替尼(AXI)的长期抗肿瘤活性和相关机制。一种VEGFR靶向药物,在LC细胞系中。
    将三种LC细胞系(NCI-H727、UMC-11和NCI-H835)与它们各自的EC50AXI浓度一起孵育6天。在孵化结束时,进行FACS实验和Western印迹分析以检查细胞周期的变化和凋亡的激活。添加了显微镜分析以描述存在时衰老和有丝分裂突变的机制。
    AXI对LC细胞系的主要作用是通过间接DNA损伤阻止肿瘤生长。值得注意的是,AXI在细胞系中以不同的方式触发这种反应,如诱导衰老或有丝分裂灾难。当DNA损伤减轻时,该药物似乎失去了功效,如在NCI-H835细胞中观察到的。
    AXI在LC肿瘤细胞中影响细胞活力和增殖的能力凸显了其作为治疗剂的潜力。DNA损伤的作用以及随之而来的衰老激活似乎是AXI发挥其功能的先决条件。
    UNASSIGNED: Lung carcinoids (LCs) are a type of neuroendocrine tumor (NET) that originate in the bronchopulmonary tract. LCs account for 20-25% of all NETs and approximately 1-2% of lung cancers. Given the highly vascularized nature of NETs and their tendency to overexpress vascular growth factor receptors (VEGFR), inhibiting angiogenesis appears as a potential therapeutic target in slowing down tumor growth and spread. This study evaluated the long-term antitumor activity and related mechanisms of axitinib (AXI), a VEGFR-targeting drug, in LC cell lines.
    UNASSIGNED: Three LC cell lines (NCI-H727, UMC-11 and NCI-H835) were incubated with their respective EC50 AXI concentrations for 6 days. At the end of the incubation, FACS experiments and Western blot analyses were performed to examine changes in the cell cycle and the activation of apoptosis. Microscopy analyses were added to describe the mechanisms of senescence and mitotic catastrophe when present.
    UNASSIGNED: The primary effect of AXI on LC cell lines is to arrest tumor growth through an indirect DNA damage. Notably, AXI triggers this response in diverse manners among the cell lines, such as inducing senescence or mitotic catastrophe. The drug seems to lose its efficacy when the DNA damage is mitigated, as observed in NCI-H835 cells.
    UNASSIGNED: The ability of AXI to affect cell viability and proliferation in LC tumor cells highlights its potential as a therapeutic agent. The role of DNA damage and the consequent activation of senescence seem to be a prerequisite for AXI to exert its function.
    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
    有丝分裂突变(MC)途径在肝细胞癌(HCC)进展和肿瘤微环境(TME)调节中起重要作用。然而,MC异质性与免疫逃避和治疗反应的相关机制尚不清楚.
    基于先前发表的94个高度相关的MC基因,研究了来自癌症基因组图谱(TCGA)的HCC患者数据以及免疫特征和预后分层的变化。通过单细胞RNA测序和空间转录组(ST)分析评估MCG的时间和空间特异性差异。多个外部数据库(GEO,ICGC)用于构建MC相关风险评分模型。
    从TCGA中鉴定HCC患者的两种MC相关亚型,在免疫特征和预后风险分层方面存在明显差异。空间定位进一步将低MC肿瘤区域与显著的免疫逃逸相关信号联系起来。结合MC风险评分和传统指标的列线图对于早期预测HCC患者预后具有巨大的效果。
    MC异质性使HCC的免疫逃逸和治疗抵抗成为可能。MC基因标签是肝癌的可靠预后指标。通过揭示HCC的明确免疫和空间异质性,我们的综合方法为临床决策提供了情境治疗策略.
    UNASSIGNED: The mitotic catastrophe (MC) pathway plays an important role in hepatocellular carcinoma (HCC) progression and tumor microenvironment (TME) regulation. However, the mechanisms linking MC heterogeneity to immune evasion and treatment response remain unclear.
    UNASSIGNED: Based on 94 previously published highly correlated genes for MC, HCC patients\' data from the Cancer Genome Atlas (TCGA) and changes in immune signatures and prognostic stratification were studied. Time and spatial-specific differences for MCGs were assessed by single-cell RNA sequencing and spatial transcriptome (ST) analysis. Multiple external databases (GEO, ICGC) were employed to construct an MC-related riskscore model.
