Deubiquitinating Enzymes

去泛素化酶
  • 文章类型: Journal Article
    多发性骨髓瘤(MM)是成人第二常见的血液肿瘤。免疫调节药物(IMiDs),如沙利度胺和来那度胺(Len),是治疗多发性骨髓瘤的有效药物。Len可以招募IKZF1和IKZF3到cereblon(CRBN),cullin4-RINGE3连接酶(CRL4)的底物受体,促进它们的泛素化和降解,并最终抑制骨髓瘤细胞的增殖。然而,MM患者随着时间的推移会对IMiDs产生耐药性,导致疾病复发和恶化。为了探索可能增强IMID对MM敏感性的可能方法,在这项研究中,我们使用邻近标记技术TurboID和定量蛋白质组学将Lys-63特异性去泛素酶BRCC36鉴定为CRBN相互作用蛋白.生化实验表明,BRISC复合物中的BRCC36通过特异性裂解CRBN上的K63连接的聚泛素链保护CRBN免受溶酶体降解。进一步的研究发现,与BRISC复合物亚基SHMT2结合的小分子化合物SHIN1可以通过升高BRCC36来上调CRBN。SHIN1和Len的组合可以进一步增加MM细胞对IMiDs的敏感性。因此,本研究为探索SHIN1和Len联合治疗MMs的可能策略提供了依据.
    Multiple myeloma (MM) is the second most common hematological tumor in adults. Immunomodulatory drugs (IMiDs), such as thalidomide and lenalidomide (Len), are effective drugs for the treatment of multiple myeloma. Len can recruit IKZF1 and IKZF3 to cereblon (CRBN), a substrate receptor of the cullin 4-RING E3 ligase (CRL4), promote their ubiquitination and degradation, and finally inhibit the proliferation of myeloma cells. However, MM patients develop resistance to IMiDs over time, leading to disease recurrence and deterioration. To explore the possible approaches that may enhance the sensitivity of IMiDs to MM, in this study, we used the proximity labeling technique TurboID and quantitative proteomics to identify Lys-63-specific deubiquitinase BRCC36 as a CRBN-interacting protein. Biochemical experiments demonstrated that BRCC36 in the BRISC complex protects CRBN from lysosomal degradation by specifically cleaving the K63-linked polyubiquitin chain on CRBN. Further studies found that a small-molecule compound SHIN1, which binds to BRISC complex subunit SHMT2, can upregulate CRBN by elevating BRCC36. The combination of SHIN1 and Len can further increase the sensitivity of MM cells to IMiDs. Therefore, this study provides the basis for the exploration of a possible strategy for the SHIN1 and Len combination treatment for MM.
    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
    泛素化,一种普遍且高度动态的可逆翻译后修饰,受到去泛素化酶(DUBs)超家族的严格调控。其中,含OTU结构域的泛素醛结合蛋白1(OTUB1)是OTU去泛素家族的关键成员,在各种癌症中作为肿瘤调节剂发挥关键作用。然而,其在BLCA(BLCA)中的具体参与及其临床意义仍然不明确。本研究旨在阐明OTUB1在BLCA中的生物学功能及其对临床预后的影响。我们的调查显示,BLCA中OTUB1的表达增加,与不利的临床结果相关。通过体内和体外实验,我们证明,增加OTUB1水平促进BLCA肿瘤发生和进展,同时赋予顺铂治疗耐药性。值得注意的是,我们建立了一个涉及OTUB1、β-catenin、坏死,BLCA,描述它们之间的监管相互作用。机械上,我们发现OTUB1通过去泛素化和稳定β-catenin发挥其影响,导致其核易位。随后,核β-catenin增强c-myc和cyclinD1的转录活性,同时抑制RIPK3和MLKL的表达,从而促进BLCA进展和顺铂耐药。重要的是,我们的临床数据提示OTUB1/β-catenin/RIPK3/MLKL轴有望成为BLCA的潜在生物标志物.
