RNF5

RNF5
  • 文章类型: Journal Article
    背景:心肌梗塞(MI)是全球范围内发病率和死亡率高的主要疾病。然而,现有的治疗方法远不能令人满意,使得探索有效的分子靶标更加势在必行。先前已报道E3泛素连接酶RING指蛋白5(RNF5)通过调节泛素化介导的蛋白质降解而参与几种疾病。然而,很少有报道关注它在心血管疾病中的作用,包括MI。
    方法:在本研究中,我们通过精确的CRISPR介导的基因组编辑建立了RNF5基因敲除小鼠,并在9-11周龄的雄性C57BL/6小鼠中使用了左冠状动脉前降支结扎.随后,进行血清生化分析和心脏组织病理学检查。此外,我们设计了调节RNF5表达的腺病毒,并对新生大鼠心肌细胞进行氧-葡萄糖剥夺(OGD)以模拟缺血条件,证明RNF5操作对细胞活力的影响。基因和蛋白质表达分析提供了对分子机制的见解。严格采用统计方法来评估实验结果的重要性。
    结果:我们发现RNF5在接受OGD治疗的小鼠和NRCM的梗死心脏组织中下调。RNF5基因敲除导致小鼠心脏功能障碍加剧,更严重的炎症反应,MI手术后细胞凋亡增加。体外,RNF5敲低加剧了OGD诱导的细胞活性下降,细胞凋亡增加,而RNF5过表达则有相反的作用。机械上,已证明,由凋亡信号调节激酶1(ASK1)激活引发的激酶级联受RNF5的密切调节,并介导了RNF5在MI期间的保护功能。
    结论:我们证明了RNF5对心肌梗死的保护作用,其功能依赖于抑制ASK1的激活,这为心肌梗死相关网络增加了新的调节成分,并有望实现新的治疗策略。
    BACKGROUND: Myocardial infarction (MI) is a major disease with high morbidity and mortality worldwide. However, existing treatments are far from satisfactory, making the exploration of potent molecular targets more imperative. The E3 ubiquitin ligase RING finger protein 5 (RNF5) has been previously reported to be involved in several diseases by regulating ubiquitination-mediated protein degradation. Nevertheless, few reports have focused on its function in cardiovascular diseases, including MI.
    METHODS: In this study, we established RNF5 knockout mice through precise CRISPR-mediated genome editing and utilized left anterior descending coronary artery ligation in 9-11-week-old male C57BL/6 mice. Subsequently, serum biochemical analysis and histopathological examination of heart tissues were performed. Furthermore, we engineered adenoviruses for modulating RNF5 expression and subjected neonatal rat cardiomyocytes to oxygen-glucose deprivation (OGD) to mimic ischemic conditions, demonstrating the impact of RNF5 manipulation on cellular viability. Gene and protein expression analysis provided insights into the molecular mechanisms. Statistical methods were rigorously employed to assess the significance of experimental findings.
    RESULTS: We found RNF5 was downregulated in infarcted heart tissue of mice and NRCMs subjected to OGD treatment. RNF5 knockout in mice resulted in exacerbated heart dysfunction, more severe inflammatory responses, and increased apoptosis after MI surgery. In vitro, RNF5 knockdown exacerbated the OGD-induced decline in cell activity, increased apoptosis, while RNF5 overexpression had the opposite effect. Mechanistically, it was proven that the kinase cascade initiated by apoptosis signal-regulating kinase 1 (ASK1) activation was closely regulated by RNF5 and mediated RNF5\'s protective function during MI.
    CONCLUSIONS: We demonstrated the protective effect of RNF5 on myocardial infarction and its function was dependent on inhibiting the activation of ASK1, which adds a new regulatory component to the myocardial infarction associated network and promises to enable new therapeutic strategy.
