CDC20

Cdc20
  • 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
    通过利用机器学习,我们可以识别与复发相关的基因,入侵,和肿瘤干性,从而发现新的治疗靶点。
    首先,我们从GEO数据库中获得了与复发和侵袭相关的基因集,一个全面的基因表达数据库。然后我们采用加权基因共表达网络分析(WGCNA)来鉴定核心基因模块并对其进行功能富集分析。接下来,我们利用随机森林和随机生存森林算法来计算关键模块内的基因,从而鉴定出三个关键基因。随后,使用Kaplan-Meier工具选择这些关键基因之一进行预后分析和潜在药物筛选.最后,为了检查CDC20在肺腺癌(LUAD)中的作用,我们进行了各种体外和体内实验,包括伤口愈合试验,集落形成试验,Transwell迁移分析,流式细胞术细胞周期分析,西方印迹,和小鼠肿瘤模型实验。
    首先,我们从两个数据集中总共收集了279个样本,GSE166722和GSE31210,鉴定91个与复发相关的差异表达基因,入侵,肺腺癌的干性。功能富集分析表明,这些关键基因簇主要参与微管结合,主轴,染色体区域,细胞器裂变,核分裂。接下来,使用机器学习,我们鉴定并验证了三个hub基因(CDC45,CDC20,TPX2),CDC20与肿瘤干性的相关性最高,既往研究有限。此外,我们发现CDC20与临床病理特征密切相关,总体生存率(OS)较差,无进展间隔(PFI),无进展生存期(PFS),肺腺癌患者的不良预后。最后,我们的功能研究表明,敲低CDC20可以抑制癌细胞的迁移,入侵,扩散,细胞周期进程,和肿瘤的生长可能通过MAPK信号通路。
    CDC20已成为监测治疗反应的新型生物标志物,复发,和肺腺癌患者的疾病进展。由于其重要性,需要进一步研究CDC20作为潜在的治疗靶点.研究CDC20的作用可以为开发新的治疗方法和改善患者预后提供有价值的见解。
    UNASSIGNED: By utilizing machine learning, we can identify genes that are associated with recurrence, invasion, and tumor stemness, thus uncovering new therapeutic targets.
    UNASSIGNED: To begin, we obtained a gene set related to recurrence and invasion from the GEO database, a comprehensive gene expression database. We then employed the Weighted Gene Co-expression Network Analysis (WGCNA) to identify core gene modules and perform functional enrichment analysis on them. Next, we utilized the random forest and random survival forest algorithms to calculate the genes within the key modules, resulting in the identification of three crucial genes. Subsequently, one of these key genes was selected for prognosis analysis and potential drug screening using the Kaplan-Meier tool. Finally, in order to examine the role of CDC20 in lung adenocarcinoma (LUAD), we conducted a variety of in vitro and in vivo experiments, including wound healing assay, colony formation assays, Transwell migration assays, flow cytometric cell cycle analysis, western blotting, and a mouse tumor model experiment.
    UNASSIGNED: First, we collected a total of 279 samples from two datasets, GSE166722 and GSE31210, to identify 91 differentially expressed genes associated with recurrence, invasion, and stemness in lung adenocarcinoma. Functional enrichment analysis revealed that these key gene clusters were primarily involved in microtubule binding, spindle, chromosomal region, organelle fission, and nuclear division. Next, using machine learning, we identified and validated three hub genes (CDC45, CDC20, TPX2), with CDC20 showing the highest correlation with tumor stemness and limited previous research. Furthermore, we found a close association between CDC20 and clinical pathological features, poor overall survival (OS), progression-free interval (PFI), progression-free survival (PFS), and adverse prognosis in lung adenocarcinoma patients. Lastly, our functional research demonstrated that knocking down CDC20 could inhibit cancer cell migration, invasion, proliferation, cell cycle progression, and tumor growth possibly through the MAPK signaling pathway.
    UNASSIGNED: CDC20 has emerged as a novel biomarker for monitoring treatment response, recurrence, and disease progression in patients with lung adenocarcinoma. Due to its significance, further research studying CDC20 as a potential therapeutic target is warranted. Investigating the role of CDC20 could lead to valuable insights for developing new treatments and improving patient outcomes.
