Ubiquitylation

泛素化
  • 文章类型: Journal Article
    癌症干细胞(CSC)负责肿瘤的化学耐药性,芳香烃受体(AHR)对于维持CSC特性是不可缺少的。这里,我们旨在研究孕激素受体膜成分1(PGRMC1)和AHR之间的相互作用如何有助于非小细胞肺癌(NSCLC)中CSC表型的维持.临床数据和组织微阵列分析表明PGRMC1表达升高的患者预后较差。此外,PGRMC1过表达通过调节AHR泛素化在体外和体内增强CSC表型和化疗抗性。然后我们确定了PGRMC1和AHR之间的特异性相互作用位点。质谱筛选鉴定了含有56(TRIM56)作为E3连接酶靶向AHR的三方基序。值得注意的是,PGRMC1过表达抑制了TRIM56和AHR之间的相互作用。总的来说,我们的研究揭示了一种涉及PGRMC1,AHR,和TRIM56,为开发NSCLC治疗中的CSC靶向策略提供了见解。
    Cancer stem cells (CSCs) are responsible for tumor chemoresistance, and the aryl hydrocarbon receptor (AHR) is indispensable for maintaining CSC characteristics. Here, we aimed to investigate how the interaction between progesterone receptor membrane component 1 (PGRMC1) and AHR contributes to the maintenance of CSC phenotypes in non-small cell lung cancer (NSCLC). Clinical data and tissue microarray analyses indicated that patients with elevated PGRMC1 expression had poorer prognoses. Moreover, PGRMC1 overexpression enhanced CSC phenotypes and chemotherapy resistance in vitro and in vivo by modulating AHR ubiquitination. We then determined the specific interaction sites between PGRMC1 and AHR. Mass spectrometry screening identified tripartite motif containing 56 (TRIM56) as the E3 ligase targeting AHR. Notably, PGRMC1 overexpression inhibited the interaction between TRIM56 and AHR. Overall, our study revealed a regulatory mechanism that involves PGRMC1, AHR, and TRIM56, providing insights for developing CSC-targeting strategies in NSCLC 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
    植物免疫稳态是通过平衡的免疫激活和抑制来实现的。在避免自身免疫的同时进行有效的防御.在拟南芥中,破坏丝裂原激活蛋白(MAP)激酶级联触发mkk1/22(SUMM2)介导的自身免疫的核苷酸结合富含亮氨酸重复序列(NLR)抑制。通过RNAi屏幕,我们鉴定了PUB5,一种推定的植物U盒E3连接酶,作为SUMM2介导的自身免疫的关键调节因子。与典型的E3连接酶相比,PUB5稳定CRCK3,一种参与SUMM2激活的钙调蛋白结合受体样细胞质激酶。密切相关的E3连接酶,PUB44的功能与PUB5相反,通过单单双胍化和内化降解CRCK3。此外,CRCK3,在根中高表达,在植物物种中保守,赋予对尖孢镰刀菌的抗性,一种毁灭性的土壤传播的真菌病原体,在拟南芥和棉花中。这些发现证明了E3连接酶对在微调激酶蛋白稳定中的拮抗作用,以调节NLR介导的自身免疫,并强调了自身免疫活化剂在控制植物根部对真菌病原体的免疫中的功能。
    Plant immune homeostasis is achieved through a balanced immune activation and suppression, enabling effective defense while averting autoimmunity. In Arabidopsis, disrupting a mitogen-activated protein (MAP) kinase cascade triggers nucleotide-binding leucine-rich-repeat (NLR) SUPPRESSOR OF mkk1/2 2 (SUMM2)-mediated autoimmunity. Through an RNAi screen, we identify PUB5, a putative plant U-box E3 ligase, as a critical regulator of SUMM2-mediated autoimmunity. In contrast to typical E3 ligases, PUB5 stabilizes CRCK3, a calmodulin-binding receptor-like cytoplasmic kinase involved in SUMM2 activation. A closely related E3 ligase, PUB44, functions oppositely with PUB5 to degrade CRCK3 through monoubiquitylation and internalization. Furthermore, CRCK3, highly expressed in roots and conserved across plant species, confers resistance to Fusarium oxysporum, a devastating soil-borne fungal pathogen, in both Arabidopsis and cotton. These findings demonstrate the antagonistic role of an E3 ligase pair in fine-tuning kinase proteostasis for the regulation of NLR-mediated autoimmunity and highlight the function of autoimmune activators in governing plant root immunity against fungal pathogens.
