deubiquitination

去泛素化
  • 文章类型: Journal Article
    非酒精性脂肪性肝病(NAFLD),包括其更严重的表现非酒精性脂肪性肝炎(NASH),是全球公共卫生挑战。这里,我们探讨去泛素化酶RPN11在NAFLD和NASH中的作用。肝细胞特异性RPN11敲除小鼠免受饮食诱导的肝脏脂肪变性,胰岛素抵抗,还有脂肪性肝炎.机械上,RPN11去泛素化并稳定METTL3,以增强酰基辅酶A(CoA)合成酶短链家族成员3(ACSS3)的m6A修饰和表达,产生丙酰辅酶A通过组蛋白丙酰化上调脂质代谢基因。RPN11-METTL3-ACSS3-组蛋白丙酸化途径在NAFLD患者的肝脏中被激活。Capzimin改善NAFLD对RPN11的药理抑制作用,NASH,和相关的代谢紊乱的小鼠和降低的脂质含量培养的人肝细胞在2D和3D。这些结果表明,RPN11是NAFLD/NASH的新型调节因子,抑制RPN11具有治疗潜力。
    Nonalcoholic fatty liver disease (NAFLD), including its more severe manifestation nonalcoholic steatohepatitis (NASH), is a global public health challenge. Here, we explore the role of deubiquitinating enzyme RPN11 in NAFLD and NASH. Hepatocyte-specific RPN11 knockout mice are protected from diet-induced liver steatosis, insulin resistance, and steatohepatitis. Mechanistically, RPN11 deubiquitinates and stabilizes METTL3 to enhance the m6A modification and expression of acyl-coenzyme A (CoA) synthetase short-chain family member 3 (ACSS3), which generates propionyl-CoA to upregulate lipid metabolism genes via histone propionylation. The RPN11-METTL3-ACSS3-histone propionylation pathway is activated in the livers of patients with NAFLD. Pharmacological inhibition of RPN11 by Capzimin ameliorated NAFLD, NASH, and related metabolic disorders in mice and reduced lipid contents in human hepatocytes cultured in 2D and 3D. These results demonstrate that RPN11 is a novel regulator of NAFLD/NASH and that suppressing RPN11 has therapeutic potential for the 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
    背景:肺腺癌(LUAD)是全球范围内高度侵袭性和快速致命的恶性肿瘤。胶原蛋白XVII(COL17A1)与各种原瘤过程有关。然而,COL17A1在LUAD进展中的功能和机制仍然难以捉摸。
    方法:通过定量PCR进行COL17A1和泛素特异性蛋白酶22(USP22)mRNA分析,并通过免疫印迹和免疫组织化学检测其蛋白水平。通过确定细胞活力来评估功能影响,扩散,凋亡,入侵,迁移,和体外铁凋亡,以及异种移植物在体内的生长。免疫共沉淀(Co-IP)和IP实验用于检查USP22/COL17A1相互作用和COL17A1去泛素化。使用环己酰亚胺处理来分析COL17A1蛋白稳定性。
    结果:COL17A1和USP22在人LUAD组织和细胞系中上调。功能上,COL17A1敲低作用于抑制LUAD细胞生长,入侵,和迁移以及在体外促进细胞凋亡和铁凋亡。COL17A1敲低可以降低LUAD细胞在体内的致瘤性。机械上,USP22通过增强COL17A1的去泛素化来稳定和上调COL17A1。此外,COL17A1的再表达可以逆转USP22沉默诱导的LUAD细胞的表型改变。
    结论:我们的研究结果表明,USP22稳定的COL17A1在LUAD中具有致癌活性。我们建议USP22和COL17A1将是建立针对LUAD的治疗方法的潜在靶标。
    BACKGROUND: Lung adenocarcinoma (LUAD) is a highly aggressive and rapidly fatal malignancy worldwide. Collagen XVII (COL17A1) has been implicated in various protumorigenic processes. However, the functions and mechanisms of COL17A1 in LUAD progression still remain elusive.
    METHODS: COL17A1 and ubiquitin-specific protease 22 (USP22) mRNA analysis was performed by quantitative PCR, and their protein levels were detected by immunoblotting and immunohistochemistry. The functional influence was evaluated by determining cell viability, proliferation, apoptosis, invasion, migration, and ferroptosis in vitro, as well as xenograft growth in vivo. Co-immunoprecipitation (Co-IP) and IP experiments were used to examine the USP22/COL17A1 interaction and COL17A1 deubiquitination. Cycloheximide treatment was used to analyze COL17A1 protein stability.
