Neuropathogenesis

神经发病机制
  • 文章类型: Journal Article
    寨卡病毒(ZIKV),蚊子传播的黄病毒,与受感染母亲所生的婴儿的小头畸形以及成人的格林-巴利综合症密切相关。ZIKV神经元细胞感染的每种细胞类型(放射状神经胶质细胞,神经元祖细胞,星形胶质细胞,小胶质细胞,和胶质母细胞瘤干细胞)和非神经元细胞(原代成纤维细胞,表皮角质形成细胞,树突状细胞,单核细胞,巨噬细胞,和支持细胞)-对其细胞生理学表现出自己的特征性变化,并对疾病产生各种影响。这里,我们对ZIKV生命周期及其细胞靶标进行了深入的回顾,并讨论感染如何引起神经病理学的当前知识,以及研究人员目前正在采取什么方法来进一步推进这些知识。ZIKV神经发病机制的一个关键方面是病毒通过多种机制诱导的神经元凋亡,包括细胞周期失调,线粒体片段化,ER压力,和展开的蛋白质反应。这些,反过来,导致p53介导的内在细胞死亡途径的激活。包括干细胞和共培养在内的全谱感染模型,Transwells模拟血液组织屏障,大脑区域特异性类器官,并为ZIKV研究开发了动物模型。
    Zika virus (ZIKV), a mosquito-borne flavivirus, is prominently associated with microcephaly in babies born to infected mothers as well as Guillain-Barré Syndrome in adults. Each cell type infected by ZIKV-neuronal cells (radial glial cells, neuronal progenitor cells, astrocytes, microglia cells, and glioblastoma stem cells) and non-neuronal cells (primary fibroblasts, epidermal keratinocytes, dendritic cells, monocytes, macrophages, and Sertoli cells)-displays its own characteristic changes to their cell physiology and has various impacts on disease. Here, we provide an in-depth review of the ZIKV life cycle and its cellular targets, and discuss the current knowledge of how infections cause neuropathologies, as well as what approaches researchers are currently taking to further advance such knowledge. A key aspect of ZIKV neuropathogenesis is virus-induced neuronal apoptosis via numerous mechanisms including cell cycle dysregulation, mitochondrial fragmentation, ER stress, and the unfolded protein response. These, in turn, result in the activation of p53-mediated intrinsic cell death pathways. A full spectrum of infection models including stem cells and co-cultures, transwells to simulate blood-tissue barriers, brain-region-specific organoids, and animal models have been developed for ZIKV research.
    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
    神经系统并发症,急性和慢性,在受COVID-19影响的个体中普遍报告。在这种情况下,对SARS-CoV-2在中枢神经系统(CNS)起源的特定细胞中的发病机理的理解是相关的。本研究探讨了SARS-CoV-2临床分离株在神经起源的人类细胞系中的感染生物学,例如胶质母细胞瘤(U87-MG)。神经母细胞瘤(SHSY5Y)和小胶质细胞(C20)。尽管通过抗刺突蛋白抗体的免疫荧光显示出明显的感染证据,观察到所有三种神经细胞系对感染病毒的复制具有高度限制性。虽然U87-MG胶质母细胞瘤细胞没有细胞病变效应和低病毒滴度,没有复制的迹象,SHSY5Y神经母细胞瘤细胞表现出细胞病变作用,并伴有气泡形成,但没有活病毒的证据.C20小胶质细胞既没有显示细胞病变作用的迹象,也没有活的病毒。超微结构研究表明,受感染的神经细胞中存在细胞内病毒体。在感染的SHSY5Y细胞中存在脂滴表明对宿主细胞代谢的影响。所有神经细胞系中病毒RNA水平随时间的降低表明病毒复制受限。总之,这项研究强调了神经细胞对SARS-CoV-2感染的有限易感性.神经细胞系对SARS-CoV-2的可容许性降低可能表明,除了有效抑制病毒复制的细胞内因子外,它们固有的支持病毒进入的受体表达较低。该研究结果促使进一步研究SARS-CoV-2感染神经细胞的机制。
    Neurological complications, both acute and chronic, are reported commonly in COVID-19 affected individuals. In this context, the understanding of pathogenesis of SARS-CoV-2 in specific cells of central nervous system (CNS) origin is relevant. The present study explores infection biology of a clinical isolate of SARS-CoV-2 in human cell lines of neural origin such as the glioblastoma (U87-MG), neuroblastoma (SHSY5Y) and microglia (C20). Despite