    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
    F-box蛋白通过泛素化和随后的靶蛋白降解在精子发生的各种细胞过程中发挥重要作用。它们是SKP1-cullin1-F-box蛋白(SCF)E3连接酶复合物的底物识别亚基。F-box蛋白介导的蛋白水解的失调可能导致人类和小鼠的男性不育。新兴的研究揭示了生理功能,病理证据,和雄性生殖细胞发育中F-box蛋白的生化底物,这敦促我们回顾目前对F盒蛋白如何促进精子发生的理解。更多的功能和机制研究将有助于明确F-box蛋白在精子发生中的作用。这将为针对男性不育的F-box蛋白靶向诊断和治疗的逻辑设计铺平道路,因为许多F-box蛋白的生精作用仍然难以捉摸。
    F-box proteins play essential roles in various cellular processes of spermatogenesis by means of ubiquitylation and subsequent target protein degradation. They are the substrate-recognition subunits of SKP1-cullin 1-F-box protein (SCF) E3 ligase complexes. Dysregulation of F‑box protein‑mediated proteolysis could lead to male infertility in humans and mice. The emerging studies revealed the physiological function, pathological evidence, and biochemical substrates of F-box proteins in the development of male germ cells, which urging us to review the current understanding of how F‑box proteins contribute to spermatogenesis. More functional and mechanistic study will be helpful to define the roles of F-box protein in spermatogenesis, which will pave the way for the logical design of F-box protein-targeted diagnosis and therapies for male infertility, as the spermatogenic role of many F-box proteins remains elusive.
    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:肝细胞癌(HCC)是一种常见的恶性肿瘤,铁凋亡是一种由过度脂质过氧化引起的细胞死亡的新形式。近年来,在癌症治疗中,泛素化修饰系统在肿瘤发生和转移中起着至关重要的作用。越来越多的证据表明泛素调节参与该过程的铁凋亡相关底物。然而,利用泛素化修饰调节铁凋亡治疗HCC的确切机制尚不清楚.
    方法:在本研究中,我们使用免疫组织化学和蛋白质印迹技术检测了TRIM33在HCC中的表达。通过体外和体内实验验证了TRIM33的功能作用。为了评估铁中毒的水平,线粒体超氧化物水平,MDA水平,Fe2+水平,并评估细胞活力。通过免疫沉淀筛选和确认TRIM33的下游底物,免疫荧光染色,和泛素化修饰实验。
    结果:我们的发现表明,TRIM33在体外和体内抑制HCC细胞的生长和转移,同时促进其对铁凋亡的敏感性。机械地讲,TRIM33通过E3连接酶依赖性降解TFRC(该过程的已知抑制剂)诱导细胞铁凋亡,从而阐明了TFRC经过TRIM33修饰的特定类型和位点。
    结论:总之,我们的研究揭示了TRIM33在HCC治疗中的重要作用,同时为其功能提供了机制支持.此外,还强调了泛素化修饰导致TFRC降解的重要性,这一见解可能对未来的靶向治疗有价值。
    BACKGROUND: Hepatocellular carcinoma (HCC) is a common malignancy, and ferroptosis is a novel form of cell death driven by excessive lipid peroxidation. In recent years, ferroptosis has been widely utilized in cancer treatment, and the ubiquitination modification system has been recognized to play a crucial role in tumorigenesis and metastasis. Increasing evidence suggests that ubiquitin regulates ferroptosis-related substrates involved in this process. However, the precise mechanism of utilizing ubiquitination modification to regulate ferroptosis for HCC treatment remains unclear.
    METHODS: In this study, we detected the expression of TRIM33 in HCC using immunohistochemistry and western blotting techniques. The functional role of TRIM33 was verified through both in vitro and in vivo experiments. To evaluate the level of ferroptosis, mitochondrial superoxide levels, MDA levels, Fe2+ levels, and cell viability were assessed. Downstream substrates of TRIM33 were screened and confirmed via immunoprecipitation, immunofluorescence staining, and ubiquitination modification experiments.