    UNASSIGNED: Identification of two MC-related subtypes in HCC patients from TCGA, with clear differences in immune signatures and prognostic risk stratification. Spatial mapping further associates low MC tumor regions with significant immune escape-related signaling. Nomogram combining MC riskscore and traditional indicators was validated great effect for early prediction of HCC patient outcomes.
    UNASSIGNED: MC heterogeneity enables immune escape and therapy resistance in HCC. The MC gene signature serves as a reliable prognostic indicator for liver cancer. By revealing clear immune and spatial heterogeneity of HCC, our integrated approach provides contextual therapeutic strategies for optimal clinical decision-making.
    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
    肾小球足细胞,一个终末分化的细胞,对肾小球滤过屏障的完整性至关重要。足细胞重新进入有丝分裂期导致损伤或死亡,称为有丝分裂灾难(MC),显着促进糖尿病肾病(DN)的进展。此外,P62介导的自噬通量已显示出调节DN诱导的足细胞损伤。虽然以前的研究,包括我们的,已经证明熊果酸(UA)通过在高糖条件下增强自噬来减轻足细胞损伤,UA对DN的保护功能和潜在的调节机制尚未完全阐明.为探讨足细胞损伤在DN进展中的调控机制,和UA治疗对DN进展的保护功能,我们利用db/db小鼠和高糖(HG)诱导的足细胞模型在体内和体外,有或没有UA管理。我们的发现表明,UA治疗通过改善生化指标来减少DN进展。P62的积累导致小鼠双分钟基因2(MDM2)在DN期间在足细胞中调节MC,UA通过增强P62介导的自噬来改善。此外,NF-κBp65或TNF-α的过表达在体内和体外均消除了UA的保护作用。总的来说,我们的结果提供了强有力的证据,表明UA可能是DN的潜在治疗剂,通过靶向自噬P62积累通过NF-κB/MDM2/Notch1途径抑制足细胞MC进行调节。
    The glomerular podocyte, a terminally differentiated cell, is crucial for the integrity of the glomerular filtration barrier. The re-entry of podocytes into the mitotic phase results in injuries or death, known as mitotic catastrophe (MC), which significantly contributes to the progression of diabetic nephropathy (DN). Furthermore, P62-mediated autophagic flux has been shown to regulate DN-induced podocyte injury. Although previous studies, including ours, have demonstrated that ursolic acid (UA) mitigates podocyte injury by enhancing autophagy under high glucose conditions, the protective functions and potential regulatory mechanisms of UA against DN have not been fully elucidated. For aiming to investigate the regulatory mechanism of podocyte injuries in DN progression, and the protective function of UA treatment against DN progression, we utilized db/db mice and high glucose (HG)-induced podocyte models in vivo and in vitro, with or without UA administration. Our findings indicate that UA treatment reduced DN progression by improving biochemical indices. P62 accumulation led to Murine Double Minute gene 2 (MDM2)-regulated MC in podocytes during DN, which was ameliorated by UA through enhanced P62-mediated autophagy. Additionally, the overexpression of NF-κB p65 or TNF-α abolished the protective effects of UA both in vivo and in vitro. Overall, our results provide strong evidence that UA could be a potential therapeutic agent for DN, regulated by inhibiting podocyte MC through the NF-κB/MDM2/Notch1 pathway by targeting autophagic-P62 accumulation.
