CDC2 Protein Kinase

CDC2 蛋白激酶
  • 文章类型: Journal Article
    组织来源的细胞外囊泡(EV)正在成为维持器官稳态的关键参与者,这显示了作为下一代候选药物的广泛来源。然而,组织EV的详细功能和治疗潜力仍未得到充分研究.这里,通过批量和单细胞RNA测序分析结合超微结构组织检查,我们首先揭示了原位肝组织EVs(LT-EVs)有助于部分肝切除术(PHX)后复杂的肝脏再生过程,肝细胞是再生肝脏中组织EV的主要来源。纳米级和蛋白质组学分析进一步确定肝细胞特异性组织EV(Hep-EV)在PHx后被增强以释放并携带增殖信息。此外,通过AAV-shRab27a在体内靶向抑制Hep-EV释放证实了Hep-EV是协调肝再生所需要的。机械上,来自再生肝脏的Hep-EV通过细胞周期依赖性激酶1(Cdk1)活性促进细胞周期进程,从而相互刺激肝细胞增殖。值得注意的是,补充来自再生肝脏的Hep-EV表现出翻译潜力并改善肝脏再生不足。这项研究提供了一个功能和机制框架,表明再生Hep-EV的释放控制着快速的肝脏再生,从而丰富了我们对器官再生和治疗中生理和内源性组织EV的理解。
    Tissue-derived extracellular vesicles (EVs) are emerging as pivotal players to maintain organ homeostasis, which show promise as a next-generation candidate for medical use with extensive source. However, the detailed function and therapeutic potential of tissue EVs remain insufficiently studied. Here, through bulk and single-cell RNA sequencing analyses combined with ultrastructural tissue examinations, we first reveal that in situ liver tissue EVs (LT-EVs) contribute to the intricate liver regenerative process after partial hepatectomy (PHx), and that hepatocytes are the primary source of tissue EVs in the regenerating liver. Nanoscale and proteomic profiling further identify that the hepatocyte-specific tissue EVs (Hep-EVs) are strengthened to release with carrying proliferative messages after PHx. Moreover, targeted inhibition of Hep-EV release via AAV-shRab27a in vivo confirms that Hep-EVs are required to orchestrate liver regeneration. Mechanistically, Hep-EVs from the regenerating liver reciprocally stimulate hepatocyte proliferation by promoting cell cycle progression through Cyclin-dependent kinase 1 (Cdk1) activity. Notably, supplementing with Hep-EVs from the regenerating liver demonstrates translational potential and ameliorates insufficient liver regeneration. This study provides a functional and mechanistic framework showing that the release of regenerative Hep-EVs governs rapid liver regeneration, thereby enriching our understanding of physiological and endogenous tissue EVs in organ regeneration and 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
    目的:我们的目的是在内镜黏膜下剥离术(ESD)治疗的早期胃癌(EGC)患者中鉴定异时性胃癌(MGC)的预测标志物。
    方法:来自接受ESD的EGC患者,在初次EGC诊断时,对非癌性胃粘膜样本进行大量RNA测序.这包括23名患有MGC的患者,和23名对照患者,没有额外的胃肿瘤超过3年(1:1匹配的年龄,性别,和幽门螺杆菌感染状态)。确定了候选的差异表达基因,使用实时定量聚合酶链反应和使用胃细胞系的细胞活力测定从中选择生物标志物。55名MGC患者和125名对照的独立验证队列用于标记验证。我们还检查了胃肠上皮化生的严重程度,一种已知的癌前病变,在初步诊断。
    结果:从发现队列中,确定了86个候选基因,其中选择KDF1和CDK1作为MGC的标记,这在验证队列中得到了证实。CERB5和AKT2同种型被鉴定为与肠上皮化生相关的标志物,并且与对照组相比,在MGC患者中也高表达(p<0.01)。将这些标志物与临床数据(年龄,性别,幽门螺杆菌和肠上皮化生的严重程度)得出MGC预测的曲线下面积(AUC)为0.91(95%CI,0.85-0.97)。
    结论:评估非癌性胃粘膜中的生物标志物可能是预测EGC患者中MGC和识别发生MGC风险较高的患者的有用方法,谁可以从严格的监控中受益。
    OBJECTIVE: We aimed to identify predictive markers for metachronous gastric cancer (MGC) in early gastric cancer (EGC) patients curatively treated with endoscopic submucosal dissection (ESD).