    RESULTS: Our findings demonstrate that TRIM33 inhibits the growth and metastasis of HCC cells both in vitro and in vivo while promoting their susceptibility to ferroptosis. Mechanistically speaking, TRIM33 induces cellular ferroptosis through E3 ligase-dependent degradation of TFRC-a known inhibitor of this process-thus elucidating the specific type and site at which TFRC undergoes modification by TRIM33.
    CONCLUSIONS: In summary, our study reveals an important role for TRIM33 in HCC treatment while providing mechanistic support for its function. Additionally highlighted is the significance of ubiquitination modification leading to TFRC degradation-an insight that may prove valuable for future targeted therapies.
    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
    CBLC(CBL原癌基因C)是E3泛素蛋白连接酶,在癌症中起关键作用。然而,CBLC在结直肠癌(CRC)中的作用和机制尚未完全阐明。本研究的目的是探讨CBLC在CRC中的功能及其潜在的分子机制。高CBLC水平在CRC患者的肿瘤组织中得到证实,其表达与TNM分期呈正相关。接下来,我们使用功能的获得或丧失探讨了CBLC在CRC中的作用.对于生物功能分析,CCK-8细胞增殖,菌落形成,流式细胞术,划痕,和transwell分析共同表明CBLC过表达促进细胞增殖,细胞周期进程,移民和入侵。正如观察到的,CBLC敲除表现出完全相反的效果,导致体外致瘤性受损。异种移植研究表明,CBLC过表达加速肿瘤生长并促进肿瘤向肺转移,同时证实了CBLC敲低对CRC细胞的致瘤性和转移能力的抑制作用。此外,探讨了CBLC在CRC中的分子机制。CBLC诱导激活ERK信号通路,进一步导致其促肿瘤作用。值得注意的是,CBLC通过其泛素连接酶活性降低ABI1(Abelson相互作用蛋白1,候选肿瘤抑制因子)蛋白水平,而ABI1上调消除了CBLC对CRC肿瘤发生的影响。一起来看,这些结果表明,CBLC通过触发ABI1的泛素化和降解以及激活ERK信号通路,在CRC中充当肿瘤启动子.CBLC可能是CRC的潜在新靶标。
    CBLC (CBL proto-oncogene C) is an E3 ubiquitin protein ligase that plays a key role in cancers. However, the function and mechanism of CBLC in colorectal cancer (CRC) has not been fully elucidated. The aim of this study was to investigate the function of CBLC in CRC and its underlying molecular mechanism. High CBLC levels were certified in tumor tissues of CRC patients, and its expression was positively associated with TNM stage. Next, we explored the role of CBLC in CRC using gain or loss of function. For biological function analysis, CCK-8 cell proliferation, colony formation, flow cytometry, scratch, and transwell assays collectively suggested that CBLC overexpression promoted cell proliferation, cell cycle progression, migration and invasion. As observed, CBLC knockdown exhibited exactly opposite effects, resulting in impaired tumorigenicity in vitro. Xenograft studies displayed that CBLC overexpression accelerated tumor growth and promoted tumor metastasis to the lung, while the inhibitory effects of CBLC knockdown on tumorigenicity and metastasis ability of CRC cells was also confirmed. Furthermore, the molecular mechanism of CBLC in CRC was explored. CBLC induced the activation of ERK signaling pathway, further leading to its pro-tumor role. Notably, CBLC decreased ABI1 (Abelson interactor protein-1, a candidate tumor suppressor) protein levels through its ubiquitin ligase activity, while ABI1 upregulation abolished the effects of CBLC on the tumorigenesis of CRC. Taken together, these results demonstrate that CBLC acts as a tumor promoter in CRC through triggering the ubiquitination and degradation of ABI1 and activating the ERK signaling pathway. CBLC may be a potential novel target for CRC.