    Ubiquitination, a prevalent and highly dynamic reversible post-translational modification, is tightly regulated by the deubiquitinating enzymes (DUBs) superfamily. Among them, OTU Domain-Containing Ubiquitin Aldehyde-Binding Protein 1 (OTUB1) stands out as a critical member of the OTU deubiquitinating family, playing a pivotal role as a tumor regulator across various cancers. However, its specific involvement in BLCA (BLCA) and its clinical significance have remained ambiguous. This study aimed to elucidate the biofunctions of OTUB1 in BLCA and its implications for clinical prognosis. Our investigation revealed heightened OTUB1 expression in BLCA, correlating with unfavorable clinical outcomes. Through in vivo and in vitro experiments, we demonstrated that increased OTUB1 levels promote BLCA tumorigenesis and progression, along with conferring resistance to cisplatin treatment. Notably, we established a comprehensive network involving OTUB1, β-catenin, necroptosis, and BLCA, delineating their regulatory interplay. Mechanistically, we uncovered that OTUB1 exerts its influence by deubiquitinating and stabilizing β-catenin, leading to its nuclear translocation. Subsequently, nuclear β-catenin enhances the transcriptional activity of c-myc and cyclin D1 while suppressing the expression of RIPK3 and MLKL, thereby fostering BLCA progression and cisplatin resistance. Importantly, our clinical data suggest that the OTUB1/β-catenin/RIPK3/MLKL axis holds promise as a potential biomarker for BLCA.
    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
    去泛素酶(DUB)对于维持蛋白质稳态和将蛋白质组装成功能复合物至关重要。尽管人们对DUBs的生物学功能越来越感兴趣,DUB在调节肠干细胞(ISC)和肠道稳态中的作用仍然未知。这里,我们通过诱导敲低成人中肠ISC和肠母细胞(EB)中DUBs的表达进行体内RNAi筛选,以鉴定果蝇中肠道稳态的DUB调节因子。我们筛选了43个DUB,并确定了ISC稳态所需的8个DUB。usp1,CG7857,usp5,rpn8,usp10和csn5的敲除减少了ISC/EB的数量,而CG4968和usp8的敲低增加了ISC/EB的数量。此外,ISC/EBs中usp1,CG4968,CG7857或rpn8的敲低破坏了肠屏障的完整性并缩短了寿命,表明这些DUB对维持肠道稳态的要求。此外,我们提供证据表明Usp1通过调节Notch信号活性介导ISC谱系分化。我们的研究表明,第一次,维持果蝇肠道稳态所需的去泛素酶,并为DUB和肠道稳态之间的功能联系提供新的见解。
    Deubiquitinases (DUBs) are essential for the maintenance of protein homeostasis and assembly of proteins into functional complexes. Despite growing interest in DUBs biological functions, the roles of DUBs in regulating intestinal stem cells (ISCs) and gut homeostasis remain largely unknown. Here, we perform an in vivo RNAi screen through induced knock-down of DUBs expression in adult midgut ISCs and enteroblasts (EBs) to identify DUB regulators of intestinal homeostasis in Drosophila. We screen 43 DUBs and identify 8 DUBs that are required for ISCs homeostasis. Knocking-down of usp1, CG7857, usp5, rpn8, usp10 and csn5 decreases the number of ISCs/EBs, while knocking-down of CG4968 and usp8 increases the number of ISCs/EBs. Moreover, knock-down of usp1, CG4968, CG7857, or rpn8 in ISCs/EBs disrupts the intestinal barrier integrity and shortens the lifespan, indicating the requirement of these DUBs for the maintenance of gut homeostasis. Furthermore, we provide evidences that USP1 mediates ISC lineage differentiation via modulating the Notch signaling activity. Our study identifies, for the first time, the deubiquitinases required for the maintenance of intestinal homeostasis in Drosophila, and provide new insights into the functional links between the DUBs and intestinal homeostasis.
    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)通过调节各种生物过程来维持细胞内蛋白质稳态和细胞功能。泛素化,一种常见的翻译后修饰,在蛋白质降解的调节中起着至关重要的作用,信号转导,以及其他生理和病理过程,并参与各种癌症的发病机理,包括骨肉瘤.骨肉瘤,最常见的原发性恶性骨肿瘤,具有高转移潜能和不良预后的特点。这是一种难治性骨病,主要治疗方式为手术联合化疗。越来越多的证据表明UPS异常与骨肉瘤的进展密切相关。由于泛素化系统的复杂性和多效性,泛素化过程中的每一步都可以被药物靶向。近年来,针对泛素系统的抑制剂的研究和开发逐渐增加,显示出巨大的临床应用潜力。本文就泛素化系统在骨肉瘤发生发展和治疗中的作用作一综述。以及研究进展,希望通过靶向泛素化系统中的有效分子来改善骨肉瘤患者的治疗效果和预后。
    The ubiquitin-proteasome system (UPS) maintains intracellular protein homeostasis and cellular function by regulating various biological processes. Ubiquitination, a common post-translational modification, plays a crucial role in the regulation of protein degradation, signal transduction, and other physiological and pathological processes, and is involved in the pathogenesis of various cancers, including osteosarcoma. Osteosarcoma, the most common primary malignant bone tumor, is characterized by high metastatic potential and poor prognosis. It is a refractory bone disease, and the main treatment modalities are surgery combined with chemotherapy. Increasing evidence suggests a close association between UPS abnormalities and the progression of osteosarcoma. Due to the complexity and pleiotropy of the ubiquitination system, each step in the ubiquitination process can be targeted by drugs. In recent years, research and development of inhibitors targeting the ubiquitin system have increased gradually, showing great potential for clinical application. This article reviews the role of the ubiquitination system in the development and treatment of osteosarcoma, as well as research progress, with the hope of improving the therapeutic effects and prognosis of osteosarcoma patients by targeting effective molecules in the ubiquitination system.