    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
    RNF5是一种E3泛素连接酶,参与各种生理过程,如蛋白质定位和癌症进展。最近的研究表明,RNF5通过促进STING和MAVS的泛素化和降解,显著抑制抗病毒先天性免疫,它们是必需的衔接蛋白,以及它们的下游信号IRF3。RNF5的丰度受到宿主因子和病毒两者的微妙调控。已发现宿主因子限制RNF5介导的泛素化,通过不同的机制保持STING或MAVS的稳定性。同时,病毒已经开发出巧妙的策略来劫持RNF5来泛素化和降解免疫蛋白。此外,最近的研究揭示了RNF5在各种病毒生命周期中的多方面作用,包括SARS-CoV-2和KSHV。基于这些新发现,RNF5代表调节抗病毒免疫的新手段。在这次审查中,综述了RNF5在抗病毒免疫和病毒生命周期中作用的最新研究进展。这种全面的理解可以为探索在病毒感染期间专注于靶向RNF5的潜在治疗应用提供有价值的见解。
    RNF5 is an E3 ubiquitin ligase involved in various physiological processes such as protein localization and cancer progression. Recent studies have shown that RNF5 significantly inhibits antiviral innate immunity by promoting the ubiquitination and degradation of STING and MAVS, which are essential adaptor proteins, as well as their downstream signal IRF3. The abundance of RNF5 is delicately regulated by both host factors and viruses. Host factors have been found to restrict RNF5-mediated ubiquitination, maintaining the stability of STING or MAVS through distinct mechanisms. Meanwhile, viruses have developed ingenious strategies to hijack RNF5 to ubiquitinate and degrade immune proteins. Moreover, recent studies have revealed the multifaceted roles of RNF5 in the life cycle of various viruses, including SARS-CoV-2 and KSHV. Based on these emerging discoveries, RNF5 represents a novel means of modulating antiviral immunity. In this review, we summarize the latest research on the roles of RNF5 in antiviral immunity and virus life cycle. This comprehensive understanding could offer valuable insights into exploring potential therapeutic applications focused on targeting RNF5 during viral infections.
    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
    在整个生命周期中,病毒与细胞宿主因子相互作用,从而影响传播,主机范围,细胞嗜性和发病机制。戊型肝炎病毒(HEV)是一种被低估的RNA病毒,迄今为止对病毒-宿主相互作用网络的了解有限。这里,两种相关的高通量哺乳动物双杂交方法(MAPPIT和KISS)用于筛选与HEV相互作用的宿主蛋白。对蛋白质功能进行了检查,涉及途径,以及它们与其他病毒的关系。我们确定了37个ORF2命中,ORF3为187,ORF4为91。一些命中在不同病毒的生命周期中具有功能。我们专注于SHARPIN和RNF5作为ORF3的候选命中,因为它们参与病毒感染期间的RLR-MAVS途径和干扰素(IFN)诱导。敲除(KO)SHARPIN和RNF5导致ORF3转染后不同的IFN应答,与野生型细胞相比。此外,SHARPINKO细胞感染增加,RNF5KO细胞感染减少。总之,MAPPIT和KISS是研究病毒与宿主相互作用的有价值的工具,提供对鲜为人知的HEV生命周期的见解。我们进一步提供了两种确定的命中作为HEV生命周期中的新宿主因子的证据。
    Throughout their life cycle, viruses interact with cellular host factors, thereby influencing propagation, host range, cell tropism and pathogenesis. The hepatitis E virus (HEV) is an underestimated RNA virus in which knowledge of the virus-host interaction network to date is limited. Here, two related high-throughput mammalian two-hybrid approaches (MAPPIT and KISS) were used to screen for HEV-interacting host proteins. Promising hits were examined on protein function, involved pathway(s), and their relation to other viruses. We identified 37 ORF2 hits, 187 for ORF3 and 91 for ORF4. Several hits had functions in the life cycle of distinct viruses. We focused on SHARPIN and RNF5 as candidate hits for ORF3, as they are involved in the RLR-MAVS pathway and interferon (IFN) induction during viral infections. Knocking out (KO) SHARPIN and RNF5 resulted in a different IFN response upon ORF3 transfection, compared to wild-type cells. Moreover, infection was increased in SHARPIN KO cells and decreased in RNF5 KO cells. In conclusion, MAPPIT and KISS are valuable tools to study virus-host interactions, providing insights into the poorly understood HEV life cycle. We further provide evidence for two identified hits as new host factors in the HEV life cycle.
