OGT

OGT
  • 文章类型: Journal Article
    Tau病是几种神经退行性疾病的统称,其特征在于细胞内过度磷酸化的微管相关蛋白Tau(P-tau)的积累。我们最近的报告揭示了双氢青蒿素(DHA)通过增强O-连接-N-乙酰氨基葡萄糖(O-GlcNAcylation)修饰对海马中过表达人Tau(hTau)的小鼠的神经保护作用。然而,DHA是否可以通过特异性促进TauO-GlcNAcylation改善hTau转基因小鼠的突触和认知功能尚不清楚。这里,我们引入了hTau转基因小鼠,更理想的tau蛋白病变模型,研究DHA对TauO-GlcNAcylation的影响。我们报道,DHA治疗减轻了Barnes迷宫中海马CA1LTP和空间学习和记忆的缺陷,并减轻了hTau转基因小鼠的上下文恐惧条件测试。机械上,我们发现,DHA通过上调TauO-GlcNAcylation和减弱Tau过度磷酸化发挥了显着的保护作用。通过分子对接,我们发现DHA和O-GlcNAc转移酶(OGT)之间的稳定结合。我们进一步报道DHA处理对OGT的表达没有影响,但它促进了OGT核出口,从而增强OGT介导的TauO-GlcNAcylation。一起来看,这些结果表明,DHA通过促进OGT的细胞质易位和重建TauO-GlcNAcylation/磷酸化的平衡发挥神经保护作用,增强Tau的O-GlcNAcylation,这表明DHA可能是抗tau蛋白病的潜在治疗剂。
    Tauopathy is a collective term for several neurodegenerative diseases characterized by the intracellular accumulation of hyperphosphorylated microtubule-associated protein Tau (P-tau). Our recent report has revealed the neuroprotective effect of dihydroartemisinin (DHA) on mice overexpressing human Tau (hTau) in the hippocampus by enhancing O-linked-N-Acetylglucosaminylation (O-GlcNAcylation) modification. However, whether DHA can improve synaptic and cognitive function in hTau transgenic mice by specifically promoting Tau O-GlcNAcylation is still unclear. Here, we introduced hTau transgenic mice, a more optimal tauopathy model, to study the effect of DHA on Tau O-GlcNAcylation. We reported that DHA treatment alleviated the deficits of hippocampal CA1 LTP and spatial learning and memory in the Barnes maze and context fear conditioning tests in hTau transgenic mice. Mechanically, we revealed that DHA exerted a significant protective effect by upregulating Tau O-GlcNAcylation and attenuating Tau hyperphosphorylation. Through molecular docking, we found a stable binding between DHA and O-GlcNAc transferase (OGT). We further reported that DHA treatment had no effect on the expression of OGT, but it promoted OGT nuclear export, thereby enhancing OGT-mediated Tau O-GlcNAcylation. Taken together, these results indicate that DHA exerts neuroprotective effect by promoting cytoplasmic translocation of OGT and rebuilding the balance of Tau O-GlcNAcylation/phosphorylation, enhancing O-GlcNAcylation of Tau, suggesting that DHA may be a potential therapeutic agent against tauopathy.