showing clear evidence of infection by immunofluorescence with an anti-spike protein antibody, all the three neural cell lines were observed to be highly restrictive to the replication of the infecting virus. While the U87-MG glioblastoma cells demonstrated no cytopathic effects and a low viral titre with no signs of replication, the SHSY5Y neuroblastoma cells exhibited cytopathic effects with bleb formation but no evidence of viable virus. The C20 microglial cells showed neither signs of cytopathic effects nor viable virus. Ultrastructural studies demonstrated intracellular virions in infected neural cells. The presence of lipid droplets in infected SHSY5Y cells suggested an impact on host cell metabolism. The decrease in viral RNA levels over time in all the neural cell lines suggested restricted viral replication. In conclusion, this study highlights the limited susceptibility of neural cells to SARS-CoV-2 infection. This reduced permissibility of neural cell lines to SARS-CoV-2 may point to their inherent lower expression of receptors that support viral entry in addition to the intracellular factors that potently inhibit viral replication. The study findings prompt further investigation into the mechanisms of SARS-CoV-2 infection of neural cells.
    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
    蜱传黄病毒(TBFV)可引起严重的神经侵袭性疾病,可能导致超过50%的幸存者死亡或长期神经功能缺损。已经提出了黄病毒侵袭中枢神经系统(CNS)的多种机制,包括轴突运输,胞吞,内皮感染,和特洛伊木马路线。黄病毒可能利用不同或多种神经侵袭机制,这取决于特定的病毒,感染部位,和宿主的变异性。在这项工作中,我们已经表明,BALB/cJ小鼠感染Powassan病毒谱系I(Powassan病毒)或谱系II(鹿tick病毒)导致CNS感染的不同空间嗜性,这与每个谱系的独特临床表现相关。受感染大脑的比较转录组学证明了不同免疫途径和下游宿主反应的激活。最终,比较病理学和转录组学与小鼠模型中的不同临床体征一致。这些结果表明,由于Powassan病毒的两个谱系的固有差异,临床病例中出现了不同的疾病表现。
    Tick-borne flaviviruses (TBFV) can cause severe neuroinvasive disease which may result in death or long-term neurological deficit in over 50% of survivors. Multiple mechanisms for invasion of the central nervous system (CNS) by flaviviruses have been proposed including axonal transport, transcytosis, endothelial infection, and Trojan horse routes. Flaviviruses may utilize different or multiple mechanisms of neuroinvasion depending on the specific virus, infection site, and host variability. In this work we have shown that the infection of BALB/cJ mice with either Powassan virus lineage I (Powassan virus) or lineage II (deer tick virus) results in distinct spatial tropism of infection in the CNS which correlates with unique clinical presentations for each lineage. Comparative transcriptomics of infected brains demonstrates the activation of different immune pathways and downstream host responses. Ultimately, the comparative pathology and transcriptomics are congruent with different clinical signs in a murine model. These results suggest that the different disease presentations occur in clinical cases due to the inherent differences in the two lineages of Powassan virus.