    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
    肺癌是一种常见的恶性肿瘤,其特征是铁性凋亡,由过度的脂质过氧化引起的铁依赖性细胞死亡形式。泛素化系统的破坏在肿瘤的发展和扩散中起着至关重要的作用。近年来,人们对利用铁性凋亡治疗肺癌越来越感兴趣;然而,泛素化调节铁凋亡的确切机制尚不清楚.我们使用数据库分析了肺癌组织与正常组织相比的STUB1表达模式,并进行了免疫组织化学。通过体外和体内功能获得和功能丧失实验研究了STUB1的功能作用。丙二醛水平,Fe2+含量,和细胞活力测定用于评估铁凋亡状态。通过筛选鉴定了STUB1的下游靶标,并使用免疫沉淀和泛素化测定法进行了验证。我们的发现表明,STUB1在肺癌细胞中下调,并在体外和体内充当其生长和转移的抑制剂,同时促进铁凋亡。机械上,STUB1通过E3连接酶依赖性降解铁凋亡抑制因子HSPB1诱导铁凋亡。此外,我们的研究阐明了STUB1介导的HSPB1修饰的具体类型和位点。这项研究确立了STUB1作为影响肺癌细胞增殖以及与其相关的上皮-间质转化过程的肿瘤抑制剂。重要的是,我们的工作强调了STUB1在泛素化介导的HSPB1降解中的作用,为肺癌的潜在治疗提供了见解.
    Lung cancer is a common malignancy characterized by ferroptosis, an iron-dependent form of cell death caused by excessive lipid peroxidation. The disruption of the ubiquitination system plays a crucial role in tumor development and spread. In recent years, there has been increasing interest in utilizing ferroptosis for lung cancer treatment; however, the precise mechanism of how ubiquitination modulates ferroptosis remains unclear. We used databases to analyze STUB1 expression patterns in lung cancer tissues compared to normal tissues and performed immunohistochemistry. The functional role of STUB1 was investigated through gain-of-function and loss-of-function experiments both in vitro and in vivo. Malondialdehyde levels, Fe2+ content, and cell viability assays were employed to evaluate ferroptosis status. Downstream targets of STUB1 were identified through screening and validated using immunoprecipitation and ubiquitination assays. Our findings demonstrate that STUB1 is downregulated in lung cancer cells and functions as an inhibitor of their growth and metastasis both in vitro and in vivo while promoting ferroptosis. Mechanistically, STUB1 induces ferroptosis through E3 ligase-dependent degradation of the ferroptosis suppressor HSPB1. Furthermore, our study elucidated the specific types and sites of modification on HSPB1 mediated by STUB1. This research establishes STUB1 as a tumor suppressor influencing proliferation of lung cancer cells as well as the epithelial-mesenchymal transition process associated with it. Importantly, our work highlights the role of STUB1 in ubiquitination-mediated degradation of HSPB1, providing insights for potential treatments for lung cancer.
    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
    动脉粥样硬化是一种慢性动脉疾病,可引起各种类型的心血管功能障碍。血管平滑肌细胞(VSMC),动脉粥样硬化斑块的主要成分,在动脉粥样硬化形成过程中从收缩转变为合成表型。泛素化在动脉粥样硬化中调节VSMC表型至关重要,它可以被去泛素酶反向调节。然而,去泛素酶对动脉粥样硬化的具体作用尚未完全阐明.在这项研究中,在人主动脉平滑肌细胞中进行RNAi筛选以探索OTU家族去泛素酶的作用。这表明沉默OTUB1抑制PDGF-BB刺激的VSMC表型转换。使用Apoe-/-小鼠的进一步体内研究表明,VSMC中OTUB1的敲除减轻了晚期的动脉粥样硬化斑块负担,并导致稳定的斑块表型。此外,体外沉默OTUB1可以抑制PDGF-BB刺激下的VSMC增殖和迁移。无偏RNA测序数据表明敲低OTUB1影响VSMC分化,附着力,和扩散。泛素化蛋白的质谱证实与细胞生长和迁移相关的蛋白被差异泛素化。机械上,我们发现OTUB1以其催化三联体识别PDGFRβ的K707残基泛素化,从而减少PDGFRβ的K48连接的泛素化。抑制VSMCs中的OTUB1可通过泛素-蛋白酶体途径促进PDGFRβ降解,因此,它有利于防止VSMCs表型转换。这些发现表明,敲低OTUB1改善了VSMCs的表型转换和动脉粥样硬化进展,表明OTUB1可能是未来有价值的翻译治疗靶点。