    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
    人类INF2基因突变导致常染色体显性遗传的局灶性节段肾小球硬化(FSGS)-一种以足细胞丢失为特征的疾病,疤痕,以及随后的肾脏变性。要了解INF2相关的致病性,我们检查了致病性INF2对肾上皮细胞系和人原代足细胞的影响。我们的研究表明,有丝分裂细胞的发生率增加,具有多余的微管组织中心促进多极纺锤体组装,导致核异常,特别是多微核。外源性致病性INF2的表达水平与内源性INF2相似。无论使用的表达方法(逆转录病毒感染或质粒转染)或使用的启动子(LTR或CMV),都观察到异常的核表型。并且在外源野生型INF2表达时不存在。这表明致病性INF2的作用不是由于过表达或实验细胞操作,而是针对致病性INF2的内在特性。INF2催化结构域的失活防止了异常核形成。致病性INF2触发了转录辅因子MRTF易位到细胞核中。RNA测序揭示了转录组的深刻变化,这可能主要归因于MRTF-SRF转录复合物的持续激活。细胞最终经历有丝分裂灾难和死亡。减少MRTF-SRF激活减轻多微核,降低细胞死亡的程度。我们的结果,如果在动物模型中验证,可以提供对驱动INF2相关FSGS肾小球变性的机制的见解,并可能提出阻碍FSGS进展的潜在治疗策略。
    Mutations in the human INF2 gene cause autosomal dominant focal segmental glomerulosclerosis (FSGS)-a condition characterized by podocyte loss, scarring, and subsequent kidney degeneration. To understand INF2-linked pathogenicity, we examined the effect of pathogenic INF2 on renal epithelial cell lines and human primary podocytes. Our study revealed an increased incidence of mitotic cells with surplus microtubule-organizing centers fostering multipolar spindle assembly, leading to nuclear abnormalities, particularly multi-micronucleation. The levels of expression of exogenous pathogenic INF2 were similar to those of endogenous INF2. The aberrant nuclear phenotypes were observed regardless of the expression method used (retrovirus infection or plasmid transfection) or the promoter (LTR or CMV) used, and were absent with exogenous wild type INF2 expression. This indicates that the effect of pathogenic INF2 is not due to overexpression or experimental cell manipulation, but instead to the intrinsic properties of pathogenic INF2. Inactivation of the INF2 catalytic domain prevented aberrant nuclei formation. Pathogenic INF2 triggered the translocation of the transcriptional cofactor MRTF into the nucleus. RNA sequencing revealed a profound alteration in the transcriptome that could be primarily attributed to the sustained activation of the MRTF-SRF transcriptional complex. Cells eventually underwent mitotic catastrophe and death. Reducing MRTF-SRF activation mitigated multi-micronucleation, reducing the extent of cell death. Our results, if validated in animal models, could provide insights into the mechanism driving glomerular degeneration in INF2-linked FSGS and may suggest potential therapeutic strategies for impeding FSGS progression.
    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
    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:历史上认为与其他癌症适应症相比,肾细胞癌(RCC)对放射疗法(RT)的反应较低。然而,通过单部分和多部分立体定向消融放射治疗(SABR)的精确高剂量辐射输送的进步导致了更好的结果和减少治疗相关的毒性,引发了人们对使用RT治疗RCC的新兴趣。此外,许多研究表明,包括化疗在内的某些治疗剂可以增加肿瘤对RT的敏感性,导致对结合这些治疗的兴趣越来越大。这里,我们在肿瘤靶向脂质体制剂中开发了两种放射增敏剂的合理组合,用于增强RCC的RT.本研究的目的是评估结合mTOR抑制剂依维莫司(E)和生存素抑制剂YM155(Y)的肿瘤靶向脂质体制剂在增强RCC肿瘤对放射的敏感性中的功效。
    方法:我们稍微修改了我们先前公开的肿瘤靶向脂质体制剂,以开发E和Y在单一脂质体制剂(EY-L)中的合理组合,并评估其在体外RCC细胞系和体内RCC肿瘤中的功效。我们进一步研究了EY-L对RCC细胞系和肿瘤的放射敏感性,并探索了放射增敏的潜在机制。
    结果:在具有免疫活性的同基因RCC小鼠模型中,EY-L在包含原发性肿瘤生长和改善存活率方面优于相应的单一载药制剂E-L和Y-L。与E-L和Y-L相比,EY-L还表现出明显更高的体外RCC细胞对辐射的敏感性。此外,在异种移植物和鼠RCC模型中,EY-L对放射治疗敏感的RCC肿瘤。EY-L通过下调多个细胞周期检查点和DNA损伤修复途径介导的有丝分裂突变的诱导可能是增强放射治疗的原因。
    结论:综合来看,我们的研究证明了战略性联合治疗通过抑制DNA损伤修复和显著增加有丝分裂突变,使RCC对放射治疗敏感的疗效.这种联合疗法可能会在RCC患者治疗期间用于增强放射疗法。
    BACKGROUND: Renal cell carcinoma (RCC) was historically considered to be less responsive to radiation therapy (RT) compared to other cancer indications. However, advancements in precision high-dose radiation delivery through single-fraction and multi-fraction stereotactic ablative radiotherapy (SABR) have led to better outcomes and reduced treatment-related toxicities, sparking renewed interest in using RT to treat RCC. Moreover, numerous studies have revealed that certain therapeutic agents including chemotherapies can increase the sensitivity of tumors to RT, leading to a growing interest in combining these treatments. Here, we developed a rational combination of two radiosensitizers in a tumor-targeted liposomal formulation for augmenting RT in RCC. The objective of this study is to assess the efficacy of a tumor-targeted liposomal formulation combining the mTOR inhibitor everolimus (E) with the survivin inhibitor YM155 (Y) in enhancing the sensitivity of RCC tumors to radiation.
