TRIM5α

TRIM5 α
  • 文章类型: Journal Article
    病毒感染所施加的进化压力导致了具有有效抗病毒活性的各种细胞蛋白的发展。其中一些被称为抗病毒限制因子。含TRIpartnal基序的蛋白5α(TRIM5α)是逆转录病毒的经过充分研究的限制因子,在保护特定慢病毒的跨灵长类动物传播方面表现出病毒和宿主物种特异性功能。这种特异性是通过主要在其C端B30.2/PRYSPRY结构域内的阳性选择在宿主基因水平上实现的,负责逆转录病毒衣壳的高度特异性识别。然而,最近的工作挑战了这种范式,证明TRIM5α是逆转录元件以及系统发育上不同病毒家族的限制因子,通过B30.2/PRYSPRY识别病毒基因产物起类似作用。该抗病毒活性谱提出了关于该蛋白质的遗传和结构可塑性作为识别潜在的多种病毒分子模式的介体的问题。这篇综述强调了B30.2/PRYSPRY域对逆转录病毒的动态进化足迹,同时探索了TRIM5α的全部其他域所赋予的指导“特异性”,这可能是其最近发现的滥交。
    The evolutionary pressures exerted by viral infections have led to the development of various cellular proteins with potent antiviral activities, some of which are known as antiviral restriction factors. TRIpartite Motif-containing protein 5 alpha (TRIM5α) is a well-studied restriction factor of retroviruses that exhibits virus- and host-species-specific functions in protecting against cross-primate transmission of specific lentiviruses. This specificity is achieved at the level of the host gene through positive selection predominantly within its C-terminal B30.2/PRYSPRY domain, which is responsible for the highly specific recognition of retroviral capsids. However, more recent work has challenged this paradigm, demonstrating TRIM5α as a restriction factor for retroelements as well as phylogenetically distinct viral families, acting similarly through the recognition of viral gene products via B30.2/PRYSPRY. This spectrum of antiviral activity raises questions regarding the genetic and structural plasticity of this protein as a mediator of the recognition of a potentially diverse array of viral molecular patterns. This review highlights the dynamic evolutionary footprint of the B30.2/PRYSPRY domain in response to retroviruses while exploring the guided \'specificity\' conferred by the totality of TRIM5α\'s additional domains that may account for its recently identified promiscuity.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Preprint
    线粒体蛋白质的泛素化为线粒体自噬机制的下游募集提供了基础,然而,机器本身的泛素化是否有助于线粒体自噬尚不清楚。这里,我们表明,关键的线粒体自噬调节因子TBK1的K63连接的聚泛素化对其线粒体自噬功能至关重要。这种修饰由泛素连接酶TRIM5α催化。线粒体损伤触发TRIM5α的自动泛素化及其与泛素结合自噬衔接子的相互作用,包括NDP52,视神经磷酸酶,和NBR1。自噬适配器,与TRIM27一起,使TRIM5α与TBK1接合。具有完整泛素化功能的TRIM5α对于在Parkin依赖性和Parkin非依赖性线粒体自噬途径中受损线粒体上的活性TBK1的适当积累是必需的。此外,我们表明TRIM5α可以直接招募受损线粒体的自噬启动机制。我们的数据支持一个模型,其中TRIM5α提供了自放大功能,线粒体定位,泛素为基础,TBK1和线粒体自噬适配器的组装平台,最终需要招募核心自噬机制。
    Ubiquitination of mitochondrial proteins provides a basis for the downstream recruitment of mitophagy machinery, yet whether ubiquitination of the machinery itself contributes to mitophagy is unknown. Here, we show that K63-linked polyubiquitination of the key mitophagy regulator TBK1 is essential for its mitophagy functions. This modification is catalyzed by the ubiquitin ligase TRIM5α. Mitochondrial damage triggers TRIM5α\'s auto-ubiquitination and its interaction with ubiquitin-binding autophagy adaptors including NDP52, optineurin, and NBR1. Autophagy adaptors, along with TRIM27, enable TRIM5α to engage with TBK1. TRIM5α with intact ubiquitination function is required for the proper accumulation of active TBK1 on damaged mitochondria in Parkin-dependent and Parkin-independent mitophagy pathways. Additionally, we show that TRIM5α can directly recruit autophagy initiation machinery to damaged mitochondria. Our data support a model in which TRIM5α provides a self-amplifying, mitochondria-localized, ubiquitin-based, assembly platform for TBK1 and mitophagy adaptors that is ultimately required to recruit the core autophagy 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