    METHODS: From EGC patients who underwent ESD, bulk RNA sequencing was performed on non-cancerous gastric mucosa samples at the time of initial EGC diagnosis. This included 23 patients who developed MGC, and 23 control patients without additional gastric neoplasms for over 3 years (1:1 matched by age, sex, and Helicobacter pylori infection state). Candidate differentially-expressed genes were identified, from which biomarkers were selected using real-time quantitative polymerase chain reaction and cell viability assays using gastric cell lines. An independent validation cohort of 55 MGC patients and 125 controls was used for marker validation. We also examined the severity of gastric intestinal metaplasia, a known premalignant condition, at initial diagnosis.
    RESULTS: From the discovery cohort, 86 candidate genes were identified of which KDF1 and CDK1 were selected as markers for MGC, which were confirmed in the validation cohort. CERB5 and AKT2 isoform were identified as markers related to intestinal metaplasia and were also highly expressed in MGC patients compared to controls (p < 0.01). Combining these markers with clinical data (age, sex, H. pylori and severity of intestinal metaplasia) yielded an area under the curve (AUC) of 0.91 (95% CI, 0.85-0.97) for MGC prediction.
    CONCLUSIONS: Assessing biomarkers in non-cancerous gastric mucosa may be a useful method for predicting MGC in EGC patients and identifying patients with a higher risk of developing MGC, who can benefit from rigorous surveillance.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Editorial
    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
    拓扑异构酶1(Top1)控制DNA拓扑,减轻复制和转录过程中的DNA超螺旋,并且对于有丝分裂进展到G1期至关重要。酪氨酸基-DNA磷酸二酯酶1(TDP1)介导捕获的Top1-DNA共价复合物(Top1cc)的去除。这里,我们鉴定了有丝分裂过程中TDP1在残基S61处的CDK1依赖性磷酸化。S61磷酸化缺陷的TDP1变体(TDP1-S61A)被困在有丝分裂染色体上,引发DNA损伤和有丝分裂缺陷。此外,我们表明,有丝分裂中的Top1cc修复是通过MUS81依赖性DNA修复机制发生的。喜树碱或蚜虫素诱导的复制胁迫导致TDP1-S61A在常见的脆性位点富集,过度刺激MUS81依赖性染色单体断裂,后期桥梁,和微核,最终在G1期形成53BP1核体。我们的发现为TDP1动力学的细胞周期依赖性调节提供了新的见解,以修复有丝分裂期间捕获的Top1-DNA共价复合物,从而防止复制应激后的基因组不稳定。
    Topoisomerase 1 (Top1) controls DNA topology, relieves DNA supercoiling during replication and transcription, and is critical for mitotic progression to the G1 phase. Tyrosyl-DNA phosphodiesterase 1 (TDP1) mediates the removal of trapped Top1-DNA covalent complexes (Top1cc). Here, we identify CDK1-dependent phosphorylation of TDP1 at residue S61 during mitosis. A TDP1 variant defective for S61 phosphorylation (TDP1-S61A) is trapped on the mitotic chromosomes, triggering DNA damage and mitotic defects. Moreover, we show that Top1cc repair in mitosis occurs via a MUS81-dependent DNA repair mechanism. Replication stress induced by camptothecin or aphidicolin leads to TDP1-S61A enrichment at common fragile sites, which over-stimulates MUS81-dependent chromatid breaks, anaphase bridges, and micronuclei, ultimately culminating in the formation of 53BP1 nuclear bodies during G1 phase. Our findings provide new insights into the cell cycle-dependent regulation of TDP1 dynamics for the repair of trapped Top1-DNA covalent complexes during mitosis that prevents genomic instability following replication stress.