    METHODS: We slightly modified our previously published tumor-targeted liposomal formulation to develop a rational combination of E and Y in a single liposomal formulation (EY-L) and assessed its efficacy in RCC cell lines in vitro and in RCC tumors in vivo. We further investigated how well EY-L sensitizes RCC cell lines and tumors toward radiation and explored the underlying mechanism of radiosensitization.
    RESULTS: EY-L outperformed the corresponding single drug-loaded formulations E-L and Y-L in terms of containing primary tumor growth and improving survival in an immunocompetent syngeneic mouse model of RCC. EY-L also exhibited significantly higher sensitization of RCC cells towards radiation in vitro than E-L and Y-L. Additionally, EY-L sensitized RCC tumors towards radiation therapy in xenograft and murine RCC models. EY-L mediated induction of mitotic catastrophe via downregulation of multiple cell cycle checkpoints and DNA damage repair pathways could be responsible for the augmentation of radiation therapy.
    CONCLUSIONS: Taken together, our study demonstrated the efficacy of a strategic combination therapy in sensitizing RCC to radiation therapy via inhibition of DNA damage repair and a substantial increase in mitotic catastrophe. This combination therapy may find its use in the augmentation of radiation therapy during the treatment of RCC patients.
    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
    有丝分裂是细胞周期的关键阶段,由庞大的监管机构网络控制,以应对多种内部和外部因素。裂殖酵母裂殖酵母可能由于突变或药物治疗而表现出灾难性的有丝分裂表型。引起灾难性有丝分裂的因素之一是脂质代谢紊乱,由于例如乙酰辅酶A/生物素羧化酶(cut6)中的突变,在脂肪酸合成酶(fas2/lsd1)中,或在脂质代谢(cbf11)基因的转录调节因子中,以及用脂肪酸合成抑制剂治疗。先前表明,氯化铵可以部分挽救脂质代谢突变体中的有丝分裂保真度。在这项研究中,我们证明了多种氮源可以改善有丝分裂的保真度。此外,这种改善不仅限于脂质代谢紊乱,还适用于许多无关的有丝分裂突变体。有趣的是,部分挽救不是通过恢复脂质代谢状态来实现的,而是间接的。我们的结果强调了氮利用率在有丝分裂保真度中的新作用。
    Mitosis is a crucial stage in the cell cycle, controlled by a vast network of regulators responding to multiple internal and external factors. The fission yeast Schizosaccharomyces pombe demonstrates catastrophic mitotic phenotypes due to mutations or drug treatments. One of the factors provoking catastrophic mitosis is a disturbed lipid metabolism, resulting from, for example, mutations in the acetyl-CoA/biotin carboxylase (cut6), fatty acid synthase (fas2, also known as lsd1) or transcriptional regulator of lipid metabolism (cbf11) genes, as well as treatment with inhibitors of fatty acid synthesis. It has been previously shown that mitotic fidelity in lipid metabolism mutants can be partially rescued by ammonium chloride supplementation. In this study, we demonstrate that mitotic fidelity can be improved by multiple nitrogen sources. Moreover, this improvement is not limited to lipid metabolism disturbances but also applies to a number of unrelated mitotic mutants. Interestingly, the partial rescue is not achieved by restoring the lipid metabolism state, but rather indirectly. Our results highlight a novel role for nitrogen availability in mitotic fidelity.