    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
    妊娠期母体寨卡病毒(ZIKV)感染与严重的宫内生长受限(IUGR)有关。胎盘损伤,新陈代谢紊乱,和新生儿神经异常。这里,我们调查了母体ZIKV感染对胎盘营养转运蛋白和营养敏感途径的影响.免疫活性(C57BL/6)小鼠在妊娠日(GD)12.5注射低(103PFU-ZIKVPE243)或高(5X107PFU-ZIKVPE243)ZIKV滴度,并在GD18.5(足月)收集组织。男性胎儿的胎儿胎盘生长受损,ZIKV感染标记物的胎盘表达更高,真核翻译起始因子2(eIF2α),但较低水平的磷酸-eIF2α。女性胎儿的胎儿胎盘生长没有差异,胎盘ZIKV感染标志物无明显改变。此外,ZIKV促进女性胎盘中1型葡萄糖转运蛋白(Slc2a1/Glut1)的表达增加,6-磷酸葡萄糖水平降低,氨基酸转运潜力没有差异。相比之下,ZIKV不影响男性胎盘中的葡萄糖转运蛋白,但下调钠偶联中性氨基酸2(Snat2)转运蛋白的表达。我们还观察到ZIKV感染的妊娠中己糖胺生物合成途径(HBP)和O-GlcNAcylation的性别依赖性差异,表明ZIKV可以干扰胎盘营养感知。我们的发现强调了由母体ZIKV感染引起的胎盘分子改变,揭示养分运输,传感,和可用性。我们的结果还表明,女性和男性胎盘采用不同的应对机制来应对ZIKV诱导的代谢变化,为先天性寨卡综合征的治疗方法提供见解。
    Maternal Zika virus (ZIKV) infection during pregnancy has been associated with severe intrauterine growth restriction (IUGR), placental damage, metabolism disturbances, and newborn neurological abnormalities. Here, we investigated the impact of maternal ZIKV infection on placental nutrient transporters and nutrient-sensitive pathways. Immunocompetent (C57BL/6) mice were injected with Low (103 PFU-ZIKVPE243) or High (5 × 107 PFU-ZIKVPE243) ZIKV titers at gestational day (GD) 12.5, and tissue was collected at GD18.5 (term). Fetal-placental growth was impaired in male fetuses, which exhibited higher placental expression of the ZIKV infective marker, eukaryotic translation initiation factor 2 (eIF2α), but lower levels of phospho-eIF2α. There were no differences in fetal-placental growth in female fetuses, which exhibited no significant alterations in placental ZIKV infective markers. Furthermore, ZIKV promoted increased expression of glucose transporter type 1 (Slc2a1/Glut1) and decreased levels of glucose-6-phosphate in female placentae, with no differences in amino acid transport potential. In contrast, ZIKV did not impact glucose transporters in male placentae but downregulated sodium-coupled neutral amino acid 2 (Snat2) transporter expression. We also observed sex-dependent differences in the hexosamine biosynthesis pathway (HBP) and O-GlcNAcylation in ZIKV-infected pregnancies, showing that ZIKV can disturb placental nutrient sensing. Our findings highlight molecular alterations in the placenta caused by maternal ZIKV infection, shedding light on nutrient transport, sensing, and availability. Our results also suggest that female and male placentae employ distinct coping mechanisms in response to ZIKV-induced metabolic changes, providing insights into therapeutic approaches for congenital Zika syndrome.
    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
    泛素化被认为是肝内胆管癌(iCCA)发展的关键因素。在这里,我们确定泛素特异性肽酶8(USP8)是通过稳定OGT促进iCCA细胞肿瘤发生的关键调节因子。USP8在人肿瘤组织中过表达并且与较差的存活率相关。此外,质谱和免疫共沉淀分析表明USP8与OGT相互作用。USP8是OGT的真正去泛素酶。它以去泛素化活性依赖性方式稳定OGT。同时,DUB-IN3,USP8抑制剂,还可以抑制肝内胆管癌的恶性程度。此外,USP8耗竭促进iCCA对培米卡替尼的反应。总之,我们的研究结果指出了USP8作为OGT的去泛素化酶的催化作用.USP8-OGT轴可能是iCCA治疗的潜在靶标。