    Atherosclerosis is a chronic artery disease that causes various types of cardiovascular dysfunction. Vascular smooth muscle cells (VSMCs), the main components of atherosclerotic plaque, switch from contractile to synthetic phenotypes during atherogenesis. Ubiquitylation is crucial in regulating VSMC phenotypes in atherosclerosis, and it can be reversely regulated by deubiquitinases. However, the specific effects of deubiquitinases on atherosclerosis have not been thoroughly elucidated. In this study, RNAi screening in human aortic smooth muscle cells was performed to explore the effects of OTU family deubiquitinases, which revealed that silencing OTUB1 inhibited PDGF-BB-stimulated VSMC phenotype switch. Further in vivo studies using Apoe-/- mice revealed that knockdown of OTUB1 in VSMCs alleviated atherosclerosis plaque burden in the advanced stage and led to a stable plaque phenotype. Moreover, VSMC proliferation and migration upon PDGF-BB stimulation could be inhibited by silencing OTUB1 in vitro. Unbiased RNA-sequencing data indicated that knocking down OTUB1 influenced VSMC differentiation, adhesion, and proliferation. Mass spectrometry of ubiquitinated protein confirmed that proteins related to cell growth and migration were differentially ubiquitylated. Mechanistically, we found that OTUB1 recognized the K707 residue ubiquitylation of PDGFRβ with its catalytic triad, thereby reducing the K48-linked ubiquitylation of PDGFRβ. Inhibiting OTUB1 in VSMCs could promote PDGFRβ degradation via the ubiquitin-proteasome pathway, so it was beneficial in preventing VSMCs\' phenotype switch. These findings revealed that knocking down OTUB1 ameliorated VSMCs\' phenotype switch and atherosclerosis progression, indicating that OTUB1 could be a valuable translational therapeutic target in the future.
    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
    FBXO43是F盒蛋白的FBXO亚家族的成员,已知是减数分裂期间的监管中心。大量数据显示FBXO43在许多人类癌症中过表达。然而,FBXO43是否以及如何影响细胞周期进程和癌细胞的生长仍然难以捉摸。在这项研究中,我们提供了第一个证据,显示FBXO43在细胞周期进程和癌细胞生长中的关键作用。具体来说,FBXO43作为阳性细胞周期调节因子,在各种类型的人类癌症中具有致癌活性,包括非小细胞肺癌,肝细胞癌和肉瘤。机械上,FBXO43与AKT1诱导的磷酸化SKP2相互作用,导致SKP2自身泛素化和随后的蛋白酶体降解减少。一起来看,我们的研究表明,FBXO43通过稳定SKP2促进细胞周期进程,并且FBXO43可作为潜在的抗癌靶点.
    FBXO43 is a member of the FBXO subfamily of F-box proteins, known to be a regulatory hub during meiosis. A body of data showed that FBXO43 is overexpressed in a number of human cancers. However, whether and how FBXO43 affects cell cycle progression and growth of cancer cells remain elusive. In this study, we provide first piece of evidence, showing a pivotal role of FBXO43 in cell cycle progression and growth of cancer cells. Specifically, FBXO43 acts as a positive cell cycle regulator with an oncogenic activity in variety types of human cancer, including non-small cell lung cancer, hepatocellular carcinoma and sarcoma. Mechanistically, FBXO43 interacts with phosphorylated SKP2 induced by AKT1, leading to reduced SKP2 auto-ubiquitylation and subsequent proteasome degradation. Taken together, our study demonstrates that FBXO43 promotes cell cycle progression by stabilizing SKP2, and FBXO43 could serve as a potential anti-cancer target.