    RESULTS: COL17A1 and USP22 were upregulated in human LUAD tissues and cell lines. Functionally, COL17A1 knockdown acted for the suppression of LUAD cell growth, invasion, and migration as well as promotion of cell apoptosis and ferroptosis in vitro. COL17A1 knockdown could diminish the tumorigenicity of LUAD cells in vivo. Mechanistically, USP22 stabilized and upregulated COL17A1 by enhancing the deubiquitination of COL17A1. Additionally, reexpression of COL17A1 could reverse USP22 silencing-induced phenotype changes of LUAD cells in vitro.
    CONCLUSIONS: Our findings demonstrate that USP22-stabilized COL17A1 possesses oncogenic activity in LUAD. We propose that USP22 and COL17A1 would be potential targets for the establishment of therapeutic approaches against LUAD.
    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,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
    转录因子ELONGATedHY5(HY5)是幼苗光形态发生的中心中心。E3泛素(Ub)连接酶成分光形态发生1(COP1)通过泛素化抑制HY5蛋白积累。然而,HY5去泛素化的过程,它拮抗E3连接酶介导的泛素化以维持HY5稳态从未被研究过。这里,我们鉴定了拟南芥去泛素化酶,Ub特异性蛋白酶14(UBP14)与HY5物理相互作用并通过去泛素化增强其蛋白质稳定性。缺乏UBP14功能的da3-1突变体表现出长的下胚轴表型,UBP14缺乏导致HY5在黑暗至光照期间无法快速积累。此外,UBP14优选稳定HY5的非磷酸化形式,其更容易与下游靶基因结合。HY5促进UBP14的表达和蛋白质积累,以促进光形态发生。因此,我们的发现建立了UBP14通过去泛素化来稳定HY5蛋白以促进拟南芥中的光形态发生的机制。
    Transcription factor ELONGATED HYPOCOTYL5 (HY5) is the central hub for seedling photomorphogenesis. E3 ubiquitin (Ub) ligase CONSTITUTIVE PHOTOMORPHOGENIC 1 (COP1) inhibits HY5 protein accumulation through ubiquitination. However, the process of HY5 deubiquitination, which antagonizes E3 ligase-mediated ubiquitination to maintain HY5 homeostasis has never been studied. Here, we identified that Arabidopsis thaliana deubiquitinating enzyme, Ub-SPECIFIC PROTEASE 14 (UBP14) physically interacts with HY5 and enhances its protein stability by deubiquitination. The da3-1 mutant lacking UBP14 function exhibited a long hypocotyl phenotype, and UBP14 deficiency led to the failure of rapid accumulation of HY5 during dark to light. In addition, UBP14 preferred to stabilize nonphosphorylated form of HY5 which is more readily bound to downstream target genes. HY5 promoted the expression and protein accumulation of UBP14 for positive feedback to facilitate photomorphogenesis. Our findings thus established a mechanism by which UBP14 stabilizes HY5 protein by deubiquitination to promote photomorphogenesis in A. thaliana.
    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
    背景:快速增殖是多形性胶质母细胞瘤(GBM)的标志,也是其复发的主要原因。异常泛素化与各种疾病有关,包括癌症.在我们的初步研究中,我们确定泛素结合酶E2S(UBE2S)作为潜在的神经胶质瘤生物标志物,与胶质瘤分级和蛋白磷酸酶1,调节亚基105(Ki67)表达水平密切相关。然而,潜在的分子机制仍然难以捉摸。NF-κB是促进GBM增殖的重要信号通路。靶向NF-κB的直接干预没有产生预期的结果,促使探索调控NF-κB的新分子成为新的方向。
    方法:本研究采用包括酵母双杂交和免疫沉淀的方法来揭示UBE2S和A激酶相互作用蛋白1(AKIP1)之间的相互作用。使用激光共聚焦显微镜观察UBE2S和AKIP1的定位。利用双荧光素酶报告基因观察NF-κB的激活。
    结果:我们的发现表明UBE2S缺乏显著阻碍GBM进展,在体外和体内。机械上,UBE2S在招募泛素特异性肽酶15(USP15)中起着至关重要的作用,促进AKIP1上K11连接的泛素化的去除。此动作增强了GBM上下文中的AKIP1稳定性。AKIP1水平的增加进一步增加了核因子κB(NF-κB)的转录活性,导致NF-κB通路调节的下游基因上调,从而促进GBM进展。
    结论:总之,我们的研究结果揭示了UBE2S/AKIP1-NF-κB轴在调节GBM进展中的作用,并提供了新的证据支持UBE2S作为GBM的潜在药物靶标.