    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E3连接酶UBE3C促进胞浆蛋白和内质网(ER)膜蛋白的蛋白酶体降解。UBE3C被提议在RNF185/MBRLER相关降解(ERAD)分支的下游发挥作用,有助于选择膜蛋白的ERAD。这里,我们报告说,UBE3C促进了错误折叠的CFTR的ERAD,即使不存在RNF185及其功能性直系同源RNF5(RNF5/185)。与RNF5/185不同,UBE3C对错误折叠的CFTR的泛素化作用有限。当与RNF5/185消融结合时,UBE3C敲低(KD)导致质膜上的功能性ΔF508-CFTR通道额外增加,特别是在临床使用的CFTR调节剂的存在下。有趣的是,尽管UBE3CKD未能减弱insig-1的ERAD,但它降低了错误折叠的ΔY490-ABCB1的ERAD并增加了细胞表面表达。UBE3CKD还稳定了ΔF508-CFTR的成熟形式,并增加了T70-CFTR的细胞表面水平,VI类CFTR突变体。这些结果表明,UBE3C在错误折叠的CFTR和ABCB1的ERAD中起着至关重要的作用,甚至在不依赖RNF5/185的ERAD通路内,它也可能参与维持CFTR的外周质量控制。
    The ubiquitin E3 ligase UBE3C promotes the proteasomal degradation of cytosolic proteins and endoplasmic reticulum (ER) membrane proteins. UBE3C is proposed to function downstream of the RNF185/MBRL ER-associated degradation (ERAD) branch, contributing to the ERAD of select membrane proteins. Here, we report that UBE3C facilitates the ERAD of misfolded CFTR, even in the absence of both RNF185 and its functional ortholog RNF5 (RNF5/185). Unlike RNF5/185, UBE3C had a limited impact on the ubiquitination of misfolded CFTR. UBE3C knockdown (KD) resulted in an additional increase in the functional ∆F508-CFTR channels on the plasma membrane when combined with the RNF5/185 ablation, particularly in the presence of clinically used CFTR modulators. Interestingly, although UBE3C KD failed to attenuate the ERAD of insig-1, it reduced the ERAD of misfolded ∆Y490-ABCB1 and increased cell surface expression. UBE3C KD also stabilized the mature form of ∆F508-CFTR and increased the cell surface level of T70-CFTR, a class VI CFTR mutant. These results suggest that UBE3C plays a vital role in the ERAD of misfolded CFTR and ABCB1, even within the RNF5/185-independent ERAD pathway, and it may also be involved in maintaining the peripheral quality control of CFTR.
    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
    泛素化是蛋白质的重要翻译后修饰(PTM)之一,在调节底物降解以确保细胞稳态中起着至关重要的作用。环指蛋白5(RNF5)是哺乳动物中抑制STING介导的干扰素(IFN)信号传导的必需E3泛素连接酶。然而,RNF5在STING/IFN途径中的功能在硬骨鱼中仍然不清楚。这里,我们报道过表达乌鱼RNF5(bcRNF5)抑制STING介导的bcIFNa转录活性,DrIFNφ1,NF-κB和ISRE启动子和抗SVCV的抗病毒活性。此外,bcRNF5的敲减增加了宿主基因的表达,包括bcIFNa,bcIFNb,bcILβ,bcMX1和bcViperin,还增强了宿主细胞的抗病毒能力。免疫荧光(IF)和免疫共沉淀(Co-IP)测定证实bcRNF5主要位于细胞质中并与bcSTING相互作用。bcRNF5和MG132共同表达的bcSTING蛋白的表达水平减弱了这种减弱作用,提示bcRNF5介导的bcSTING降解依赖于蛋白酶体途径。随后,Co-IP和免疫印迹(IB)实验鉴定bcRNF5触发bcSTING的K48-连接而不是K63-连接的泛素化。总之,以上结果得出结论,RNF5通过增强K48连接的泛素化和黑鱼中STING的蛋白酶降解来抑制STING/IFN信号传导。
    Ubiquitination is one of the important post-translational modifications (PTMs) of proteins that plays a vital role in regulating substrate degradation to ensure cellular homeostasis. Ring finger protein 5 (RNF5) is an essential E3 ubiquitin ligase for inhibiting STING-mediated interferon (IFN) signaling in mammals. Nevertheless, the function of RNF5 in STING/IFN pathway remains obscure in teleost. Here, we reported that over-expression of black carp RNF5 (bcRNF5) inhibited STING-mediated transcription activity of bcIFNa, DrIFNφ1, NF-κB and ISRE promoters and antiviral activity against SVCV. Moreover, knockdown of bcRNF5 increased the expression of host genes, including bcIFNa, bcIFNb, bcILβ, bcMX1 and bcViperin, and also enhanced the antiviral capability of host cells. Immunofluorescence (IF) and Co-immunoprecipitation (Co-IP) assay confirmed that bcRNF5 was mainly localized in the cytoplasm and interacted with bcSTING. The expression level of bcSTING protein was attenuated by co-expressed bcRNF5 and MG132 treatment rescued this attenuating effect, suggesting that bcRNF5-mediated bcSTING degradation was dependent on the proteasome pathway. Subsequent, Co-IP and immunoblot (IB) experiments identified that bcRNF5 triggered the K48-linked but not K63-linked ubiquitination of bcSTING. Altogether, above results conclude that RNF5 suppresses STING/IFN signaling by enhancing K48-linked ubiquitination and protease degradation of STING in black carp.