    Ubiquitination was considered to be a crucial factor in intrahepatic cholangiocarcinoma (iCCA) development. Herein, we identified Ubiquitin-specific peptidase 8 (USP8) as a key regulator for promoting the tumorigenesis of iCCA cell via stabilizing OGT. USP8 was overexpressed in human tumor tissues and correlated with worse survival. Moreover, the mass spectrometry and co-immunoprecipitation analysis indicated that USP8 interacted with OGT. USP8 worked as a bona fide deubiquitylase of OGT. It stabilized OGT in a deubiquitylation activity-dependent manner. Meanwhile, DUB-IN3, the USP8 inhibitor, could also restrain the malignancy of intrahepatic cholangiocarcinoma. In addition, USP8 depletion promoted the response of iCCA to pemigatinib. In conclusion, our findings pointed to a previously undocumented catalytic role for USP8 as a deubiquitinating enzyme of OGT. The USP8-OGT axis could be a potential target for iCCA 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
    循环系统是贯穿全身的封闭管道系统,由以下两部分组成:心血管系统和淋巴系统。血液恶性肿瘤通常在循环系统中生长和繁殖,直接或间接影响其功能。这些恶性肿瘤包括多发性骨髓瘤,白血病,和淋巴瘤。O-连接的β-N-乙酰葡糖胺(O-GlcNAc)转移酶(OGT)通过O-GlcNAc修饰调节底物蛋白的功能和稳定性。OGT异常表达与肿瘤发生密切相关,包括血液恶性肿瘤,结直肠癌,肝癌,乳腺癌,和前列腺癌。在细胞中,OGT可以与多种蛋白质组装形成复合物,以行使相关的生物学功能,如OGT/HCF-1,OGT/TET,NSL,然后调节葡萄糖代谢,基因转录,细胞增殖,和其他生物过程,从而影响血液系统恶性肿瘤的发展。本文综述了参与OGT在细胞中组装的复合物以及相关OGT复合物在血液系统恶性肿瘤中的作用。解开由OGT复合物调节的复杂网络将有助于更好地理解血液系统恶性肿瘤的发展和进展。
    The circulatory system is a closed conduit system throughout the body and consists of two parts as follows: the cardiovascular system and the lymphatic system. Hematological malignancies usually grow and multiply in the circulatory system, directly or indirectly affecting its function. These malignancies include multiple myeloma, leukemia, and lymphoma. O-linked β-N-acetylglucosamine (O-GlcNAc) transferase (OGT) regulates the function and stability of substrate proteins through O-GlcNAc modification. Abnormally expressed OGT is strongly associated with tumorigenesis, including hematological malignancies, colorectal cancer, liver cancer, breast cancer, and prostate cancer. In cells, OGT can assemble with a variety of proteins to form complexes to exercise related biological functions, such as OGT/HCF-1, OGT/TET, NSL, and then regulate glucose metabolism, gene transcription, cell proliferation, and other biological processes, thus affecting the development of hematological malignancies. This review summarizes the complexes involved in the assembly of OGT in cells and the role of related OGT complexes in hematological malignancies. Unraveling the complex network regulated by the OGT complex will facilitate a better understanding of hematologic malignancy development and progression.