    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
    在酿酒酵母中,有两个翻译终止因素,eRF1(Sup45)和eRF3(Sup35),这对生存能力至关重要。先前的研究表明,这些基因中无义突变的存在会导致突变等位基因(sup35-n和sup45-n)的扩增,这似乎是这种细胞存活所必需的。然而,这种现象的机制尚不清楚。在这项研究中,我们使用RNA-Seq和蛋白质组分析揭示了在细胞适应sup35-218无义等位基因引入过程中发生的全套基因表达变化。我们的分析表明,控制细胞周期的基因转录发生了显着变化:后期促进复合物APC/C(APC9,CDC23)及其激活剂CDC20的基因表达减少,转录因子FKH1的表达增加,主要的细胞周期激酶CDC28和诱导DNA生物合成的细胞周期蛋白。我们提出了一个模型,根据该模型,酵母对翻译终止因子基因中的无义突变的适应是由于细胞周期进程延迟超过G2-M阶段而发生的。这导致S和G2期的扩展以及突变体sup35-n等位基因的拷贝数增加。
    In yeast Saccharomyces cerevisiae, there are two translation termination factors, eRF1 (Sup45) and eRF3 (Sup35), which are essential for viability. Previous studies have revealed that presence of nonsense mutations in these genes leads to amplification of mutant alleles (sup35-n and sup45-n), which appears to be necessary for the viability of such cells. However, the mechanism of this phenomenon remained unclear. In this study, we used RNA-Seq and proteome analysis to reveal the complete set of gene expression changes that occur during cellular adaptation to the introduction of the sup35-218 nonsense allele. Our analysis demonstrated significant changes in the transcription of genes that control the cell cycle: decreases in the expression of genes of the anaphase promoting complex APC/C (APC9, CDC23) and their activator CDC20, and increases in the expression of the transcription factor FKH1, the main cell cycle kinase CDC28, and cyclins that induce DNA biosynthesis. We propose a model according to which yeast adaptation to nonsense mutations in the translation termination factor genes occurs as a result of a delayed cell cycle progression beyond the G2-M stage, which leads to an extension of the S and G2 phases and an increase in the number of copies of the mutant sup35-n allele.
    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:慢性粒细胞白血病干细胞(CML-LSCs)被认为是酪氨酸激酶抑制剂(TKIs)耐药和CML复发的主要诱因。泛素化,翻译后修饰,与慢性粒细胞白血病的恶化过程有关。更详细地了解他们的串扰需要进一步调查。我们的研究旨在利用生物信息学分析探索CML-LSC中潜在的泛素化相关基因,这些基因可能是根除LSCs的靶标。方法:从GSE47927和iUCD数据库获得正常造血干细胞(HSC)和LSC之间的泛素化修饰相关差异表达基因(UUC-DEGs)。随后,利用STRING数据库和Cytoscape平台内的MCODE插件,通过蛋白质-蛋白质相互作用(PPI)网络分析鉴定了中心UUC-DEGs.hTFtarget研究了集线器UUC-DEGs的上游调节网络,PROMO,miRDB和miRWalk数据库。然后通过CIBERSORT算法和“ggcorrplot”软件包分析集线器UUC-DEGS与免疫细胞之间的相关性。最后,我们验证了集线器UUC-DEGs在CML动物模型中的功能,GSE24739数据集的CML细胞系和CD34+细胞。结果:4个hubUUC基因之间存在很强的关联(AURKA,Fancd2,Cdc20和Uhrf1)的LSCs和CD4+/CD8+T细胞的浸润,NK细胞和单核细胞。构建了8个TFs和23个潜在靶向这4个hub基因的miRNA。在这些中枢基因中,发现Fancd2,Cdc20和Uhrf1在CML-LSC中高度表达,敲低导致CML细胞增殖的显著抑制。结论:从生物信息学分析的角度,UHRF1和CDC20被鉴定为CML-LSCs和CML发病机制中的新的关键泛素化相关基因。
    Purpose: Chronic myeloid leukemia stem cells (CML-LSCs) are posited as the primary instigators of resistance to tyrosine kinase inhibitors (TKIs) and recurrence of CML. Ubiquitination, a post-translational modification, has been implicated in the worsening process of CML. A more detailed understanding of their crosstalk needs further investigation. Our research aims to explore the potential ubiquitination-related genes in CML-LSC using bioinformatics analysis that might be the target for the eradication of LSCs. Methods: The ubiquitination modification-related differentially expressed genes (UUC-DEGs) between normal hematopoietic stem cells (HSCs) and LSCs were obtained from GSE47927 and iUUCD database. Subsequently, the hub UUC-DEGs were identified through protein-protein interaction (PPI) network analysis utilizing the STRING database and the MCODE plug-in within the Cytoscape platform. The upstream regulation network of the hub UUC-DEGs was studied by hTFtarget, PROMO, miRDB and miRWalk databases respectively. Then the correlation between the hub UUC-DEGs and the immune cells was analyzed by the CIBERSORT algorithm and \"ggcorrplot\" package. Finally, we validated the function of hub UUC-DEGs in CML animal models, CML cell lines and CD34+ cells of the GSE24739 dataset. Results: There is a strong association between the 4 hub UUC genes (AURKA, Fancd2, Cdc20 and Uhrf1) of LSCs and the infiltration of CD4+/CD8+ T cells, NK cells and monocytes. 8 TFs and 23 miRNAs potentially targeted these 4 hub genes were constructed. Among these hub genes, Fancd2, Cdc20 and Uhrf1 were found to be highly expressed in CML-LSC, which knocking down resulted in significant inhibition of CML cell proliferation. Conclusions: From the perspective of bioinformatics analysis, UHRF1 and CDC20 were identified as the novel key ubiquitination-related genes in CML-LSCs and the pathogenesis of CML.