    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
    胶质母细胞瘤(GBM),成人中最常见和最致命的脑癌,其特点是生存时间短、死亡率高。由于GBM细胞对常规治疗方法的抗性,科学兴趣集中在寻找替代和有效的辅助治疗。S-腺苷蛋氨酸(AdoMet),经过充分研究的生理甲基供体,已经成为一种有前途的抗癌化合物和多种癌症相关信号通路的调节剂。我们在这里首次报道了AdoMet选择性抑制U87MG的活力和增殖,U343MG,和U251MGGBM细胞。在这些细胞系中,AdoMet诱导S和G2/M细胞周期停滞和凋亡,并下调参与同源重组DNA修复的蛋白质的表达和激活,包括RAD51、BRCA1和Chk1。此外,AdoMet能够将DNA维持在受损状态,如增加的γH2AX/H2AX比所示。AdoMet通过抑制AuroraB激酶表达促进有丝分裂突变,磷酸化,和定位导致GBM细胞经历有丝分裂突变诱导的死亡。最后,AdoMet抑制DNA修复并诱导细胞周期停滞,凋亡,和患者来源的GBM细胞中的有丝分裂突变。鉴于这些结果,AdoMet可以被认为是GBM治疗中的潜在佐剂。
    Glioblastoma (GBM), the most frequent and lethal brain cancer in adults, is characterized by short survival times and high mortality rates. Due to the resistance of GBM cells to conventional therapeutic treatments, scientific interest is focusing on the search for alternative and efficient adjuvant treatments. S-Adenosylmethionine (AdoMet), the well-studied physiological methyl donor, has emerged as a promising anticancer compound and a modulator of multiple cancer-related signaling pathways. We report here for the first time that AdoMet selectively inhibited the viability and proliferation of U87MG, U343MG, and U251MG GBM cells. In these cell lines, AdoMet induced S and G2/M cell cycle arrest and apoptosis and downregulated the expression and activation of proteins involved in homologous recombination DNA repair, including RAD51, BRCA1, and Chk1. Furthermore, AdoMet was able to maintain DNA in a damaged state, as indicated by the increased γH2AX/H2AX ratio. AdoMet promoted mitotic catastrophe through inhibiting Aurora B kinase expression, phosphorylation, and localization causing GBM cells to undergo mitotic catastrophe-induced death. Finally, AdoMet inhibited DNA repair and induced cell cycle arrest, apoptosis, and mitotic catastrophe in patient-derived GBM cells. In light of these results, AdoMet could be considered a potential adjuvant in GBM therapy.
    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
    靶向polo-box结构域(PBD)小分子用于polo样激酶1(PLK1)抑制是靶激酶结构域(KD)的可行替代方案,这可以避免ATP竞争性抑制剂的泛选择性和剂量限制性毒性。然而,它们在这些环境中的疗效仍然很低,无法达到临床要求.在这里,我们利用基于结构的高通量虚拟筛选来寻找能够通过靶向PBD抑制PLK1的新型化学支架,并鉴定了初始命中分子化合物1a.基于铅化合物1a,进行了结构优化的方法,合成了几个系列的具有萘酰亚胺结构基序的衍生物。化合物4Bb被鉴定为新的有效PLK1抑制剂,其KD值为0.29μM。4Bb可以靶向PLK1PBD抑制PLK1活性,随后抑制PLK1与胞质分裂蛋白调节因子1(PRC1)的相互作用,最终导致耐药肺癌细胞的有丝分裂灾难。此外,4Bb可以用谷胱甘肽(GSH)的巯基进行亲核取代,以通过耗尽GSH来干扰氧化还原稳态。通过调节细胞周期机制和增加细胞氧化应激,4Bb对多种癌细胞和耐药癌细胞表现出有效的细胞毒性。皮下和口服4Bb可以有效抑制体内耐药肿瘤的生长,在正常组织中无副作用的荷瘤小鼠的存活时间加倍。因此,我们的研究提供了一种口服方法,用于耐药肺癌治疗的结构新型PLK1抑制剂。
    Targeting polo-box domain (PBD) small molecule for polo-like kinase 1 (PLK1) inhibition is a viable alternative to target kinase domain (KD), which could avoid pan-selectivity and dose-limiting toxicity of ATP-competitive inhibitors. However, their efficacy in these settings is still low and inaccessible to clinical requirement. Herein, we utilized a structure-based high-throughput virtual screen to find novel chemical scaffold capable of inhibiting PLK1 via targeting PBD and identified an initial hit molecule compound 1a. Based on the lead compound 1a, a structural optimization approach was carried out and several series of derivatives with naphthalimide