    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
    O-连接的β-N-乙酰葡糖胺(O-GlcNAc)转移酶(OGT)是催化细胞内所有O-GlcNAc酰化反应的唯一酶。先前的研究发现,OGT水平在细胞分裂过程中会振荡。具体来说,OGT丰度在有丝分裂期间下调,但是缺乏潜在的机制。在这里,我们证明OGT被泛素E3连接酶泛素化,后期促进复合物/环小体(APC/C)-细胞分裂周期20(Cdc20)。我们表明APC/CCdc20通过保守的破坏盒(D盒)与OGT相互作用:Arg-351/Leu-354,其废除使OGT稳定。由于APC/CCdc20-底物结合通常在引发泛素化事件之前,我们还使用了质谱,并将OGTLys-352映射为泛素化位点,这是OGT与APC/C亚基关联的先决条件。有趣的是,在癌症基因组图谱中,R351C是子宫癌突变体,表明D-box的突变与肿瘤发生有关。矛盾的是,我们发现R351C和D-box突变体(R351A/L354A)在小鼠异种移植模型中抑制子宫癌,可能是由于细胞分裂和增殖受损。总之,我们提出了一个模型,其中OGTLys-352泛素化引发其与APC/C的结合,然后APC/CCdc20通过D-box与OGT合作进行有丝分裂破坏。我们的工作不仅突出了细胞周期中调节OGT的关键机制,而且还揭示了糖基化与细胞分裂机制之间的相互协调。
    O-linked β-N-acetylglucosamine (O-GlcNAc) transferase (OGT) is the sole enzyme that catalyzes all O-GlcNAcylation reactions intracellularly. Previous investigations have found that OGT levels oscillate during the cell division process. Specifically, OGT abundance is downregulated during mitosis, but the underlying mechanism is lacking. Here we demonstrate that OGT is ubiquitinated by the ubiquitin E3 ligase, anaphase promoting complex/cyclosome (APC/C)-cell division cycle 20 (Cdc20). We show that APC/CCdc20 interacts with OGT through a conserved destruction box (D-box): Arg-351/Leu-354, the abrogation of which stabilizes OGT. As APC/CCdc20-substrate binding is often preceded by a priming ubiquitination event, we also used mass spectrometry and mapped OGT Lys-352 to be a ubiquitination site, which is a prerequisite for OGT association with APC/C subunits. Interestingly, in The Cancer Genome Atlas, R351C is a uterine carcinoma mutant, suggesting that mutations of the D-box are linked with tumorigenesis. Paradoxically, we found that both R351C and the D-box mutants (R351A/L354A) inhibit uterine carcinoma in mouse xenograft models, probably due to impaired cell division and proliferation. In sum, we propose a model where OGT Lys-352 ubiquitination primes its binding with APC/C, and then APC/CCdc20 partners with OGT through the D-box for its mitotic destruction. Our work not only highlights the key mechanism that regulates OGT during the cell cycle, but also reveals the mutual coordination between glycosylation and the cell division machinery.
    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
    O-连接-β-D-N-乙酰葡糖胺(O-GlcNAc)糖基化(O-GlcNAcylation),由O-GlcNAc转移酶(OGT)和O-GlcNAcase(OGA)动态调节,是参与多个细胞过程的翻译后修饰。蛋白质的O-GlcNAcylation可以通过与其他翻译后修饰的串扰来调节其生物学功能,比如磷酸化,泛素化,乙酰化,和甲基化。肝脏疾病是全世界死亡的主要原因,该疾病的关键病理特征,比如炎症,纤维化,脂肪变性,和肿瘤发生,没有完全理解。O-GlcNAcylation的失调已被证明与一些严重的肝细胞应激有关。病毒性肝炎,肝纤维化,非酒精性脂肪酸肝病(NAFLD),恶性进展,肝细胞癌(HCC)的耐药通过多分子信号通路。这里,我们总结了O-GlcNAcylation与肝脏病理过程之间的新联系,并提供了有关肝病治疗策略发展的信息。
    O-linked-β-D-N-acetylglucosamine (O-GlcNAc) glycosylation (O-GlcNAcylation), which is dynamically regulated by O-GlcNAc transferase (OGT) and O-GlcNAcase (OGA), is a post-translational modification involved in multiple cellular processes. O-GlcNAcylation of proteins can regulate their biological functions via crosstalk with other post-translational modifications, such as phosphorylation, ubiquitination, acetylation, and methylation. Liver diseases are a major cause of death worldwide; yet, key pathological features of the disease, such as inflammation, fibrosis, steatosis, and tumorigenesis, are not fully understood. The dysregulation of O-GlcNAcylation has been shown to be involved in some severe hepatic cellular stress, viral hepatitis, liver fibrosis, nonalcoholic fatty acid liver disease (NAFLD), malignant progression, and drug resistance of hepatocellular carcinoma (HCC) through multiple molecular signaling pathways. Here, we summarize the emerging link between O-GlcNAcylation and hepatic pathological processes and provide information about the development of therapeutic strategies for liver diseases.