    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
    纺锤体组装检查点(SAC)通过监测动粒-微管附着来确保细胞分裂过程中忠实的染色体分离。植物产生序列保守和发散的SAC成分,SAC激活如何导致这些蛋白质在未连接的动体上组装以防止细胞进入后期,这在很大程度上是未知的。在拟南芥中,在整个有丝分裂过程中,在动子上检测到非经典BUB3.3蛋白,与仅与未连接染色体相关的MAD1和植物特异性BUB1/MAD3家族蛋白BMF3不同。当BUB3.3因基因突变而丢失时,有丝分裂细胞经常进入后期,染色体错位,并在被低剂量的微管解聚剂oryzalin攻击后呈现滞后的染色体,导致微核的形成。令人惊讶的是,其他SAC蛋白的动粒定位不需要BUB3.3,反之亦然。相反,BUB3.3通过两个内部重复基序与BMF3特异性结合,这两个内部重复基序不是BMF3动子定位所必需的。这种相互作用使BMF3能够招募CDC20,一个下游的SAC目标,独立的动臂。一起来看,我们的发现表明,植物SAC利用非常规的蛋白质相互作用来阻止有丝分裂,随着BUB3.3指导BMF3在CDC20招募中的角色,而不是在真菌和动物中观察到的BUB1/MAD3蛋白的募集。这种独特的机制突出了植物如何适应保守细胞周期机制的不同版本以实现专门的SAC控制。
    The spindle assembly checkpoint (SAC) ensures faithful chromosome segregation during cell division by monitoring kinetochore-microtubule attachment. Plants produce both sequence-conserved and diverged SAC components, and it has been largely unknown how SAC activation leads to the assembly of these proteins at unattached kinetochores to prevent cells from entering anaphase. In Arabidopsis thaliana, the noncanonical BUB3.3 protein was detected at kinetochores throughout mitosis, unlike MAD1 and the plant-specific BUB1/MAD3 family protein BMF3 that associated with unattached chromosomes only. When BUB3.3 was lost by a genetic mutation, mitotic cells often entered anaphase with misaligned chromosomes and presented lagging chromosomes after they were challenged by low doses of the microtubule depolymerizing agent oryzalin, resulting in the formation of micronuclei. Surprisingly, BUB3.3 was not required for the kinetochore localization of other SAC proteins or vice versa. Instead, BUB3.3 specifically bound to BMF3 through two internal repeat motifs that were not required for BMF3 kinetochore localization. This interaction enabled BMF3 to recruit CDC20, a downstream SAC target, to unattached kinetochores. Taken together, our findings demonstrate that plant SAC utilizes unconventional protein interactions for arresting mitosis, with BUB3.3 directing BMF3\'s role in CDC20 recruitment, rather than the recruitment of BUB1/MAD3 proteins observed in fungi and animals. This distinct mechanism highlights how plants adapted divergent versions of conserved cell cycle machinery to achieve specialized SAC control.