    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
    由冠状病毒SARS-CoV-2引起的COVID-19大流行揭示了许多问题和医学发现,特别是,关于病毒对中枢神经系统(CNS)和周围神经系统(PNS)的影响。本文是一篇叙述性综述,深入探讨了COVID-19与NS之间的复杂相互作用。因此,本文阐述了由病毒引起的广泛的神经表现和神经退行性疾病。它仔细考虑了SARS-CoV-2到达NS的路线,包括嗅觉系统,当然,血行途径,在讨论病毒的神经发病机制的直接和间接机制时,也包括在内。除了像中风这样的神经病,脑炎,格林-巴利综合征,帕金森病,和多发性硬化症,重点领域也是诊断的挑战,治疗,以及在大流行期间对这些疾病的管理。审查还审查了用于预防这些疾病的战略和干预方法,以及与介导COVID-19引起的神经系统作用有关的ACE2受体。这个详细的概述,将研究结果与案例数据相结合,旨在应对这一流行病挑战,以期将来更好的患者护理和结果。
    The COVID-19 pandemic caused by the coronavirus SARS-CoV-2 revealed a huge number of problems as well as discoveries in medicine, notably, regarding the effects of the virus on the central nervous system (CNS) and peripheral nervous system (PNS). This paper is a narrative review that takes a deep dive into the complex interactions between COVID-19 and the NS. Therefore, this paper explains the broad range of neurological manifestations and neurodegenerative diseases caused by the virus. It carefully considers the routes through which SARS-CoV-2 reaches the NS, including the olfactory system and of course, the hematogenous route, which are also covered when discussing the virus\'s direct and indirect mechanisms of neuropathogenesis. Besides neurological pathologies such as stroke, encephalitis, Guillain-Barré syndrome, Parkinson\'s disease, and multiple sclerosis, the focus area is also given to the challenges of making diagnosis, treatment, and management of these conditions during the pandemic. The review also examines the strategic and interventional approaches utilized to prevent these disorders, as well as the ACE2 receptors implicated in the mediation of neurological effects caused by COVID-19. This detailed overview, which combines research outputs with case data, is directed at tackling this pandemic challenge, with a view toward better patient care and outcomes in the future.
    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
    背景:EcoHIV是一种在小鼠CD4+T细胞中复制的嵌合HIV,巨噬细胞,和小胶质细胞(但不在神经元中),引起持久的神经认知障碍,类似于HIV感染者的神经认知疾病。本研究旨在开发对EcoHIV敏感的原代小鼠脑培养物,以研究HIV感染对神经元完整性的间接影响。
    结果:我们使用了两个编码EGFP和小鼠骨髓源性巨噬细胞(BMM)的EcoHIV克隆,混合的小鼠脑细胞,或从两个野生型小鼠品系中富集小鼠神经胶质细胞来测试EcoHIV复制效率,生产性感染细胞的身份,神经元凋亡和完整性。EcoHIV在BMM中有效复制。在混合脑细胞培养中,EcoHIV靶向小胶质细胞但不引起神经元凋亡。相反,小胶质细胞的生产性感染激活了它们,突触素表达受损,树枝状密度,和神经元的轴突结构。通过抗逆转录病毒培养,可以防止小胶质细胞中的EcoHIV复制和感染过程中的神经元结构变化。
    结论:在鼠脑细胞培养中,小胶质细胞中的EcoHIV复制在很大程度上是与感染小鼠和HIV感染者的认知疾病相关的神经元功能障碍的原因。这些培养物提供了进一步研究HIV神经发病机制及其控制的工具。
    EcoHIV is a chimeric HIV that replicates in mice in CD4+ T cells, macrophages, and microglia (but not in neurons), causing lasting neurocognitive impairment resembling neurocognitive disease in people living with HIV. The present study was designed to develop EcoHIV-susceptible primary mouse brain cultures to investigate the indirect effects of HIV infection on neuronal integrity.
    We used two EcoHIV clones encoding EGFP and mouse bone marrow-derived macrophages (BMM), mixed mouse brain cells, or enriched mouse glial cells from two wild-type mouse strains to test EcoHIV replication efficiency, the identity of productively infected cells, and neuronal apoptosis and integrity. EcoHIV replicated efficiently in BMM. In mixed brain cell cultures, EcoHIV targeted microglia but did not cause neuronal apoptosis. Instead, the productive infection of the microglia activated them and impaired synaptophysin expression, dendritic density, and axonal structure in the neurons. EcoHIV replication in the microglia and neuronal structural changes during infection were prevented by culture with an antiretroviral.