    细胞内限制因子TRIM5α抑制内源性LINE-1逆转录元件。它在感知细胞质LINE-1复合物时诱导先天免疫信号级联,从而强调其对于保护人类基因组免受有害的反转录转座事件的重要性。这里,我们表明,TRIM5α的RING域内存在频繁的SNP,导致变体H43Y,与TRIM5αWT相比,以更高的效率阻断LINE-1逆行转位。在细胞质中检测LINE-1复合物时,TRIM5αH43Y比TRIM5αWT更有效地激活NF-κB和AP-1信号通路,触发LINE-1启动子的强阻断。有趣的是,H43Y等位基因失去其抗病毒功能,提示其对内源性LINE-1元件的增强活性是其在群体中维持的驱动力.因此,我们的研究表明,限制因子和传感器TRIM5α的H43Y变体在人群中持续存在,因为它以更高的效率保护了我们的基因组免受不受控制的LINE-1反转座。
    The intracellular restriction factor TRIM5α inhibits endogenous LINE-1 retroelements. It induces innate immune signaling cascades upon sensing of cytoplasmic LINE-1 complexes, thereby underlining its importance for protecting the human genome from harmful retrotransposition events. Here, we show that a frequent SNP within the RING domain of TRIM5α, resulting in the variant H43Y, blocks LINE-1 retrotransposition with higher efficiency compared to TRIM5α WT. Upon sensing of LINE-1 complexes in the cytoplasm, TRIM5α H43Y activates both NF-κB and AP-1 signaling pathways more potently than TRIM5α WT, triggering a strong block of the LINE-1 promoter. Interestingly, the H43Y allele lost its antiviral function suggesting that its enhanced activity against endogenous LINE-1 elements is the driving force behind its maintenance within the population. Thus, our study suggests that the H43Y variant of the restriction factor and sensor TRIM5α persists within the human population since it preserves our genome from uncontrolled LINE-1 retrotransposition with higher efficiency.
    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
    TRIM5α是一种宿主抗逆转录病毒限制因子,可破坏人类免疫缺陷病毒(HIV)病毒体并触发先天免疫信号。TRIM5α还通过TRIMosome形成介导靶蛋白的自噬降解。我们先前表明TRIM5α促进EB病毒(EBV)Rta泛素化并减弱EBV裂解进程。在这项研究中,我们试图阐明TRIM5α是否可以与EBV衣壳蛋白相互作用并诱导其降解。进行谷胱甘肽S-转移酶(GST)下拉和免疫沉淀测定以鉴定相互作用的蛋白质,并产生突变体以研究关键结合结构域和泛素化位点。结果表明,TRIM5α与BORF1直接结合,BORF1是一种EBV衣壳蛋白,具有核定位信号(NLS),能够将EBV衣壳蛋白转运到宿主核中以促进衣壳组装。TRIM5α促进BORF1泛素化,这需要TRIM5αPRY/SPRY域中的表面斑块区域。TRIM5α的表达也降低了BORF1(6KR)的稳定性,所有赖氨酸残基都突变成精氨酸的突变体。然而,氯喹处理恢复了BORF1(6KR)的稳定性,表明TRIM5α通过直接识别其底物进行自噬降解而使BORF1不稳定。这些结果揭示了TRIM5α超越逆转录病毒的抗病毒影响的新见解。
    TRIM5α is a host anti-retroviral restriction factor that destroys human immunodeficiency virus (HIV) virions and triggers innate immune signaling. TRIM5α also mediates the autophagic degradation of target proteins via TRIMosome formation. We previously showed that TRIM5α promotes Epstein-Barr virus (EBV) Rta ubiquitination and attenuates EBV lytic progression. In this study, we sought to elucidate whether TRIM5α can interact with and induce the degradation of EBV capsid proteins. Glutathione S-transferase (GST) pulldown and immunoprecipitation assays were conducted to identify interacting proteins, and mutants were generated to investigate key binding domains and ubiquitination sites. Results showed that TRIM5α binds directly with BORF1, an EBV capsid protein with a nuclear localization signal (NLS) that enables the transport of EBV capsid proteins into the host nucleus to facilitate capsid assembly. TRIM5α promotes BORF1 ubiquitination, which requires the surface patch region in the TRIM5α PRY/SPRY domain. TRIM5α expression also decreases the stability of BORF1(6KR), a mutant with all lysine residues mutated to arginine. However, chloroquine treatment restores the stability of BORF1(6KR), suggesting that TRIM5α destabilizes BORF1 via direct recognition of its substrate for autophagic degradation. These results reveal novel insights into the antiviral impact of TRIM5α beyond retroviruses.