    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:治疗性低温(MTH)是围手术期防治心肌缺血再灌注损伤(MIRI)的重要方法。在蛋白质翻译后修饰线粒体蛋白以调节线粒体功能是改善心肌缺血再灌注损伤的机制之一。本研究探讨了浅低温治疗改善心肌缺血再灌注损伤与COX10的O-GlcNAcylation水平之间的关系。
    方法:我们使用体内Langendorff模型和体外缺氧/复氧(H/R)细胞模型来研究MTH对心肌缺血再灌注损伤的影响。组织学变化,心肌酶,氧化应激,和线粒体结构/功能进行评估。机理研究涉及各种分子生物学方法,如ELISA,免疫沉淀(IP),WB,和免疫荧光。
    结果:我们的研究结果表明,MTH上调了COX10的O-GlcNAC化水平,改善了线粒体功能,抑制ROS的表达,改善心肌缺血再灌注损伤。在体内,MTH能有效缓解缺血再灌注引起的心功能不全,心肌损伤,线粒体损伤,和氧化还原失衡。体外,OGT抑制剂ALX抑制OGT介导的O-GlcNA酰化信号通路,下调COX10的O-Glc酰化水平,促进ROS释放,并抵消了MTH的保护作用。相反,OGA抑制剂ThG显示出与ALX相反的作用,进一步证实MTH激活OGT介导的O-GlcNAcylation信号通路发挥心脏保护作用。
    结论:总之,MTH激活OGT介导的O-糖基化修饰的COX10调节线粒体功能,改善心肌缺血再灌注损伤,为MTH的临床应用提供了重要的理论依据。
    OBJECTIVE: Mild therapeutic hypothermia (MTH) is an important method for perioperative prevention and treatment of myocardial ischemia-reperfusion injury (MIRI). Modifying mitochondrial proteins after protein translation to regulate mitochondrial function is one of the mechanisms for improving myocardial ischemia-reperfusion injury. This study investigated the relationship between shallow hypothermia treatment improving myocardial ischemia-reperfusion injury and the O-GlcNAcylation level of COX10.
    METHODS: We used in vivo Langendorff model and in vitro hypoxia/reoxygenation (H/R) cell model to investigate the effects of MTH on myocardial ischemia-reperfusion injury. Histological changes, myocardial enzymes, oxidative stress, and mitochondrial structure/function were assessed. Mechanistic studies involved various molecular biology methods such as ELISA, immunoprecipitation (IP), WB, and immunofluorescence.
    RESULTS: Our research results indicate that MTH upregulates the O-GlcNACylation level of COX10, improves mitochondrial function, and inhibits the expression of ROS to improve myocardial ischemia-reperfusion injury. In vivo, MTH effectively alleviates ischemia-reperfusion induced cardiac dysfunction, myocardial injury, mitochondrial damage, and redox imbalance. In vitro, the OGT inhibitor ALX inhibits the OGT mediated O-GlcNA acylation signaling pathway, downregulates the O-Glc acylation level of COX10, promotes ROS release, and counteracts the protective effect of MTH. On the contrary, the OGA inhibitor ThG showed opposite effects to ALX, further confirming that MTH activated the OGT mediated O-GlcNAcylation signaling pathway to exert cardioprotective effects.
    CONCLUSIONS: In summary, MTH activates OGT mediated O-glycosylation modified COX10 to regulate mitochondrial function and improve myocardial ischemia-reperfusion injury, which provides important theoretical basis for the clinical application of MTH.