    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
    SETDB2是准确染色体分离所需的H3K9组蛋白甲基转移酶。据报道,其H3K9组蛋白甲基转移酶活性在中期与染色体有关。这里,我们确认SETDB2是有丝分裂和准确染色体分离所必需的。然而,这些功能与其组蛋白甲基转移酶活性无关。进一步分析表明,SETDB2可以与BUBR1相互作用,并且是CDC20与BUBR1和APC/C复合物结合以及CYCLINB1降解所必需的。SETDB2调节CDC20与BUBR1或APC/C复合物结合的能力,细胞周期蛋白B1的稳定性也独立于其组蛋白甲基转移酶的活性。这些结果表明,SETDB2与BUBR1相互作用以促进CDC20与BUBR1和APC3的结合,然后降解CYCLINB1以确保准确的染色体分离和有丝分裂,独立于其组蛋白甲基转移酶活性。
    SETDB2 is a H3K9 histone methyltransferase required for accurate chromosome segregation. Its H3K9 histone methyltransferase activity was reported to be associated with chromosomes during metaphase. Here, we confirm that SETDB2 is required for mitosis and accurate chromosome segregation. However, these functions are independent of its histone methyltransferase activity. Further analysis showed that SETDB2 can interact with BUBR1, and is required for CDC20 binding to BUBR1 and APC/C complex and CYCLIN B1 degradation. The ability of SETDB2 to regulate the binding of CDC20 to BUBR1 or APC/C complex, and stabilization of CYCLIN B1 are also independent of its histone methyltransferase activity. These results suggest that SETDB2 interacts with BUBR1 to promote binding of CDC20 to BUBR1 and APC3, then degrades CYCLIN B1 to ensure accurate chromosome segregation and mitosis, independently of its histone methyltransferase activity.
    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
    暂无摘要。
    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:HeberFERON是α2b和γ干扰素的共同制剂,基于它们的协同作用,这表明其在基底细胞癌中的临床优势优于单个干扰素。在胶质母细胞瘤(GBM)中,HeberFEON已显示出有希望的临床前和临床结果。这导致我们设计了一个微阵列实验,旨在鉴定与U-87MG模型中单个干扰素相比,HeberFERON的独特作用所涉及的分子机制。
    方法:转录表达谱包括对照组(未治疗)和接受α2b-干扰素的三组,使用IlluminaHT-12微阵列平台进行γ-干扰素和HeberFERON。无监督的基因和样本分组方法,差异表达基因的鉴定,应用功能富集和网络分析计算生物学方法来鉴定HeberFERON的独特转录模式。大多数代表性基因的验证通过qPCR进行。对于用HeberFERON处理24小时的细胞的细胞周期分析,48和72小时,我们使用流式细胞术。
    结果:基于基因表达谱,三种处理显示出不同的行为。富集分析确定了几个有丝分裂细胞周期相关事件,特别是从前中期到后期,这是Heberferon的唯一目标。FOXM1转录因子网络参与多个细胞周期阶段,在GBM中高度表达,大幅下调。流式细胞术实验证实了HeberFERON以剂量和时间依赖性方式对细胞周期的作用,治疗后24小时有明显的细胞停滞。尽管p53没有下调,在功能上富集了几个参与其调节活性的基因。网络分析还揭示了p53与HeberFERON靶向基因的强关系。我们提出了一个机械模型来解释这种独特的行为,基于PKR和ATF3的同时激活,p53磷酸化改变,以及其减少的MDM2介导的泛素化和从细胞核到细胞质的输出。PLK1,AURKB,BIRC5和CCNB1基因,所有这些都由FOXM1监管,在这一模式中也起着核心作用。这些和其他相互作用可以解释G2/M阻滞和HeberFERON对U-87MG增殖的影响。
    结论:我们提出了与U-87MG模型中单个干扰素治疗相比,HeberFERON独特行为的分子机制,其中细胞周期相关事件高度相关。
    BACKGROUND: HeberFERON is a co-formulation of α2b and γ interferons, based on their synergism, which has shown its clinical superiority over individual interferons in basal cell carcinomas. In glioblastoma (GBM), HeberFERON has displayed promising preclinical and clinical results. This led us to design a microarray experiment aimed at identifying the molecular mechanisms involved in the distinctive effect of HeberFERON compared to the individual interferons in U-87MG model.
    METHODS: Transcriptional expression profiling including a control (untreated) and three groups receiving α2b-interferon, γ-interferon and HeberFERON was performed using an Illumina HT-12 microarray platform. Unsupervised methods for gene and sample grouping, identification of differentially expressed genes, functional enrichment and network analysis computational biology methods were applied to identify distinctive transcription patterns of HeberFERON. Validation of most representative genes was performed by qPCR. For the cell cycle analysis of cells treated with HeberFERON for 24 h, 48 and 72 h we used flow cytometry.