    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
    蛋白质TRIM5正在深入研究,与其在抗病毒防御中的作用有关,然而,其潜在的作用机制仍然难以捉摸。在我们的研究中,我们对TRIM5相互作用的配偶体进行了无偏倚的鉴定,并发现了参与多种细胞功能的蛋白质.我们利用这个蛋白质组学数据集来揭示TRIM5在线粒体自噬中的作用,在多种人类疾病中受损的线粒体质量控制系统。线粒体损伤触发TRIM5募集至ER-线粒体接触位点,其中TRIM5与自噬体生物发生的标记物共同定位。缺乏TRIM5的细胞不能进行PRKN依赖性和PRKN非依赖性线粒体自噬途径。TRIM5敲除细胞显示线粒体功能降低和响应线粒体损伤的不受控制的免疫激活;表型与线粒体自噬中TRIM5的要求一致。机械上,我们发现TRIM5是自噬启动机制募集到受损线粒体所必需的,其中TRIM5充当支架,促进线粒体损伤的蛋白质标记与自噬起始机制之间的相互作用。
    The protein TRIM5 is under intensive investigation related to its roles in antiviral defense, yet its underlying mechanisms of action remain elusive. In our study, we performed an unbiased identification of TRIM5-interacting partners and found proteins participating in a wide variety of cellular functions. We utilized this proteomics data set to uncover a role for TRIM5 in mitophagy, a mitochondrial quality control system that is impaired in multiple human diseases. Mitochondrial damage triggers the recruitment of TRIM5 to ER-mitochondria contact sites where TRIM5 colocalizes with markers of autophagosome biogenesis. Cells lacking TRIM5 are unable to carry out PRKN-dependent and PRKN-independent mitophagy pathways. TRIM5 knockout cells show reduced mitochondrial function and uncontrolled immune activation in response to mitochondrial damage; phenotypes consistent with a requirement for TRIM5 in mitophagy. Mechanistically, we found that TRIM5 is required for the recruitment of the autophagy initiation machinery to damaged mitochondria, where TRIM5 acts as a scaffold promoting interactions between protein markers of mitochondrial damage and the autophagy initiation 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
    TRIM5α蛋白在抗逆转录病毒防御中具有多种作用,但TRIM5α作用的潜在机制尚不清楚。这里,我们使用基于APEX2的蛋白质组学来鉴定TRIM5α相互作用的配偶体。我们的蛋白质组学结果将TRIM5与其他具有抗病毒防御作用的蛋白质联系起来。此外,它们将TRIM5与线粒体自噬联系起来,一种基于自噬的线粒体质量控制模式,在几种人类疾病中受损。我们发现TRIM5是Parkin依赖性和非依赖性线粒体自噬途径所必需的,其中TRIM5向受损的线粒体募集上游自噬调节因子。缺乏泛素连接酶活性的TRIM5突变体的表达无法挽救TRIM5敲除细胞中的线粒体自噬。缺乏TRIM5的细胞在基础条件下显示降低的线粒体功能,并且比野生型细胞更容易受到免疫激活和响应线粒体损伤的死亡。一起来看,我们的研究确定了一种蛋白质的稳态作用,这种蛋白质以前完全被认为是抗病毒作用。
    The protein TRIM5α has multiple roles in antiretroviral defense, but the mechanisms underlying TRIM5α action are unclear. Here, we employ APEX2-based proteomics to identify TRIM5α-interacting partners. Our proteomics results connect TRIM5 to other proteins with actions in antiviral defense. Additionally, they link TRIM5 to mitophagy, an autophagy-based mode of mitochondrial quality control that is compromised in several human diseases. We find that TRIM5 is required for Parkin-dependent and -independent mitophagy pathways where TRIM5 recruits upstream autophagy regulators to damaged mitochondria. Expression of a TRIM5 mutant lacking ubiquitin ligase activity is unable to rescue mitophagy in TRIM5 knockout cells. Cells lacking TRIM5 show reduced mitochondrial function under basal conditions and are more susceptible to immune activation and death in response to mitochondrial damage than are wild-type cells. Taken together, our studies identify a homeostatic role for a protein previously recognized exclusively for its antiviral actions.