    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
    Mitochondrial antiviral signaling protein (MAVS) is as an adaptor in RIG-I like receptor (RLR) signaling, which plays the key role in interferon (IFN) production during host antiviral innate immune activation. MAVS is fine tuned to avoid excess IFN production, which have been extensively studied in human and mammals. However, the regulation of MAVS in teleost still remains obscure. In this manuscript, we cloned ring finger protein 5 (bcRNF5) of black carp (Mylopharyngodon piceus) and characterized this teleost E3 ubiquitin ligase as a negative regulator of MAVS. The coding region of bcRNF5 consists of 615 nucleotides which encode 205 amino acids, containing two trans-membrane domain (TM) and a ring-finger domain (RING). The transcription regulation of bcRNF5 varies in host cells in response to stimulations of LPS, poly (I:C), grass carp reovirus (GCRV) and spring viremia of carp virus (SVCV). bcRNF5 migrates around 22 KDa in immunoblot (IB) assay and distributes mainly in cytoplasm by immunofluorescent (IF) staining test. Moreover, bcRNF5 significantly inhibits bcMAVS-mediated IFN promoter transcription. In addition, both IF and co-immunoprecipitation assay showed that bcRNF5 interacts with bcMAVS. Furthermore, bcMAVS-mediated antiviral ability is distinctly impaired by bcRNF5. Taken together, these results conclude that bcRNF5, as a negative regulator of the MAVS-mediated IFN signaling, may play a key role in host protection upon virus infection in black carp.
    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
    许多人类疾病是由核膜(NE)蛋白的突变引起的。蛋白质稳态和疾病病因在NE上如何相互联系的了解很少。具体来说,目前尚不清楚促进泛素-蛋白酶体依赖性NE蛋白周转的局部泛素连接酶。这里,我们雇佣了一个短暂的,LaminB受体疾病变体作为遗传筛选中的模型底物,以揭示NE蛋白周转的关键要素。我们鉴定了泛素缀合酶(E2s)Ube2G2和Ube2D3,膜驻留的泛素连接酶(E3s)RNF5和HRD1以及对蛋白质TMEM33了解甚少。RNF5,而不是HRD1,需要TMEM33来实现有效的生物合成和功能。一旦合成,RNF5通过离开内质网动态响应NE处增加的底物水平,HRD1仍然受到限制。因此,哺乳动物蛋白质质量控制机制在不同的细胞区室之间划分,以解决局部变化的底物负荷,建立一个强大的细胞质量控制系统。
    Many human diseases are caused by mutations in nuclear envelope (NE) proteins. How protein homeostasis and disease etiology are interconnected at the NE is poorly understood. Specifically, the identity of local ubiquitin ligases that facilitate ubiquitin-proteasome-dependent NE protein turnover is presently unknown. Here, we employ a short-lived, Lamin B receptor disease variant as a model substrate in a genetic screen to uncover key elements of NE protein turnover. We identify the ubiquitin-conjugating enzymes (E2s) Ube2G2 and Ube2D3, the membrane-resident ubiquitin ligases (E3s) RNF5 and HRD1, and the poorly understood protein TMEM33. RNF5, but not HRD1, requires TMEM33 both for efficient biosynthesis and function. Once synthesized, RNF5 responds dynamically to increased substrate levels at the NE by departing from the endoplasmic reticulum, where HRD1 remains confined. Thus, mammalian protein quality control machinery partitions between distinct cellular compartments to address locally changing substrate loads, establishing a robust cellular quality control 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
    单纯疱疹性角膜炎(HSK),由单纯疱疹病毒1(HSV-1)引起,是发达国家的主要致盲疾病。HSV-1可以在宿主中终生潜伏并且不能被根除。感染引起各种细胞因子的分泌和炎性细胞的聚集。在炎症的早期阶段,主要是嗜中性粒细胞浸润角膜,CD4T细胞在随后的进展中介导疱疹性基质角膜炎的免疫病理学变化。STING/IRF3介导的I型干扰素(IFN)反应可以有效抑制病毒复制和控制感染,但是STING的活性受到各种泛素化修饰的影响。在这项研究中,我们发现感染HSV-1后,RNF5在角膜组织和角膜上皮细胞中的表达升高。免疫荧光染色证实RNF5主要表达于角膜上皮层。我们在角膜上皮细胞中沉默并过表达RNF5,然后用HSV-1接种它们。我们发现STING的表达,RNF5沉默后p-IRF3、p-TBK1和IFN-βmRNA增加。在RNF5过表达后获得相反的结果。我们还使用siRNA沉默小鼠角膜中的RNF5,然后建立HSK模型。与siRNA对照组相比,siRNA-RNF5组显着改善角膜炎症,降低临床评分和泪液病毒滴度,并显著增加角膜IFN-β表达。此外,促炎细胞因子IL-6和TNF-α在角膜组织中的表达显著降低,表明RNF5沉默可以有效促进IFN-I表达,抑制病毒复制,缓解炎症,减少角膜炎症损伤。总之,我们的结果表明,RNF5通过抑制STING/IRF3信号传导限制了HSV角膜上皮炎的I型IFN抗病毒应答.