    BACKGROUND: Rapid proliferation is a hallmark of glioblastoma multiforme (GBM) and a major contributor to its recurrence. Aberrant ubiquitination has been implicated in various diseases, including cancer. In our preliminary studies, we identified Ubiquitin-conjugating enzyme E2S (UBE2S) as a potential glioma biomarker, exhibiting close associations with glioma grade and protein phosphatase 1, regulatory subunit 105 (Ki67) expression levels. However, the underlying molecular mechanisms remained elusive. NF-κB is an important signaling pathway that promotes GBM proliferation. Direct intervention targeting NF-κB has not yielded the expected results, prompting the exploration of new molecules for regulating NF-κB as a new direction.
    METHODS: This study employed methods including yeast two-hybrid and immunoprecipitation to uncover the interaction between UBE2S and A kinase interacting protein 1 (AKIP1). Laser confocal microscopy was used to observe the localization of UBE2S and AKIP1. Dual luciferase reporter genes were utilized to observe the activation of NF-κB.
    RESULTS: Our findings demonstrate that UBE2S deficiency significantly impedes GBM progression, both in vitro and in vivo. Mechanistically, UBE2S plays a crucial role in recruiting Ubiquitin Specific Peptidase 15 (USP15), facilitating the removal of K11-linked ubiquitination on AKIP1. This action enhances AKIP1 stability within the GBM context. The resulting increase in AKIP1 levels further augments nuclear factor kappa-B (NF-κB) transcriptional activity, leading to the upregulation of downstream genes regulated by the NF-κB pathway, thereby promoting GBM progression.
    CONCLUSIONS: In summary, our findings reveal the role of the UBE2S/AKIP1-NF-κB axis in regulating GBM progression and provide novel evidence supporting UBE2S as a potential drug target for GBM.
    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
    肝脏缺血/再灌注(I/R)损伤是肝脏手术后肝功能损害的重要原因。泛素-蛋白酶体系统(UPS)在蛋白质质量控制中起着至关重要的作用,并对肝脏I/R过程产生重大影响。尽管OTU去泛素酶1(OTUD1)参与多种生物过程,其在肝脏I/R中的具体功能含义尚未完全了解。这项研究表明,OTUD1减轻了氧化应激,凋亡,和肝I/R损伤引起的炎症。机械上,OTUD1通过其催化位点半胱氨酸320残基和ETGE基序去泛素并激活核因子红系2相关因子2(NRF2),从而减轻肝脏I/R损伤。此外,施用含有ETGE基序的短肽显著减轻小鼠的肝I/R损伤。总的来说,我们的研究阐明了OTUD1在改善肝脏I/R损伤中的机制和作用,为使用ETGE-肽进行潜在治疗提供理论依据。
    Hepatic ischemia/reperfusion (I/R) injury is an important cause of liver function impairment following liver surgery. The ubiquitin-proteasome system (UPS) plays a crucial role in protein quality control and has substantial impact on the hepatic I/R process. Although OTU deubiquitinase 1 (OTUD1) is involved in diverse biological processes, its specific functional implications in hepatic I/R are not yet fully understood. This study demonstrates that OTUD1 alleviates oxidative stress, apoptosis, and inflammation induced by hepatic I/R injury. Mechanistically, OTUD1 deubiquitinates and activates nuclear factor erythroid 2-related factor 2 (NRF2) through its catalytic site cysteine 320 residue and ETGE motif, thereby attenuating hepatic I/R injury. Additionally, administration of a short peptide containing the ETGE motif significantly mitigates hepatic I/R injury in mice. Overall, our study elucidates the mechanism and role of OTUD1 in ameliorating hepatic I/R injury, providing a theoretical basis for potential treatment using ETGE-peptide.