    RESULTS: The three treatments show different behavior based on the gene expression profiles. The enrichment analysis identified several mitotic cell cycle related events, in particular from prometaphase to anaphase, which are exclusively targeted by HeberFERON. The FOXM1 transcription factor network that is involved in several cell cycle phases and is highly expressed in GBMs, is significantly down regulated. Flow cytometry experiments corroborated the action of HeberFERON on the cell cycle in a dose and time dependent manner with a clear cellular arrest as of 24 h post-treatment. Despite the fact that p53 was not down-regulated, several genes involved in its regulatory activity were functionally enriched. Network analysis also revealed a strong relationship of p53 with genes targeted by HeberFERON. We propose a mechanistic model to explain this distinctive action, based on the simultaneous activation of PKR and ATF3, p53 phosphorylation changes, as well as its reduced MDM2 mediated ubiquitination and export from the nucleus to the cytoplasm. PLK1, AURKB, BIRC5 and CCNB1 genes, all regulated by FOXM1, also play central roles in this model. These and other interactions could explain a G2/M arrest and the effect of HeberFERON on the proliferation of U-87MG.
    CONCLUSIONS: We proposed molecular mechanisms underlying the distinctive behavior of HeberFERON compared to the treatments with the individual interferons in U-87MG model, where cell cycle related events were highly relevant.
    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:越来越多的证据表明,尤其是免疫检查点抑制剂(ICIs),除前列腺癌外,有可能促进各种癌症的长期生存。新出现的证据表明焦亡,细胞死亡的免疫原性形式,可以触发抗肿瘤免疫微环境并增强免疫治疗的有效性。然而,前列腺癌中焦凋亡信号调节的潜在机制尚不清楚.
    方法:从五个独立的公共数据集中综合分析前列腺癌中人类E3连接酶的差异表达。此外,来自前列腺癌患者的组织微阵列的免疫组织化学分析证实了生物信息学分析的结果.此外,通过下一代RNA测序评估前列腺癌细胞系,以评估CDC20缺失后的转录组概况.接下来,qRT-PCR,西方印迹,环己酰亚胺测定,免疫沉淀,采用泛素化检测方法探讨CDC20与GSDME的相关性和相互作用。免疫缺陷和免疫活性小鼠模型均用于检查有或没有抗PD1抗体的CDC20抑制的抗肿瘤功效。分别。为了分析异种移植物的免疫微环境,用免疫组织化学和流式细胞术检测肿瘤组织。
    结果:对多个前列腺癌队列的分析提示CDC20是最显著的过表达E3连接酶。此外,CDC20通过靶向GSDME以依赖degron的方式进行泛素化介导的蛋白水解,对焦亡途径产生负调节作用。CDC20的敲低导致GSDME丰度增加以及响应于死亡信号从细胞凋亡到焦亡的转变。此外,在我们的同基因鼠模型中,我们发现CDC20的耗竭通过促进CD8+T淋巴细胞的浸润而显著增强抗肿瘤免疫,以及减少髓系免疫细胞。更重要的是,Apcin,一种靶向CDC20的小分子抑制剂,在小鼠前列腺癌模型中与基于抗PD1的免疫疗法表现出协同作用.
    结论:总体而言,这些发现为CDC20对GSDME介导的焦亡的上游调节提供了新的见解,CDC20与GSDME特异性相互作用,并以依赖degron的方式促进其泛素化.重要的是,我们的数据突出了靶向细胞焦亡和增强基于抗PD1的免疫疗法有效性的新分子途径.
    BACKGROUND: Increasing evidence suggests that immunotherapy, especially immune checkpoint inhibitors (ICIs), has the potential to facilitate long-term survival in various cancer besides prostate cancer. Emerging evidence indicated that pyroptosis, an immunogenic form of cell death, could trigger an anti-tumor immune microenvironment and enhance the effectiveness of immunotherapy. Nevertheless, the mechanism underlying the regulation of pyroptosis signaling in prostate cancer remains unclear.