    Herpes simplex keratitis (HSK), caused by the herpes simplex virus 1 (HSV-1), is a major blinding disease in developed countries. HSV-1 can remain latent in the host for life and cannot be eradicated. The infection causes the secretion of various cytokines and aggregation of inflammatory cells. In the early stage of inflammation, mainly neutrophils infiltrate the cornea, and CD4+ T cells mediate the immunopathological changes in herpetic stromal keratitis in the subsequent progression. The STING/IRF3-mediated type I interferon (IFN) response can effectively inhibit viral replication and control infection, but the activity of STING is affected by various ubiquitination modifications. In this study, we found that the expression of RNF5 was elevated in corneal tissues and corneal epithelial cells after infection with HSV-1. Immunofluorescence staining confirmed that RNF5 was mainly expressed in the corneal epithelial layer. We silenced and overexpressed RNF5 expression in corneal epithelial cells and then inoculated them with HSV-1. We found that the expressions of STING, p-IRF3, p-TBK1, and IFN-β mRNA increased after RNF5 silencing. The opposite results were obtained after RNF5 overexpression. We also used siRNA to silence RNF5 in the mouse cornea and then established the HSK model. Compared with the siRNA-control group, the siRNA-RNF5 group showed significantly improved corneal inflammation, reduced clinical scores and tear virus titers, and significantly increased corneal IFN-β expression. In addition, the expressions of the proinflammatory cytokines IL-6 and TNF-α in the corneal tissue were significantly decreased, indicating that RNF5 silencing could effectively promote IFN-I expression, inhibit virus replication, alleviate inflammation, and reduce corneal inflammatory damage. In summary, our results suggest that RNF5 limits the type I IFN antiviral response in HSV corneal epithelitis by inhibiting STING/IRF3 signaling.
    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
    RNF5,一种内质网(ER)E3泛素连接酶,参与ER相关蛋白质降解,保证蛋白质稳态。根据测试的肿瘤模型,RNF5发挥促肿瘤或抗肿瘤活性。本研究的目的是阐明RNF5在神经母细胞瘤和黑色素瘤中的有争议的作用。婴儿期和成年期的两种神经外胚层肿瘤,分别。在公开可用的数据集中评估RNF5基因水平,报告诊断时黑色素瘤和神经母细胞瘤原发性肿瘤的基因表达谱。RNF5药物激活剂Analog-1的治疗效果,在体外和体内神经母细胞瘤和黑色素瘤模型上进行了研究。在神经母细胞瘤和黑色素瘤患者中,RNF5的高表达与更好的预后结果相关。用类似物1处理神经母细胞瘤和黑素瘤细胞系通过抑制F1FoATP合酶活性而损害谷氨酰胺的可用性和能量代谢,从而降低了细胞活力。后一个事件导致氧化应激的显著增加,which,反过来,导致细胞死亡。同样,与仅用媒介物治疗的小鼠相比,用类似物-1治疗的携带神经母细胞瘤和黑色素瘤的小鼠显示出肿瘤生长的显著延迟。这些发现验证了RNF5作为创新药物靶标,并支持神经母细胞瘤和黑色素瘤患者早期临床试验中类似物1的开发。
    RNF5, an endoplasmic reticulum (ER) E3 ubiquitin ligase, participates to the ER-associated protein degradation guaranteeing the protein homeostasis. Depending on tumor model tested, RNF5 exerts pro- or anti-tumor activity. The aim of this study was to elucidate the controversial role of RNF5 in neuroblastoma and melanoma, two neuroectodermal tumors of infancy and adulthood, respectively. RNF5 gene levels are evaluated in publicly available datasets reporting the gene expression profile of melanoma and neuroblastoma primary tumors at diagnosis. The therapeutic effect of Analog-1, an RNF5 pharmacological activator, was investigated on in vitro and in vivo neuroblastoma and melanoma models. In both neuroblastoma and melanoma patients the high expression of RNF5 correlated with a better prognostic outcome. Treatment of neuroblastoma and melanoma cell lines with Analog-1 reduced cell viability by impairing the glutamine availability and energy metabolism through inhibition of F1Fo ATP-synthase activity. This latter event led to a marked increase in oxidative stress, which, in turn, caused cell death. Similarly, neuroblastoma- and melanoma-bearing mice treated with Analog-1 showed a significant delay of tumor growth in comparison to those treated with vehicle only. These findings validate RNF5 as an innovative drug target and support the development of Analog-1 in early phase clinical trials for neuroblastoma and melanoma patients.