    In murine brain cell cultures, EcoHIV replication in the microglia is largely responsible for the aspects of neuronal dysfunction relevant to cognitive disease in infected mice and people living with HIV. These cultures provide a tool for further study of HIV neuropathogenesis and its control.
    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
    哺乳动物正道病毒(呼肠孤病毒)是一种无包膜病毒,在肠道中建立原发性感染并传播到继发性感染部位,包括CNS。呼肠孤病毒进入涉及多个参与因素,但该病毒是如何系统性传播并靶向神经元的,目前尚不清楚。在这项研究中,我们确定鼠神经纤毛蛋白1(mNRP1)是呼肠孤病毒的受体.mNRP1使用病毒-受体相互作用的独特机制以纳摩尔亲和力结合呼肠孤病毒,由不同呼肠孤病毒衣壳亚基和多个NRP1胞外域之间的多种相互作用协调。通过交换必需的衣壳蛋白编码基因片段,我们确定多价相互作用是由外部衣壳蛋白σ3和衣壳转塔蛋白λ2介导的。使用不能结合NRP1的衣壳突变体,我们发现NRP1有助于小鼠的呼肠孤病毒传播和神经毒力。总的来说,我们的结果表明NRP1是呼肠孤病毒的进入受体,并揭示了NRP促进病毒进入和发病机制.
    Mammalian orthoreovirus (reovirus) is a nonenveloped virus that establishes primary infection in the intestine and disseminates to sites of secondary infection, including the CNS. Reovirus entry involves multiple engagement factors, but how the virus disseminates systemically and targets neurons remains unclear. In this study, we identified murine neuropilin 1 (mNRP1) as a receptor for reovirus. mNRP1 binds reovirus with nanomolar affinity using a unique mechanism of virus-receptor interaction, which is coordinated by multiple interactions between distinct reovirus capsid subunits and multiple NRP1 extracellular domains. By exchanging essential capsid protein-encoding gene segments, we determined that the multivalent interaction is mediated by outer-capsid protein σ3 and capsid turret protein λ2. Using capsid mutants incapable of binding NRP1, we found that NRP1 contributes to reovirus dissemination and neurovirulence in mice. Collectively, our results demonstrate that NRP1 is an entry receptor for reovirus and uncover mechanisms by which NRPs promote viral entry and pathogenesis.
    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
    背景:COVID-19大流行是一种健康紧急情况,由于其高传染性,对世界产生了重大影响。疾病,主要在下呼吸道,发展出许多影响多个器官的临床症状,并显示出嗅觉缺失的临床发现。几位作者研究了SARS-CoV-2感染引起的嗅觉障碍的发病机制,提出不同的假设,并显示出矛盾的结果。由于病毒可能的嗜神经性和对嗅球的直接损害仍然存在不确定性,我们研究了SARS-CoV-2和ACE2受体转录本在自体肺和嗅球组织中的表达,关于组织病理学特征。
    方法:从COVID-19大流行期间进行的200次初次尸检中随机收集25例COVID-19嗅球和肺组织。常规诊断基于临床和放射学发现,并通过验尸拭子证实。对自体FFPE肺和嗅球组织进行SARS-CoV-2和ACE2受体RNA的实时RT-PCR。对组织标本进行组织学染色,并与分子数据进行比较。
    结果:尽管SARS-CoV-2的实时RT-PCR在25个肺样本中有23个呈阳性,嗅球中不存在病毒RNA表达。ACE2受体RNA存在于所有检查的组织中,在肺样本中比嗅球高表达。
    结论:我们的发现表明,COVID-19失语症不仅是由于神经嗜性和SARS-CoV-2进入嗅球的直接作用。嗅球中SARS-CoV-2神经发病机制需要更好的阐明和进一步的研究,以减轻与病毒作用相关的嗅球损伤。
    BACKGROUND: The COVID-19 pandemic has been a health emergency with a significant impact on the world due to its high infectiousness. The disease, primarily identified in the lower respiratory tract, develops with numerous clinical symptoms affecting multiple organs and displays a clinical finding of anosmia. Several authors have investigated the pathogenetic mechanisms of the olfactory disturbances caused by SARS-CoV-2 infection, proposing different hypotheses and showing contradictory results. Since uncertainties remain about possible virus neurotropism and direct damage to the olfactory bulb, we investigated the expression of SARS-CoV-2 as well as ACE2 receptor transcripts in autoptic lung and olfactory bulb tissues, with respect to the histopathological features.