    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
    泛素化,蛋白质的关键翻译后修饰,在调节蛋白质的稳定性中起着至关重要的作用。泛素化酶和去泛素化酶的失调是各种癌症的共同特征,强调必须研究泛素连接酶和去泛素化酶(DUB),以了解致癌过程和治疗干预措施的发展。在这次审查中,我们讨论了泛素-蛋白酶体系统(UPS)在癌症的所有标志和药物发现的进展中的贡献。我们深入研究了UPS在肿瘤学中的多种功能,包括它对多种癌症相关途径的调节,它在代谢重编程中的作用,它参与肿瘤免疫反应,它在表型可塑性和多态微生物组中的功能,和其他基本的细胞功能。此外,我们全面概述了利用UPS的新型抗癌策略,包括蛋白水解靶向嵌合体(PROTACs)和分子胶的开发和应用。
    Ubiquitination, a pivotal posttranslational modification of proteins, plays a fundamental role in regulating protein stability. The dysregulation of ubiquitinating and deubiquitinating enzymes is a common feature in various cancers, underscoring the imperative to investigate ubiquitin ligases and deubiquitinases (DUBs) for insights into oncogenic processes and the development of therapeutic interventions. In this review, we discuss the contributions of the ubiquitin-proteasome system (UPS) in all hallmarks of cancer and progress in drug discovery. We delve into the multiple functions of the UPS in oncology, including its regulation of multiple cancer-associated pathways, its role in metabolic reprogramming, its engagement with tumor immune responses, its function in phenotypic plasticity and polymorphic microbiomes, and other essential cellular functions. Furthermore, we provide a comprehensive overview of novel anticancer strategies that leverage the UPS, including the development and application of proteolysis targeting chimeras (PROTACs) and molecular glues.
    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
    泛素化途径在蛋白质稳态中具有至关重要的作用,信号和先天免疫1-3。在这些途径中,E1、E2和E3蛋白的酶促级联将泛素或泛素样蛋白(Ubl)与靶蛋白赖氨酸残基缀合4。细菌编码涉及硫代谢5,6的E1和Ubl蛋白的古代亲属,但这些蛋白不介导Ubl-靶结合,剩下的问题是细菌是否可以进行泛素化样蛋白缀合。在这里,我们证明了与噬菌体防御岛相关的细菌操纵子编码完整的泛素化途径。细菌E1-E2-Ubl复合物的两种结构揭示了与经典真核生物泛素化机制的惊人结构相似之处。细菌E1具有氨基末端无活性的腺苷酸化结构域和羧基末端活性的腺苷酸化结构域,其具有含有催化半胱氨酸(CYS结构域)的移动α-螺旋插入。一种结构揭示了细菌UblC末端定位用于腺苷酸化的预反应状态,和第二结构模拟E1-E2硫代酯转移状态,其中E1CYS结构域与结合的E2相邻。我们表明,在同一途径中的去泛素酶预处理细菌Ubl,暴露其C端甘氨酸进行腺苷酸化。最后,我们显示,细菌E1和E2协作以将Ubl缀合至靶蛋白赖氨酸残基。一起,这些数据表明,细菌拥有真正的泛素化系统,与经典真核生物泛素化途径具有很强的机械和结构相似性,这表明这些途径首先出现在细菌中.