    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
    背景:MutT同源物1(MTH1)对氧化的dNTP池进行消毒以促进癌细胞的存活,并且其表达在癌症中经常上调。聚泛素化稳定MTH1以促进黑色素瘤细胞的增殖,表明泛素系统控制MTH1的稳定性和功能。然而,泛素化是否调节胃癌中的MTH1尚未得到很好的定义。本研究旨在探讨MTH1与去泛素酶之间的相互作用,USP9X,在调节增殖方面,生存,迁移,和胃癌细胞的侵袭。
    方法:在HGC-27胃癌细胞中通过共免疫沉淀(co-IP)评估了USP9X与MTH1之间的相互作用。siRNA用于干扰胃癌细胞系HGC-27和MKN-45中的USP9X表达。进行MTT测定以检查增殖,碘化丙啶(PI)和7-AAD染色测定,以评估细胞周期,进行膜联蛋白V/PI染色测定以检查细胞凋亡,和transwell分析用于确定对照的迁移和入侵,缺乏USP9X,和USP9X缺陷加上MTH1过表达的HGC-27和MKN-45胃癌细胞。
    结果:Co-IP数据显示USP9X与MTH1相互作用并去泛素化。USP9X的过表达通过下调其泛素化提高MTH1蛋白水平,而USP9X的敲低对MTH1有相反的影响。HGC-27和MKN-45细胞中的USP9X缺乏导致增殖减少,细胞周期停滞,额外的凋亡,以及有缺陷的迁移和入侵,可以通过过量的MTH1来拯救。
    结论:USP9X与MTH1相互作用并稳定MTH1以促进其增殖,生存,胃癌细胞的迁移和侵袭。
    BACKGROUND: MutT homolog 1 (MTH1) sanitizes oxidized dNTP pools to promote the survival of cancer cells and its expression is frequently upregulated in cancers. Polyubiquitination stabilizes MTH1 to facilitate the proliferation of melanoma cells, suggesting the ubiquitin system controls the stability and function of MTH1. However, whether ubiquitination regulates MTH1 in gastric cancers has not been well defined. This study aims to investigate the interaction between MTH1 and a deubiquitinase, USP9X, in regulating the proliferation, survival, migration, and invasion of gastric cancer cells.
    METHODS: The interaction between USP9X and MTH1 was evaluated by co-immunoprecipitation (co-IP) in HGC-27 gastric cancer cells. siRNAs were used to interfere with USP9X expression in gastric cancer cell lines HGC-27 and MKN-45. MTT assays were carried out to examine the proliferation, propidium iodide (PI) and 7-AAD staining assays were performed to assess the cell cycle, Annexin V/PI staining assays were conducted to examine the apoptosis, and transwell assays were used to determine the migration and invasion of control, USP9X-deficient, and USP9X-deficient plus MTH1-overexpressing HGC-27 and MKN-45 gastric cancer cells.
    RESULTS: Co-IP data show that USP9X interacts with and deubiquitinates MTH1. Overexpression of USP9X elevates MTH1 protein level by downregulating its ubiquitination, while knockdown of USP9X has the opposite effect on MTH1. USP9X deficiency in HGC-27 and MKN-45 cells causes decreased proliferation, cell cycle arrest, extra apoptosis, and defective migration and invasion, which could be rescued by excessive MTH1.
    CONCLUSIONS: USP9X interacts with and stabilizes MTH1 to promote the proliferation, survival, migration and invasion of gastric cancer cells.
    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
    杨梅素,一种在各种食物中发现的天然类黄酮,研究了其对传染性胃肠炎病毒(TGEV)的抗病毒作用。这种α-冠状病毒在全球养猪业造成了巨大的经济损失。这项研究的重点是木瓜蛋白酶(PLpro),通过介导去泛素化在冠状病毒免疫逃避中起着至关重要的作用。靶向PLpro可能潜在地破坏病毒复制并增强抗病毒反应。结果表明,杨梅素以剂量依赖性方式有效抑制TGEV诱导的细胞病变效应,EC50值为31.19μM。杨梅素在8小时的共同潜伏期后48小时内显着降低了TGEV病毒载量。进一步的研究表明,浓度为100μM的杨梅素直接灭活TGEV并抑制其细胞内复制阶段。此外,用100μM杨梅素预处理对PK-15细胞具有抗TGEV感染的保护作用。杨梅素竞争性抑制PLpro,IC50值为6.563μM。分子对接实验表明,杨梅素通过常规氢键与PLpro的Cys102残基结合,Pi-硫磺,和Pi-烷基相互作用。通过定点诱变实验证实了这种结合,表明杨梅素是预防和治疗TGEV感染的潜在候选者。
    Myricetin, a natural flavonoid found in various foods, was investigated for its antiviral effect against transmissible gastroenteritis virus (TGEV). This α-coronavirus causes significant economic losses in the global swine industry. The study focused on the papain-like protease (PLpro), which plays a crucial role in coronavirus immune evasion by mediating deubiquitination. Targeting PLpro could potentially disrupt viral replication and enhance antiviral responses. The results demonstrated that myricetin effectively inhibited TGEV-induced cytopathic effects in a dose-dependent manner, with an EC50 value of 31.19 μM. Myricetin significantly reduced TGEV viral load within 48 h after an 8-h co-incubation period. Further investigations revealed that myricetin at a concentration of 100 μM directly inactivated TGEV and suppressed its intracellular replication stage. Moreover, pretreatment with 100 μM myricetin conferred a protective effect on PK-15 cells against TGEV infection. Myricetin competitively inhibited PLpro with an IC50 value of 6.563 μM. Molecular docking experiments show that myricetin binds to the Cys102 residue of PLpro through conventional hydrogen bonds, Pi-sulfur, and Pi-alkyl interactions. This binding was confirmed through site-directed mutagenesis experiments, indicating myricetin as a potential candidate for preventing and treating TGEV infection.