    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
    含溴结构域蛋白4(BRD4)已成为前列腺癌(PCa)的有希望的治疗靶标。了解BRD4稳定性的机制可以增强BRD4靶向治疗的临床反应。在这项研究中,我们报道BRD4蛋白水平在有丝分裂过程中以PLK1依赖性方式显著降低.机械上,我们显示BRD4主要在T1186被CDK1/细胞周期蛋白B复合物磷酸化,招募PLK1在S24/S1100磷酸化BRD4,这被APC/CCdh1复合物识别用于蛋白酶体途径降解。我们发现PLK1过表达降低SPOP突变稳定的BRD4,从而使PCa细胞对BRD4抑制剂重新敏感。有趣的是,我们报道,多西他赛和JQ1的序贯治疗可显著抑制PCa.总的来说,结果支持PLK1磷酸化BRD4在M期触发其降解。多西他赛和JQ1的序贯治疗克服了BRD4积累相关的布罗莫结构域和末端外抑制剂(BETi)耐药性,这可能有助于制定治疗PCa的策略。
    Bromodomain-containing protein 4 (BRD4) has emerged as a promising therapeutic target in prostate cancer (PCa). Understanding the mechanisms of BRD4 stability could enhance the clinical response to BRD4-targeted therapy. In this study, we report that BRD4 protein levels are significantly decreased during mitosis in a PLK1-dependent manner. Mechanistically, we show that BRD4 is primarily phosphorylated at T1186 by the CDK1/cyclin B complex, recruiting PLK1 to phosphorylate BRD4 at S24/S1100, which are recognized by the APC/CCdh1 complex for proteasome pathway degradation. We find that PLK1 overexpression lowers SPOP mutation-stabilized BRD4, consequently rendering PCa cells re-sensitized to BRD4 inhibitors. Intriguingly, we report that sequential treatment of docetaxel and JQ1 resulted in significant inhibition of PCa. Collectively, the results support that PLK1-phosphorylated BRD4 triggers its degradation at M phase. Sequential treatment of docetaxel and JQ1 overcomes BRD4 accumulation-associated bromodomain and extra-terminal inhibitor (BETi) resistance, which may shed light on the development of strategies to treat PCa.
    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:食管癌(ESCA)是一种与慢性炎症和免疫失调相关的恶性肿瘤。然而,该疾病的特异性免疫状态和免疫调节的关键机制需要进一步探索。
    方法:探讨人ESCA肿瘤免疫微环境的特征及其可能的调控,我们通过飞行时间进行了质量细胞计数,单细胞RNA测序,组织的多色荧光染色,和流式细胞术分析来自未治疗患者的肿瘤和癌旁组织。
    结果:我们描绘了ESCA的免疫景观,并揭示了CD8+(组织常驻记忆CD8+T细胞(CD8+TRMs)与疾病进展密切相关。我们还揭示了CD8+TRMs在ESCA肿瘤微环境(TME)中的异质性,这与它们的分化和功能有关。此外,肿瘤中表达高水平颗粒酶B和免疫检查点的CD8+TRMs亚群(称为tTRMs)在晚期ESCA的TME中显著降低.我们表明tTRMs是在TME中预激活的肿瘤效应细胞。然后,我们证明了源自中间单核细胞(iMos)的常规树突状细胞(cDC2s)对于维持TME中CD8TRM的增殖至关重要。我们的初步研究表明,缺氧可以促进iMos的凋亡,阻碍cDC2s的成熟,这反过来又降低了CD8+TRM的增殖能力,从而促进癌症的进展。
    结论:我们的研究揭示了CD8TRMs的重要抗肿瘤作用,并初步探索了iMo/cDC2/CD8TRM免疫轴在人ESCATME中的调节作用。
    BACKGROUND: Esophageal cancer (ESCA) is a form of malignant tumor associated with chronic inflammation and immune dysregulation. However, the specific immune status and key mechanisms of immune regulation in this disease require further exploration.
    METHODS: To investigate the features of the human ESCA tumor immune microenvironment and its possible regulation, we performed mass cytometry by time of flight, single-cell RNA sequencing, multicolor fluorescence staining of tissue, and flow cytometry analyses on tumor and paracancerous tissue from treatment-naïve patients.