    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
    泛素-蛋白酶体系统(UPS)是介导真核细胞中特定蛋白质周转的主要机制。通过泛素化不必要的,损坏,或有害的蛋白质,并推动它们的降解,UPS涉及许多重要的细胞过程。几种基于UPS的新技术,包括分子胶降解剂和PROTACs(蛋白水解靶向嵌合体),以促进蛋白质降解,和DUBTACs(去泛素酶靶向嵌合体)增加蛋白质的稳定性,已经开发了。通过特异性诱导不同的泛素连接酶和不相关的靶向蛋白质之间的相互作用,分子胶降解剂和PROTACs通过泛素-蛋白酶体系统降解目标蛋白;相反,通过诱导目标蛋白质接近去泛素酶,DUBTAC用于清除可降解的聚泛素链以稳定目标蛋白质。在这次审查中,我们总结了分子胶降解剂的最新研究进展,方案,和DUBTAC及其应用。我们讨论免疫调节药物(IMiDs),磺胺类药物,CDK靶向分子胶降解剂,以及PROTACs的新发展。我们还介绍了DUBTAC的原理及其应用。最后,我们提出了与靶向蛋白质稳态相关的这三种技术的一些未来方向。
    The ubiquitin (Ub)-proteasome system (UPS) is the major machinery mediating specific protein turnover in eukaryotic cells. By ubiquitylating unwanted, damaged, or harmful proteins and driving their degradation, UPS is involved in many important cellular processes. Several new UPS-based technologies, including molecular glue degraders and PROTACs (proteolysis-targeting chimeras) to promote protein degradation, and DUBTACs (deubiquitinase-targeting chimeras) to increase protein stability, have been developed. By specifically inducing the interactions between different Ub ligases and targeted proteins that are not otherwise related, molecular glue degraders and PROTACs degrade targeted proteins via the UPS; in contrast, by inducing the proximity of targeted proteins to deubiquitinases, DUBTACs are created to clear degradable poly-Ub chains to stabilize targeted proteins. In this review, we summarize the recent research progress in molecular glue degraders, PROTACs, and DUBTACs and their applications. We discuss immunomodulatory drugs, sulfonamides, cyclin-dependent kinase-targeting molecular glue degraders, and new development of PROTACs. We also introduce the principle of DUBTAC and its applications. Finally, we propose a few future directions of these three technologies related to targeted protein homeostasis.
    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
    UBE2F,aneddylationE2,在与neddylE3RBX2/SAG偶联后,neddylateCUL5活化cul5以激活cullin-RING连接酶-5。UBE2F是否以及如何控制胰腺肿瘤发生以前是未知的。这里,我们表明UBE2F对于具有KRAS突变的人胰腺癌细胞的生长至关重要。在小鼠KrasG12D胰腺导管腺癌(PDAC)模型中,Ube2f缺失抑制cerulein诱导的胰腺炎,腺泡-导管上皮化生(ADM)和胰腺上皮内瘤变的进展。机械上,Ube2f缺失通过阻断CRL5Asb11E3连接酶的底物Diras2的泛素化使Mapk-c-Myc信号失活。生物学,DIRAS2抑制携带突变KRAS的人胰腺癌细胞的生长和存活,Diras2缺失在很大程度上挽救了Ube2f缺失诱导的表型。总的来说,Ube2f或Diras2在小鼠KrasG12DPDAC模型中发挥促肿瘤或抑瘤作用,分别。UBE2F-CRL5ASB11轴可以作为胰腺癌的有效靶标,而UBE2F或DIRAS2水平可作为PDAC患者的预后生物标志物.
    UBE2F, a neddylation E2, neddylates CUL5 to activate cullin-RING ligase-5, upon coupling with neddylation E3 RBX2/SAG. Whether and how UBE2F controls pancreatic tumorigenesis is previously unknown. Here, we showed that UBE2F is essential for the growth of human pancreatic cancer cells with KRAS mutation. In the mouse KrasG12D pancreatic ductal adenocarcinoma (PDAC) model, Ube2f deletion suppresses cerulein-induced pancreatitis, and progression of acinar-to-ductal metaplasia (ADM) and pancreatic intraepithelial neoplasia. Mechanistically, Ube2f deletion inactivates the Mapk-c-Myc signals via blocking ubiquitylation of Diras2, a substrate of CRL5Asb11 E3 ligase. Biologically, DIRAS2 suppresses growth and survival of human pancreatic cancer cells harboring mutant KRAS, and Diras2 deletion largely rescues the phenotypes induced by Ube2f deletion. Collectively, Ube2f or Diras2 plays a tumor-promoting or tumor-suppressive role in the mouse KrasG12D PDAC model, respectively. The UBE2F-CRL5ASB11 axis could serve as a valid target for pancreatic cancer, whereas the levels of UBE2F or DIRAS2 may serve as prognostic biomarkers for PDAC patients.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号