    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
    乳腺癌是全球最常见的女性恶性肿瘤。泛素特异性肽酶53(USP53)已被证明在几种实体瘤中发挥抑癌功能,但其在乳腺癌中的作用和潜在机制尚未明确阐明。因此,我们在生物信息学层面对这个问题进行了一系列详细的研究,临床组织,细胞功能和动物模型。我们发现USP53在乳腺癌标本中表达下调,并与临床分期呈负相关。功能的增益和丧失实验表明USP53抑制增殖,克隆发生,细胞周期和异种移植生长,以及诱导乳腺癌细胞凋亡和线粒体损伤。免疫共沉淀数据表明,USP53与含有11(ZMYND11)的锌指MYND型相互作用,并催化其去泛素化和稳定化。33-50个氨基酸的Cys-box结构域是USP53酶活性的关键,但对其与ZMYND11的绑定并不重要。拯救实验表明,USP53在乳腺癌细胞中的抗肿瘤作用至少部分由ZMYND11介导。USP53和ZMYND11均为乳腺癌的预后保护因素。USP53-ZMYND11轴可能是乳腺癌的潜在生物标志物或治疗靶点。这可以为诊断提供新的见解,治疗和预后。
    Breast cancer is the most common female malignancy worldwide. Ubiquitin-specific peptidase 53 (USP53) has been shown to exert cancer-suppressing functions in several solid tumors, but its role and the underlying mechanism in breast cancer has not been clearly elucidated. Therefore, we have carried out a series of detailed studies on this matter at the levels of bioinformatics, clinical tissue, cell function and animal model. We found that USP53 expression was downregulated in breast cancer specimens and was negatively correlated with the clinical stages. Gain- and loss-of-function experiments demonstrated USP53 inhibited proliferation, clonogenesis, cell cycle and xenograft growth, as well as induced apoptosis and mitochondrial damage of breast cancer cells. Co-immunoprecipitation data suggested that USP53 interacted with zinc finger MYND-type containing 11 (ZMYND11), and catalyzed its deubiquitination and stabilization. The 33-50 amino acid Cys-box domain was key for USP53 enzyme activity, but not essential for its binding with ZMYND11. The rescue experiments revealed that the anti-tumor role of USP53 in breast cancer cells was at least partially mediated by ZMYND11. Both USP53 and ZMYND11 were prognostic protective factors for breast cancer. USP53-ZMYND11 axis may be a good potential biomarker or therapeutic target for breast cancer, which can provide novel insights into the diagnosis, treatment and prognosis.
    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
    缺氧诱导因子(HIF)的泛素化和蛋白酶体介导的降解是后生动物氧敏感的核心,但是去泛素化酶(DUB)在HIF信号传导中的参与尚不清楚。这里,使用定制的DUBssgRNA文库,我们进行CRISPR/Cas9诱变筛选,以确定DUBs如何参与HIF信号传导。除了定义参与HIF激活或抑制的DUB,我们将USP43鉴定为有效激活HIF应答所需的DUB.USP43是缺氧调节的,并选择性地与HIF-1α同工型相关,虽然USP43不会改变HIF-1α的稳定性,它促进HIF-1核积累和与其靶基因的结合。机械上,USP43以缺氧和磷酸化依赖性方式与14-3-3蛋白结合以增加HIF-1的核库。一起,我们的结果强调了DUB的多功能性,说明它们可以提供重要的信号功能以及它们的催化作用。
    The ubiquitination and proteasome-mediated degradation of Hypoxia Inducible Factors (HIFs) is central to metazoan oxygen-sensing, but the involvement of deubiquitinating enzymes (DUBs) in HIF signalling is less clear. Here, using a bespoke DUBs sgRNA library we conduct CRISPR/Cas9 mutagenesis screens to determine how DUBs are involved in HIF signalling. Alongside defining DUBs involved in HIF activation or suppression, we identify USP43 as a DUB required for efficient activation of a HIF response. USP43 is hypoxia regulated and selectively associates with the HIF-1α isoform, and while USP43 does not alter HIF-1α stability, it facilitates HIF-1 nuclear accumulation and binding to its target genes. Mechanistically, USP43 associates with 14-3-3 proteins in a hypoxia and phosphorylation dependent manner to increase the nuclear pool of HIF-1. Together, our results highlight the multifunctionality of DUBs, illustrating that they can provide important signalling functions alongside their catalytic roles.
    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