    Ubiquitination pathways have crucial roles in protein homeostasis, signalling and innate immunity1-3. In these pathways, an enzymatic cascade of E1, E2 and E3 proteins conjugates ubiquitin or a ubiquitin-like protein (Ubl) to target-protein lysine residues4. Bacteria encode ancient relatives of E1 and Ubl proteins involved in sulfur metabolism5,6, but these proteins do not mediate Ubl-target conjugation, leaving open the question of whether bacteria can perform ubiquitination-like protein conjugation. Here we demonstrate that a bacterial operon associated with phage defence islands encodes a complete ubiquitination pathway. Two structures of a bacterial E1-E2-Ubl complex reveal striking architectural parallels with canonical eukaryotic ubiquitination machinery. The bacterial E1 possesses an amino-terminal inactive adenylation domain and a carboxy-terminal active adenylation domain with a mobile α-helical insertion containing the catalytic cysteine (CYS domain). One structure reveals a pre-reaction state with the bacterial Ubl C terminus positioned for adenylation, and a second structure mimics an E1-to-E2 transthioesterification state with the E1 CYS domain adjacent to the bound E2. We show that a deubiquitinase in the same pathway preprocesses the bacterial Ubl, exposing its C-terminal glycine for adenylation. Finally, we show that the bacterial E1 and E2 collaborate to conjugate Ubl to target-protein lysine residues. Together, these data reveal that bacteria possess bona fide ubiquitination systems with strong mechanistic and architectural parallels to canonical eukaryotic ubiquitination pathways, suggesting that these pathways arose first in bacteria.
    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
    人类中的几种免疫途径将泛素样蛋白与病毒和宿主分子结合,作为抗病毒防御的手段1-5。在这里,我们研究了细菌中的抗噬菌体防御系统,包含泛素样蛋白,泛素缀合酶E1和E2,以及去泛素酶。我们证明在噬菌体感染期间,该系统将泛素样蛋白与噬菌体中央尾纤维特异性结合,尾部末端的蛋白质,对于尾部组装以及识别目标宿主受体至关重要。感染后,编码这个防御系统的细胞释放出部分组装的混合物,无尾噬菌体颗粒和完全组装的噬菌体,其中中央尾纤维被共价连接的泛素样蛋白阻塞。这些噬菌体显示严重受损的感染性,解释防御系统如何保护细菌种群免受噬菌体感染的传播。我们的发现表明,泛素样蛋白的结合是整个生命树保守的抗病毒策略。
    Several immune pathways in humans conjugate ubiquitin-like proteins to virus and host molecules as a means of antiviral defence1-5. Here we studied an antiphage defence system in bacteria, comprising a ubiquitin-like protein, ubiquitin-conjugating enzymes E1 and E2, and a deubiquitinase. We show that during phage infection, this system specifically conjugates the ubiquitin-like protein to the phage central tail fibre, a protein at the tip of the tail that is essential for tail assembly as well as for recognition of the target host receptor. Following infection, cells encoding this defence system release a mixture of partially assembled, tailless phage particles and fully assembled phages in which the central tail fibre is obstructed by the covalently attached ubiquitin-like protein. These phages show severely impaired infectivity, explaining how the defence system protects the bacterial population from the spread of phage infection. Our findings demonstrate that conjugation of ubiquitin-like proteins is an antiviral strategy conserved across the tree of life.
    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
    肉毒杆菌神经毒素是已知的一些最有效的天然毒素;它们通过抑制突触小泡释放而引起弛缓性麻痹。一些血清型,特别是血清型A和B,会导致持续数月的瘫痪.因为他们的力量和毅力,肉毒杆菌神经毒素现在被用来治疗几种临床疾病,并且有兴趣使用具有新底物特异性的工程毒素扩展其临床应用。设计具有可调持久性的毒素也将是有益的。我们已经研究了小分子蛋白水解靶向嵌合体(PROTACs)的潜在用途,以改变修饰的重组肉毒杆菌神经毒素的持久性。我们还描述了一种与肉毒杆菌中毒治疗具有潜在相关性的补充方法。该第二种方法使用针对肉毒杆菌神经毒素的骆驼重链抗体,其被修饰以结合PROTAC。这些策略为使用两种不同的方法通过选择性靶向其催化轻链进行蛋白酶体降解来微调肉毒杆菌神经毒素的持久性提供了原理证明。
    Botulinum neurotoxins are some of the most potent natural toxins known; they cause flaccid paralysis by inhibiting synaptic vesicle release. Some serotypes, notably serotype A and B, can cause persistent paralysis lasting for several months. Because of their potency and persistence, botulinum neurotoxins are now used to manage several clinical conditions, and there is interest in expanding their clinical applications using engineered toxins with novel substrate specificities. It will also be beneficial to engineer toxins with tunable persistence. We have investigated the potential use of small-molecule proteolysis-targeting chimeras (PROTACs) to vary the persistence of modified recombinant botulinum neurotoxins. We also describe a complementary approach that has potential relevance for botulism treatment. This second approach uses a camelid heavy chain antibody directed against botulinum neurotoxin that is modified to bind the PROTAC. These strategies provide proof of principle for the use of two different approaches to fine tune the persistence of botulinum neurotoxins by selectively targeting their catalytic light chains for proteasomal degradation.