structural motif were synthesized. Compound 4Bb was identified as a new potent PLK1 inhibitor with a KD value of 0.29 μM. 4Bb could target PLK1 PBD to inhibit PLK1 activity and subsequently suppress the interaction of PLK1 with protein regulator of cytokinesis 1 (PRC1), finally leading to mitotic catastrophe in drug-resistant lung cancer cells. Furthermore, 4Bb could undergo nucleophilic substitution with the thiol group of glutathione (GSH) to disturb the redox homeostasis through exhausting GSH. By regulating cell cycle machinery and increasing cellular oxidative stress, 4Bb exhibited potent cytotoxicity to multiple cancer cells and drug-resistant cancer cells. Subcutaneous and oral administration of 4Bb could effectively inhibit the growth of drug-resistant tumors in vivo, doubling the survival time of tumor bearing mice without side effects in normal tissues. Thus, our study offers an orally-available, structurally-novel PLK1 inhibitor for drug-resistant lung cancer therapy.
    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: Preprint
    Survivin蛋白在修复前中期不正确的微管-动粒附着中起作用,和胞质分裂的忠实执行,两者都是染色体乘客复合体(CPC)的一部分(1)。在这种情况下,错误经常导致非整倍性,多倍体和癌症(1)。加上这种蛋白质的这些众所周知的作用,这篇论文现在首次显示了Survivin是癌细胞进入有丝分裂所必需的,而且,在缺席的情况下,HeLa细胞在早期积累,或之前报告(2,3)。完整的核层的存在和低的Cdk1活性证明了这种早期的阻断(4)。重要的是,逃避Survivin废除诱导的逮捕导致多个有丝分裂缺陷,或有丝分裂灾难,最终细胞死亡。机械上,在没有Survivin的情况下,Cdk1不会定位于中心体。此外,重组Survivin蛋白可在体外通过Cdc25磷酸酶诱导Cdk1激酶的激活。此外,存活蛋白直接与Cdc25B亚型相互作用,在体外和体内,在它缺席的情况下,无活性的胞质Cdc25B-Cdk1-细胞周期蛋白B1复合物积累。最后,与Survivin在Cdk1早期激活中的作用一致,通过Survivin废除在HeLa细胞中诱导的早期前期积累可以被功能获得Cdc25B突变体绕过,驱使细胞重新进入有丝分裂。
    The Survivin protein has roles in repairing incorrect microtubule-kinetochore attachments at prometaphase, and the faithful execution of cytokinesis, both as part of the chromosomal passenger complex (CPC) (1). In this context, errors frequently lead to aneuploidy, polyploidy and cancer (1). Adding to these well-known roles of this protein, this paper now shows for the first time that Survivin is required for cancer cells to enter mitosis, and that, in its absence, HeLa cells accumulate at early prophase, or prior to reported before (2, 3). This early prophase blockage is demonstrated by the presence of an intact nuclear lamina and low Cdk1 activity (4). Importantly, escaping the arrest induced by Survivin abrogation leads to multiple mitotic defects, or mitotic catastrophe, and eventually cell death. Mechanistically, Cdk1 does not localize at the centrosome in the absence of Survivin pointing at an impairment in signaling through the Cdc25B-Cdk1 axis. In agreement, even though Survivin directly interacts with Cdc25B, both in vitro and in vivo, in its absence, an inactive cytosolic Cdc25B-Cdk1-Cyclin B1 complex accumulates. This flaw in Cdc25B activation can however be reversed in Survivin-depleted HeLa cell extracts to which the recombinant Survivin protein is added back. Finally, a role for Survivin in the Cdc25B-mediated activation of Cdk1 is confirmed by overriding the early prophase blockage induced in cells lacking Survivin through the expression of a gain-of-function Cdc25B mutant.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号