    RESULTS: We depicted the immune landscape of the ESCA and revealed that CD8+ (tissue-resident memory CD8+ T cells (CD8+ TRMs) were closely related to disease progression. We also revealed the heterogeneity of CD8+ TRMs in the ESCA tumor microenvironment (TME), which was associated with their differentiation and function. Moreover, the subset of CD8+ TRMs in tumor (called tTRMs) that expressed high levels of granzyme B and immune checkpoints was markedly decreased in the TME of advanced ESCA. We showed that tTRMs are tumor effector cells preactivated in the TME. We then demonstrated that conventional dendritic cells (cDC2s) derived from intermediate monocytes (iMos) are essential for maintaining the proliferation of CD8+ TRMs in the TME. Our preliminary study showed that hypoxia can promote the apoptosis of iMos and impede the maturation of cDC2s, which in turn reduces the proliferative capacity of CD8+ TRMs, thereby contributing to the progression of cancer.
    CONCLUSIONS: Our study revealed the essential antitumor roles of CD8+ TRMs and preliminarily explored the regulation of the iMo/cDC2/CD8+ TRM immune axis in the human ESCA TME.
    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
    前列腺癌(PCa)是美国男性中最常见的癌症,也是癌症相关死亡的主要原因。溶质载体家族14成员1(SLC14A1)是尿素转运蛋白的成员,对调节尿液浓度很重要。然而,SLC14A1在PCa中的生理意义仍不清楚。在本研究中,通过生物信息学分析和实验,我们发现SLC14A1的表达在PCa进展中显著降低,这可能归因于SLC14A1启动子区域的过度甲基化。此外,SLC14A1启动子的低表达和高甲基化与PCa患者的不良预后密切相关。另一方面,SLC14A1过表达抑制细胞增殖和转移,同时也抑制CDK1/CCNB1通路和mTOR/MMP-9信号通路。此外,SLC14A1表达在前列腺基底型细胞中富集。总之,我们的研究表明,SLC14A1的低表达水平和启动子甲基化可能是PCa进展和预后的新指标,SLC14A1可抑制PCa的进展。
    Prostate cancer (PCa) is the most common cancer among men in the United States and the leading cause of cancer-related death. The Solute Carrier Family 14 Member 1 (SLC14A1) is a member of urea transporters which are important for the regulation of urine concentration. However, the physiological significance of SLC14A1 in PCa still remains unclear. In the present study, via bioinformatics analysis and experiments, we found that expression of SLC14A1 is significantly decreased in PCa progression, which could be attributed to hypermethylation on SLC14A1 promoter region. Moreover, its low expression and hypermethylation on SLC14A1 promoter are closely related to the poor prognosis of PCa patients. On the other hand, overexpression of SLC14A1 inhibited cell proliferation and metastasis while its overexpression also suppressed CDK1/CCNB1 pathway and mTOR/MMP-9 signaling pathway. Additionally, SLC14A1 expression is enriched in prostate basal-type cells. In summary, our study indicates that its low expression level and promoter hypermethylation of SLC14A1 may represent novel indicators for PCa progression and prognosis, and SLC14A1 could inhibit the progression of PCa.