    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
    作为人体最大的器官,骨骼肌是呼吸支持所必需的,运动启动,和维持体内平衡。已经表明,程序性细胞死亡(PCD)其中包括自噬,凋亡,和坏死,对骨骼肌的发育至关重要。关于骨骼肌,一种称为铁中毒的PCD的新形式仍然知之甚少。在这项研究中,我们观察到,铁凋亡的激活显着阻碍了C2C12成肌细胞分化为肌管,同时抑制了关键的去泛素化酶OTUB1的表达。使用OTUB1沉默的C2C12小鼠成肌细胞来研究OTUB1在铁凋亡中的功能。结果表明,体外OTUB1敲低可显著增加C2C12的铁细胞凋亡,抑制肌生成。有趣的是,由OTUB1敲除引起的铁凋亡的诱导伴随着自噬的激活。此外,OTUB1与P62蛋白相互作用,并通过去泛素化来稳定其表达,从而抑制自噬依赖性铁凋亡并促进肌生成。所有这些发现都证明了OTUB1在控制铁凋亡中的关键作用,我们建议关注OTUB1-P62轴可能是治疗和预防涉及骨骼肌疾病的有用策略。
    As the largest organ in the human body, skeletal muscle is essential for breathing support, movement initiation, and maintenance homeostasis. It has been shown that programmed cell death (PCD), which includes autophagy, apoptosis, and necrosis, is essential for the development of skeletal muscle. A novel form of PCD called ferroptosis is still poorly understood in relation to skeletal muscle. In this study, we observed that the activation of ferroptosis significantly impeded the differentiation of C2C12 myoblasts into myotubes and concurrently suppressed the expression of OTUB1, a crucial deubiquitinating enzyme. OTUB1-silenced C2C12 mouse myoblasts were used to investigate the function of OTUB1 in ferroptosis. The results show that OTUB1 knockdown in vitro significantly increased C2C12 ferroptosis and inhibited myogenesis. Interestingly, the induction of ferroptosis resulting from OTUB1 knockdown was concomitant with the activation of autophagy. Furthermore, OTUB1 interacted with the P62 protein and stabilized its expression by deubiquitinating it, thereby inhibiting autophagy-dependent ferroptosis and promoting myogenesis. All of these findings demonstrate the critical role that OTUB1 plays in controlling ferroptosis, and we suggest that focusing on the OTUB1-P62 axis may be a useful tactic in the treatment and prevention of disorders involving the skeletal muscle.
    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
    最近的研究表明,足细胞损伤在糖尿病肾病(DKD)的发展中起着至关重要的作用。蛋白质的去泛素化修饰广泛参与疾病的发生和发展。这里,我们探索去泛素化酶的作用和调节机制,OTUD5,在足细胞损伤和DKD中。RNA-seq分析表明OTUD5在HG/PA刺激的足细胞中的表达显著降低。足细胞特异性Otud5敲除会加剧1型和2型糖尿病小鼠的足细胞损伤和DKD。此外,AVV9介导的OTUD5在足细胞中的过表达显示出对DKD的治疗作用。质谱和免疫共沉淀实验揭示了一种炎症调节蛋白,TAK1,作为足细胞中OTUD5的底物。机械上,OTUD5在K158位点通过其活性位点C224去泛素化K63连接的TAK1,随后阻止TAK1的磷酸化并减少足细胞中的下游炎症反应。我们的发现显示了足细胞炎症和损伤中的OTUD5-TAK1轴,并强调了OTUD5作为DKD有希望的治疗靶标的潜力。
    Recent studies have shown the crucial role of podocyte injury in the development of diabetic kidney disease (DKD). Deubiquitinating modification of proteins is widely involved in the occurrence and development of diseases. Here, we explore the role and regulating mechanism of a deubiquitinating enzyme, OTUD5, in podocyte injury and DKD. RNA-seq analysis indicates a significantly decreased expression of OTUD5 in HG/PA-stimulated podocytes. Podocyte-specific Otud5 knockout exacerbates podocyte injury and DKD in both type 1 and type 2 diabetic mice. Furthermore, AVV9-mediated OTUD5 overexpression in podocytes shows a therapeutic effect against DKD. Mass spectrometry and co-immunoprecipitation experiments reveal an inflammation-regulating protein, TAK1, as the substrate of OTUD5 in podocytes. Mechanistically, OTUD5 deubiquitinates K63-linked TAK1 at the K158 site through its active site C224, which subsequently prevents the phosphorylation of TAK1 and reduces downstream inflammatory responses in podocytes. Our findings show an OTUD5-TAK1 axis in podocyte inflammation and injury and highlight the potential of OTUD5 as a promising therapeutic target for DKD.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号