    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
    O-GlcNAc转移酶(OGT)是一种必需的哺乳动物酶,可糖基化多种细胞内蛋白质并切割转录共调节因子宿主细胞因子1以调节细胞周期过程。通过这些催化活性以及非催化蛋白质-蛋白质相互作用,OGT维持细胞稳态。OGT的四三肽重复(TPR)结构域在底物识别中很重要,但是关于改变TPR域如何影响其细胞功能的信息很少。这里,我们研究了内源性酶缺失后,改变OGT的TPR结构域如何影响细胞生长。我们发现,破坏OGT二聚化所需的TPR残基会导致更快的细胞生长,而截短TPR结构域会减缓细胞生长。我们还发现OGT需要其13个TPR中的8个来维持细胞活力。OGT-8,就像不可行的较短OGT变体一样,错误定位,并降低了Ser/Thr糖基化活性;此外,它与大多数野生型OGT的结合配偶体的相互作用被广泛减弱。因此,虽然OGT的五个N端TPR对细胞活力不是必需的,它们是正确的亚细胞定位和介导许多OGT蛋白-蛋白相互作用所必需的。因为我们已经确定的可行的OGT截断变体保留了OGT的基本功能,这可能有助于他们的识别。
    O-GlcNAc transferase (OGT) is an essential mammalian enzyme that glycosylates myriad intracellular proteins and cleaves the transcriptional coregulator Host Cell Factor 1 to regulate cell cycle processes. Via these catalytic activities as well as noncatalytic protein-protein interactions, OGT maintains cell homeostasis. OGT\'s tetratricopeptide repeat (TPR) domain is important in substrate recognition, but there is little information on how changing the TPR domain impacts its cellular functions. Here, we investigate how altering OGT\'s TPR domain impacts cell growth after the endogenous enzyme is deleted. We find that disrupting the TPR residues required for OGT dimerization leads to faster cell growth, whereas truncating the TPR domain slows cell growth. We also find that OGT requires eight of its 13 TPRs to sustain cell viability. OGT-8, like the nonviable shorter OGT variants, is mislocalized and has reduced Ser/Thr glycosylation activity; moreover, its interactions with most of wild-type OGT\'s binding partners are broadly attenuated. Therefore, although OGT\'s five N-terminal TPRs are not essential for cell viability, they are required for proper subcellular localization and for mediating many of OGT\'s protein-protein interactions. Because the viable OGT truncation variant we have identified preserves OGT\'s essential functions, it may facilitate their identification.
    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
    造血在整个生命中持续产生来自造血干细胞(HSC)的所有类型的血细胞。代谢状态是HSC自我更新和分化的已知调节因子,但是代谢传感器是否以及如何进行O-GlcNAcylation,可以通过抑制其循环酶O-GlcNAcase(OGA)和O-GlcNAc转移酶(OGT)来调节,对造血的贡献在很大程度上仍然未知。在这里,等基因,OGA耗尽(OGAi)和OGT耗尽(OGTi)人类诱导多能干细胞(hiPSCs)的单细胞克隆成功地从主hiPSC系MUSIi012-A,从包含表观遗传记忆的CD34造血干/祖细胞(HSPC)重新编程。已建立的OGAi和OGTihiPSC表现出细胞O-GlcNAcylation的增加或减少,伴随着OGA和OGT的丧失,分别,表型和核型正常,并保留了多能性,尽管它们可能倾向于向某些细菌谱系分化。在通过中胚层诱导和内皮到造血转变进行造血分化时,我们发现OGA抑制加速了hiPSC对HSPCs的承诺,而O-GlcNAc稳态的破坏会影响其对红系谱系的承诺.来自所有组的分化的HSPC能够产生所有造血祖细胞,从而证实了它们的功能特征。总之,已建立的OGTi和OGAihiPSC的单细胞克隆为进一步剖析O-GlcNAcylation在血细胞发育中不同阶段和谱系的作用提供了有价值的平台.这些hiPSC中OGA和OGT的不完全敲除使它们容易受到额外的操纵,即,通过小分子,允许O-GlcNAcylation的分子动力学研究。
    Hematopoiesis continues throughout life to produce all types of blood cells from hematopoietic stem cells (HSCs). Metabolic state is a known regulator of HSC self-renewal and differentiation, but whether and how metabolic sensor O-GlcNAcylation, which can be modulated via an inhibition of its cycling enzymes O-GlcNAcase (OGA) and O-GlcNAc transferase (OGT), contributes to hematopoiesis remains largely unknown. Herein, isogenic, single-cell clones of OGA-depleted (OGAi) and OGT-depleted (OGTi) human induced pluripotent stem cells (hiPSCs) were successfully generated from the master hiPSC line MUSIi012-A, which were reprogrammed from CD34+ hematopoietic stem/progenitor cells (HSPCs) containing epigenetic memory. The established OGAi and OGTi hiPSCs exhibiting an increase or decrease in cellular O-GlcNAcylation concomitant with their loss of OGA and OGT, respectively, appeared normal in phenotype and karyotype, and retained pluripotency, although they may favor differentiation toward certain germ lineages. Upon hematopoietic differentiation through mesoderm induction and endothelial-to-hematopoietic transition, we found that OGA inhibition accelerates hiPSC commitment toward HSPCs and that disruption of O-GlcNAc homeostasis affects their commitment toward erythroid lineage. The differentiated HSPCs from all groups were capable of giving rise to all hematopoietic progenitors, thus confirming their functional characteristics. Altogether, the established single-cell clones of OGTi and OGAi hiPSCs represent a valuable platform for further dissecting the roles of O-GlcNAcylation in blood cell development at various stages and lineages of blood cells. The incomplete knockout of OGA and OGT in these hiPSCs makes them susceptible to additional manipulation, i.e., by small molecules, allowing the molecular dynamics studies of O-GlcNAcylation.
    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
    O-GlcNAcylation的失调已经成为几种疾病的潜在生物标志物。尤其是癌症。OGT(O-GlcNAc转移酶)在维持O-GlcNAc稳态中的作用已得到广泛研究;然而,OGA(O-GlcNAcase)在癌症中的调控仍然难以捉摸。这里,我们证明了多功能蛋白RBM14是细胞O-GlcNAcylation的调节因子。通过研究肺癌细胞中O-GlcNAcylation升高与RBM14表达增加之间的相关性,我们发现RBM14促进OGA的泛素依赖性蛋白酶体降解,最终介导细胞O-GlcNAcylation水平。此外,RBM14本身在丝氨酸521处被O-GlcNAcylated,调节其与E3连接酶TRIM33的相互作用,因此影响OGA蛋白的稳定性。此外,我们证明,丝氨酸521突变为丙氨酸消除了RBM14的致癌特性。总的来说,我们的研究结果揭示了以前未知的OGA调控机制,并提示了治疗O-GlcNAcylation失调的癌症的潜在治疗靶点.
    Dysregulation of O-GlcNAcylation has emerged as a potential biomarker for several diseases, particularly cancer. The role of OGT (O-GlcNAc transferase) in maintaining O-GlcNAc homeostasis has been extensively studied; nevertheless, the regulation of OGA (O-GlcNAcase) in cancer remains elusive. Here, we demonstrated that the multifunctional protein RBM14 is a regulator of cellular O-GlcNAcylation. By investigating the correlation between elevated O-GlcNAcylation and increased RBM14 expression in lung cancer cells, we discovered that RBM14 promotes ubiquitin-dependent proteasomal degradation of OGA, ultimately mediating cellular O-GlcNAcylation levels. In addition, RBM14 itself is O-GlcNAcylated at serine 521, regulating its interaction with the E3 ligase TRIM33, consequently affecting OGA protein stability. Moreover, we demonstrated that mutation of serine 521 to alanine abrogated the oncogenic properties of RBM14. Collectively, our findings reveal a previously unknown mechanism for the regulation of OGA and suggest a potential therapeutic target for the treatment of cancers with dysregulated O-GlcNAcylation.
    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