    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
    恒河猴(RM)中的TRIM5α多态性限制了动物的遗传库,我们可以在其中进行猿猴免疫缺陷病毒(SIV)研究,而无需首先对动物进行允许的TRIM5α基因型筛选。我们先前已经表明,TRIM5αB30.2/SPRY结构域的多态性会影响RM中SIVsmm病毒血症的水平,并且衣壳N末端结构域(CA-NTD)中的氨基酸取代(P37S/R98S)使病毒能够克服具有限制性纯合TRIM5αP/TFP基因型的RM中的限制。由于这种基因型也对感染CL757亲本来源的动物的中枢神经系统(CNS)病变的发展产生负面影响,因此我们试图生成TRIM5αTFP/TFP抗性克隆,SIV-804E-CL757-P37S/R98S(CL757-SS),使用类似的策略。出乎意料的是,CL757-SS的病毒复制在允许的TRIM5αTFP/Q或限制性TRIM5αTFP/TFP基因型的RM中受损.对在后一种动物中出现的病毒的分析导致发现了相对于其他SIV的预先存在的突变。在衣壳的C-末端结构域(CA-CTD)中的保守无序接头区域中的这种P146T取代已显示抑制HIV-1衣壳颗粒的适当形成。在TRIM5α-SS逃逸突变的情况下,将该残基恢复为脯氨酸不仅恢复了病毒复制,而且还增强了我们先前报道的嗜神经克隆的感染性,即使在具有允许的TRIM5α基因型的RM中。重要性恒河猴SIV感染已成为开发AIDS疫苗和抗逆转录病毒疗法的有价值的模型。恒河猴TRIM5α基因多态性可影响SIV复制,使得有必要对允许SIV复制的TRIM5α等位基因进行遗传筛选。这限制了可用于研究的动物库,从而使得获取满足研究参数所需的动物变得困难。我们已经构建了一个病毒克隆,在恒河猴中诱导神经艾滋病,无论其TRIM5α基因型如何,同时也强调了无序接头结构域在病毒感染性中的重要作用。
    TRIM5α polymorphism in rhesus macaques (RM) limits the genetic pool of animals in which we can perform simian immunodeficiency virus (SIV) studies without first screening animals for permissive TRIM5α genotypes. We have previously shown that polymorphisms in the TRIM5α B30.2/SPRY domain impact the level of SIVsmm viremia in RM and that amino acid substitutions (P37S/R98S) in the capsid N-terminal domain (CA-NTD) enables the virus to overcome restriction in RMs with the restrictive homozygous TRIM5αTFP/TFP genotype. Since this genotype also negatively impacted the development of central nervous system (CNS) lesions in animals infected with the parental source of CL757, we sought to generate a TRIM5αTFP/TFP-resistant clone, SIV-804E-CL757-P37S/R98S (CL757-SS), using a similar strategy. Unexpectedly, viral replication of CL757-SS was impaired in RMs with either the permissive TRIM5αTFP/Q or the restrictive TRIM5αTFP/TFP genotype. Analysis of the virus which emerged in the latter animals led to the discovery of a preexisting mutation relative to other SIVs. This P146T substitution in a conserved disordered linker region in the C-terminal domain of capsid (CA-CTD) has been shown to inhibit proper formation of HIV-1 capsid particles. Restoration of this residue to proline in the context of the TRIM5α-SS escape mutations not only restored viral replication, but also enhanced the infectivity of our previously reported neurotropic clone, even in RMs with permissive TRIM5α genotypes. IMPORTANCE SIV infection of rhesus macaques has become a valuable model for the development of AIDS vaccines and antiretroviral therapies. Polymorphisms in the rhesus macaque TRIM5α gene can affect SIV replication, making it necessary to genetically screen macaques for TRIM5α alleles that are permissive for SIV replication. This limits the pool of animals that can be used in a study, thereby making the acquisition of animals needed to fulfill study parameters difficult. We have constructed a viral clone that induces neuroAIDS in rhesus macaques regardless of their TRIM5α genotype, while also highlighting the important role the disordered linker domain plays in viral infectivity.