    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
    发育过程中不同细胞命运的产生取决于祖细胞的不对称细胞分裂。在果蝇的中枢和周围神经系统中,分别称为成神经细胞或感觉器官前体的祖细胞在有丝分裂期间使用PAR极性来控制其子细胞中的细胞命运决定。极性和细胞周期是如何耦合的,以及细胞周期机制如何调节PAR蛋白功能和细胞命运决定尚不清楚。这里,我们产生了CDK1的类似敏感等位基因,并揭示了其部分抑制作用减弱但不会消除胚胎和幼虫神经母细胞的顶端极性,并导致命运决定子极化的缺陷。我们描述了一种新的Bazooka体内磷酸化,果蝇PAR-3的同源物,在Serine180上,一个共有的CDK磷酸化位点。在某些组织背景下,Serine180的磷酸化发生在不对称分裂的细胞中,但不发生在对称分裂的邻居中。在成神经细胞中,Serine180磷酸盐破坏基底极化的时间。Serine180磷酸盐还影响感觉器官前体的规格和二元细胞命运确定以及Baz在其不对称细胞分裂过程中的定位。最后,我们显示CDK1在体外磷酸化人PAR-3上的丝氨酸-S180和等效丝氨酸。
    The generation of distinct cell fates during development depends on asymmetric cell division of progenitor cells. In the central and peripheral nervous system of Drosophila, progenitor cells respectively called neuroblasts or sensory organ precursors use PAR polarity during mitosis to control cell fate determination in their daughter cells. How polarity and the cell cycle are coupled, and how the cell cycle machinery regulates PAR protein function and cell fate determination is poorly understood. Here, we generate an analog sensitive allele of CDK1 and reveal that its partial inhibition weakens but does not abolish apical polarity in embryonic and larval neuroblasts and leads to defects in polarisation of fate determinants. We describe a novel in vivo phosphorylation of Bazooka, the Drosophila homolog of PAR-3, on Serine180, a consensus CDK phosphorylation site. In some tissular contexts, phosphorylation of Serine180 occurs in asymmetrically dividing cells but not in their symmetrically dividing neighbours. In neuroblasts, Serine180 phosphomutants disrupt the timing of basal polarisation. Serine180 phosphomutants also affect the specification and binary cell fate determination of sensory organ precursors as well as Baz localisation during their asymmetric cell divisions. Finally, we show that CDK1 phosphorylates Serine-S180 and an equivalent Serine on human PAR-3 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
    背景:胶质母细胞瘤(GBM)是一种高度侵袭性和普遍性的脑肿瘤,对治疗提出了重大挑战。SRSF9,一种RNA结合蛋白,对细胞过程至关重要,并与癌症进展有关。然而,其在GBM中的功能和机制有待澄清。
    方法:进行生物信息学分析,以探讨SRSF9在GBM中的差异表达及其与胶质瘤患者预后的相关性。通过RT-qPCR定量GBM细胞系和患者组织中的SRSF9和CDK1表达,蛋白质印迹或免疫荧光测定。MTT法评价SRSF9在GBM细胞增殖和迁移中的作用,Transwell和集落形成测定。此外,使用ChIP-PCR和双荧光素酶测定法研究了SRSF9对CDK1的转录调控。
    结果:SRSF9表达升高与GBM分期和胶质瘤患者的低生存率相关。通过功能增益和功能损失策略,证明SRSF9促进GBM细胞的增殖和迁移。生物信息学分析表明,SRSF9对细胞生长途径有影响,包括细胞周期检查点和E2F靶标。机械上,SRSF9似乎与CDK1基因的启动子结合并增加其转录水平,从而促进GBM细胞增殖。
    结论:这些发现揭示了SRSF9在GBM中的细胞功能,并强调了其对GBM的治疗潜力。
    BACKGROUND: Glioblastoma (GBM) is a highly aggressive and prevalent brain tumor that poses significant challenges in treatment. SRSF9, an RNA-binding protein, is essential for cellular processes and implicated in cancer progression. Yet, its function and mechanism in GBM need clarification.
    METHODS: Bioinformatics analysis was performed to explore differential expression of SRSF9 in GBM and its prognostic relevance to glioma patients. SRSF9 and CDK1 expression in GBM cell lines and patients\' tissues were quantified by RT-qPCR, Western blot or immunofluorescence assay. The role of SRSF9 in GBM cell proliferation and migration was assessed by MTT, Transwell and colony formation assays. Additionally, transcriptional regulation of CDK1 by SRSF9 was investigated using ChIP-PCR and dual-luciferase assays.
    RESULTS: The elevated SRSF9 expression correlates to GBM stages and poor survival of glioma patients. Through gain-of-function and loss-of-function strategies, SRSF9 was demonstrated to promote proliferation and migration of GBM cells. Bioinformatics analysis showed that SRSF9 has an impact on cell growth pathways including cell cycle checkpoints and E2F targets. Mechanistically, SRSF9 appears to bind to the promoter of CDK1 gene and increase its transcription level, thus promoting GBM cell proliferation.
    CONCLUSIONS: These findings uncover the cellular function of SRSF9 in GBM and highlight its therapeutic potential for GBM.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号