    METHODS: The differential expression of human E3 ligases in prostate cancer was integratedly analyzed from five independent public datasets. Moreover, the immunohistochemistry analysis of a tissue microarray derived from prostate cancer patients confirmed the results from the bioinformatic analysis. Furthermore, prostate cancer cell lines were evaluated via the next-generation RNA sequencing to assess transcriptomic profile upon CDC20 depletion. Next, qRT-PCR, Western blotting, cycloheximide assay, immunoprecipitation, and ubiquitination assay were employed to explore the correlation and interaction between CDC20 and GSDME. Both immune-deficient and immune-competent murine models were utilized to examine the anti-tumor efficacy of CDC20 inhibition with or without the anti-PD1 antibodies, respectively. To analyze the immune microenvironment of the xenografts, the tumor tissues were examined by immunohistochemistry and flow cytometry.
    RESULTS: The analysis of multiple prostate cancer cohorts suggested that CDC20 was the most significantly over-expressed E3 ligase. In addition, CDC20 exerted a negative regulatory effect on the pyroptosis pathway by targeting GSDME for ubiquitination-mediated proteolysis in a degron-dependent manner. Knockdown of CDC20 leads to increased GSDME abundance and a transition from apoptosis to pyroptosis in response to death signals. Furthermore, in our syngeneic murine models, we found that depletion of CDC20 significantly enhances the anti-tumor immunity by promoting the infiltration of CD8+ T lymphocytes dependent on the existence of GSDME, as well as reducing myeloid immune cells. More importantly, Apcin, a small molecular inhibitor that targets CDC20, exhibited synergistic effects with anti-PD1-based immunotherapy in murine models of prostate cancer.
    CONCLUSIONS: Overall, these findings provide new insights into the upstream regulation of GSDME-mediated pyroptosis by CDC20, which specifically interacts with GSDME and facilitates its ubiquitination in a degron-dependent manner. Importantly, our data highlight novel molecular pathways for targeting cellular pyroptosis and enhancing the effectiveness of anti-PD1-based immunotherapy.
    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
    甲基转移酶KMT5A已被认为是前列腺癌中的癌基因,因此代表了推定的治疗靶标。为了证实这个假设,我们对在转录活性雄激素受体(AR)存在下KMT5A敲低后雄激素非依赖性前列腺癌细胞系模型进行了微阵列研究,以了解在活性AR存在下哪些基因和细胞过程受KMT5A调控.我们观察到当KMT5A表达降低时,301个基因下调,而408个基因上调。KEGG通路和基因本体论分析显示,响应于KMT5A敲低,凋亡和DNA损伤信号上调,而蛋白折叠和RNA剪接下调。在这些条件下,发现最重要的非AR调节基因是CDC20,CDC20是纺锤体组装检查点的关键调节因子,在几种癌症类型中具有致癌作用.进一步的研究表明,KMT5A以甲基转移酶依赖性方式调节CDC20,以调节其启动子区域内的组蛋白H4K20甲基化,并通过p53信号通路间接调节。在临床前列腺癌样本中也观察到KMT5A和CDC20表达之间的正相关。进一步支持这个协会。因此,我们得出的结论是,KMT5A是前列腺癌治疗的有效治疗靶点,而CDC20有可能作为有效治疗靶点的生物标志物.
    The methyltransferase KMT5A has been proposed as an oncogene in prostate cancer and therefore represents a putative therapeutic target. To confirm this hypothesis, we have performed a microarray study on a prostate cancer cell line model of androgen independence following KMT5A knockdown in the presence of the transcriptionally active androgen receptor (AR) to understand which genes and cellular processes are regulated by KMT5A in the presence of an active AR. We observed that 301 genes were down-regulated whilst 408 were up-regulated when KMT5A expression was reduced. KEGG pathway and gene ontology analysis revealed that apoptosis and DNA damage signalling were up-regulated in response to KMT5A knockdown whilst protein folding and RNA splicing were down-regulated. Under these conditions, the top non-AR regulated gene was found to be CDC20, a key regulator of the spindle assembly checkpoint with an oncogenic role in several cancer types. Further investigation revealed that KMT5A regulates CDC20 in a methyltransferase-dependent manner to modulate histone H4K20 methylation within its promoter region and indirectly via the p53 signalling pathway. A positive correlation between KMT5A and CDC20 expression was also observed in clinical prostate cancer samples, further supporting this association. Therefore, we conclude that KMT5A is a valid therapeutic target for the treatment of prostate cancer and CDC20 could potentially be utilised as a biomarker for effective therapeutic targeting.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号