    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
    嵌合蛋白TRIM5α-HRH是HIV-1基因治疗的一种有前途的抗病毒因子。这种蛋白质能够通过阻断细胞质中的病毒来保护细胞免受HIV-1的侵害。我们正在开发HIV-1基因治疗方案,涉及通过慢病毒载体(LV)将TRIM5α-HRH基因传递到CD4^()T淋巴细胞中。然而,含有TRIM5α-HRH的LV具有低感染滴度,这阻止了有效的T细胞修饰。这里,我们发现TRIM5α-HRH在HEK293T细胞产生假病毒颗粒过程中的表达,以及在我们的构建中Eflα启动子的存在是滴度降低的原因。这些结果使我们能够确定进一步优化具有TRIM5α-HRH基因的LV以提高其感染滴度的方向。
    The chimeric protein TRIM5α-HRH is a promising antiviral factor for HIV-1 gene therapy. This protein is able to protect cells from HIV-1 by blocking the virus in the cytoplasm. We are developing protocol of HIV-1 gene therapy, which involves the delivery of the TRIM5α-HRH gene into CD4^(+) T-lymphocytes by lentiviral vectors (LVs). However, LVs containing TRIM5α-HRH have a low infectious titer, which prevents effective T cell modification. Here, we found that the expression of TRIM5α-HRH during pseudoviral particle production in HEK293 T cells, as well as the presence of the Eflα promoter in our construction are responsible for titer reduction. These results allow us to determine the directions for further optimization of LV with the TRIM5α-HRH gene to improve its infectious titer.
    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
    AIDS限制性基因(ARGs)如APOBEC3、TRIM5α、和BST2可以作为机体先天保护机制的免疫学检测器。ARGs影响病毒发病过程和疾病进展。由包括HIV在内的不同病毒引起的感染激活先天免疫受体,导致促炎细胞因子的产生,在诱导适应性免疫后募集和激活参与炎症过程的细胞的干扰素和信号。与病毒感染有关的基因的差异表达决定了感染的命运和随后的易感性及其临床结果。然而,关于APOBEC3基因多态性发病率的综合报道,TRIM5α,一般人群中的BST-2及其与病理状况的关系尚未得到很好的描述。因此,APOBEC3,TRIM5α,分析了健康个体的BST2多态性及其对HIV传播的影响。我们使用几个数据库进行了广泛的搜索,包括,EMBASE,PubMed(Medline),谷歌学者。APOBEC3-D,-F,-G,和-H在七个人类APOBEC3中,帮助控制病毒感染。在各种制约因素中,TRIM5α和BST-2也限制了病毒感染,然后是疾病的发展。在当前的审查中,简要介绍了APOBEC3G中的多态性,TRIM5α,以及APOBEC3G与Vif蛋白的相互作用,在不同人群中探索BST2基因。此外,这篇综述特别关注ARGs多态性(APOBEC3G,TRIM5α,和BST2)与HIV传播有关。
    AIDS restriction genes (ARGs) like APOBEC3, TRIM5α, and BST2 can act as immunological detectors of the innate protective mechanism of the body. ARGs influence the course of viral pathogenesis and progression of the disease. The infection caused by different viruses including HIV activates the innate immune receptors leading to production of proinflammatory cytokines, interferons and signals that recruit and activate cells involved in the process of inflammation following induction of adaptive immunity. Differential expression of genes involved in viral infection decide the fate and subsequent susceptibility to infection and its clinical outcome. Nevertheless, comprehensive reports on the incidence of genetic polymorphism of APOBEC3s, TRIM5α, and BST-2 in the general population and its association with pathological conditions have not been described well. Therefore, the occurrence of APOBEC3, TRIM5α, and BST2 polymorphism in healthy individuals and its impact on HIV transmission was analyzed. We conducted an extensive search using the several databases including, EMBASE, PubMed (Medline), and Google Scholar. APOBEC3-D, -F, -G, and -H out of the seven human APOBEC3s, help in the control of viral infection. Amongst various restriction factors, TRIM5α and BST-2 also restrict the viral infection followed by the development of the disease. In the current review, a brief account of the polymorphism in the APOBEC3G, TRIM5α, and BST2 genes are explored among different populations along with the interaction of APOBEC3G with Vif protein. Furthermore, this review specifically focus on ARGs polymorphism (APOBEC3G, TRIM5α, and BST2) associated with HIV transmission.
    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: English Abstract
    During their evolution, mammals have developed cellular factors interfering with retroviral replication, known as « restriction factors ». The prototype of these factors, Fv1, was characterized in the late 60\'s and blocks MLV infection. Some Fv1-like factors interfering with complex retroviruses, including HIV, have recently been discovered in primate cells. These restriction factors are referred to as Ref1, which blocksMLVreplication in human cells, and Lv1, which blocks the infection of non-human primate cells by various retroviruses, including MLV and HIV. These factors are all saturable by an excess of virus, target the viral capsid and interfere with an early step of viral replication. Lv1 and Ref1 have recently been found to be species-specific variants of a single protein called TRIM5α, a member of the TRIM protein family. The mechanism of action of these factors is still unknown. The existence of natural inhibitors of retroviral infection raises new hopes for the development of therapeutic tools against HIV infection.
    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