    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
    作为一种包膜病毒,严重急性呼吸综合征冠状病毒2(SARS-CoV-2)含有介导病毒从细胞膜释放的膜蛋白(M)。然而,SARS-CoV-2病毒体释放的分子机制仍然知之甚少。在本研究中,我们进行了RNA干扰(RNAi)筛选,并鉴定了E3连接酶RNF5,它介导SARS-CoV-2M在残基K15的泛素化,以增强病毒包膜蛋白(E)与M的相互作用,而去泛素化酶POH1负调控这一过程。M-E复合物确保病毒成熟的病毒颗粒的均匀尺寸并介导病毒体释放。此外,M从高尔基体运输到自噬体,并使用自噬体释放病毒体,该过程依赖于RNF5介导的泛素修饰和M-E相互作用。这些结果表明,SARS-CoV-2M的泛素修饰可稳定M-E复合物,并使用自噬体释放病毒体。重要性包膜病毒颗粒从宿主细胞膜上释放出来,和病毒膜蛋白(M)是这个过程的关键。更好地了解SARS-CoV-2组装和出芽的分子机制对于开发抗病毒疗法至关重要。SARS-CoV-2的包膜蛋白(E)和M形成复合物以介导病毒组装和出芽。RNF5被鉴定为作为E3连接酶发挥作用,POH1被证明是SARS-CoV-2M的去泛素化酶。这两个成分共同调节M与E的相互作用,以促进病毒的组装和出芽。泛素化的M使用自噬体进行病毒释放。我们的发现提供了对SARS-CoV-2组装和萌芽机制的见解,证明泛素化修饰和自噬在病毒复制中的重要性。
    As an enveloped virus, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) contains a membrane protein (M) that mediates viral release from cellular membranes. However, the molecular mechanisms of SARS-CoV-2 virion release remain poorly understood. In the present study, we performed RNA interference (RNAi) screening and identified the E3 ligase RNF5, which mediates the ubiquitination of SARS-CoV-2 M at residue K15 to enhance the interaction of the viral envelope protein (E) with M, whereas the deubiquitinating enzyme POH1 negatively regulates this process. The M-E complex ensures the uniform size of viral particles for viral maturation and mediates virion release. Moreover, M traffics from the Golgi apparatus to autophagosomes and uses autophagosomes for virion release, and this process is dependent on RNF5-mediated ubiquitin modification and M-E interaction. These results demonstrate that ubiquitin modification of SARS-CoV-2 M stabilizes the M-E complex and uses autophagosomes for virion release. IMPORTANCE Enveloped virus particles are released from the membranes of host cells, and viral membrane proteins (M) are critical for this process. A better understanding of the molecular mechanisms of SARS-CoV-2 assembly and budding is critical for the development of antiviral therapies. Envelope protein (E) and M of SARS-CoV-2 form complexes to mediate viral assembly and budding. RNF5 was identified to play a role as the E3 ligase, and POH1 was demonstrated to function as the deubiquitinating enzyme of SARS-CoV-2 M. The two components collectively regulate the interaction of M with E to promote viral assembly and budding. Ubiquitinated M uses autophagosomes for viral release. Our findings provide insights into the mechanisms of SARS-CoV-2 assembly and budding, demonstrating the importance of ubiquitination modification and autophagy in viral replication.
    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