    METHODS: Twenty-five COVID-19 olfactory bulbs and lung tissues were randomly collected from 200 initial autopsies performed during the COVID-19 pandemic. Routine diagnosis was based on clinical and radiological findings and were confirmed with post-mortem swabs. Real-time RT-PCR for SARS-CoV-2 and ACE2 receptor RNA was carried out on autoptic FFPE lung and olfactory bulb tissues. Histological staining was performed on tissue specimens and compared with the molecular data.
    RESULTS: While real-time RT-PCR for SARS-CoV-2 was positive in 23 out of 25 lung samples, the viral RNA expression was absent in olfactory bulbs. ACE2-receptor RNA was present in all tissues examined, being highly expressed in lung samples than olfactory bulbs.
    CONCLUSIONS: Our finding suggests that COVID-19 anosmia is not only due to neurotropism and the direct action of SARS-CoV-2 entering the olfactory bulb. The mechanism of SARS-CoV-2 neuropathogenesis in the olfactory bulb requires a better elucidation and further research studies to mitigate the olfactory bulb damage associated with virus action.
    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
    罗非鱼湖病毒(TiLV)与感染鱼的大脑中的病理变化有关,但驱动病毒的神经发病机制仍未得到充分表征。即使在周围器官中不再检测到病毒,TiLV也会在受感染鱼的大脑中建立持续感染,使治疗干预和疾病管理具有挑战性。此外,病毒在大脑中的持续存在可能会带来病毒再感染和传播的风险,并导致持续的组织损伤和神经炎症过程。在这次审查中,我们探讨了TiLV相关的神经系统疾病。我们讨论了TiLV进入中枢神经系统(CNS)的可能机制,并检查了TiLV诱导的神经炎症和脑免疫反应。最后,我们讨论未来的研究问题和需要解决的知识差距,以显著推进这一领域。
    Tilapia Lake Virus (TiLV) is associated with pathological changes in the brain of infected fish, but the mechanisms driving the virus\'s neuropathogenesis remain poorly characterized. TiLV establishes a persistent infection in the brain of infected fish even when the virus is no longer detectable in the peripheral organs, rendering therapeutic interventions and disease management challenging. Moreover, the persistence of the virus in the brain may pose a risk for viral reinfection and spread and contribute to ongoing tissue damage and neuroinflammatory processes. In this review, we explore TiLV-associated neurological disease. We discuss the possible mechanism(s) used by TiLV to enter the central nervous system (CNS) and examine TiLV-induced neuroinflammation and brain immune responses. Lastly, we discuss future research questions and knowledge gaps to be addressed to significantly advance this field.
    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
    细胞前生存和死亡途径之间的精确平衡对于稳态是极其必要的。不同形式的程序性细胞死亡已被广泛研究和报道,如细胞凋亡,坏死,焦亡,和自噬。自噬是对正常细胞功能重要的分解代谢过程。这种机器的主要目的是降解错误折叠或受损的蛋白质,无用的细胞器,和病原体,侵入细胞,从而维持细胞稳态并确保细胞成分的定期更新。自噬的这种促生存功能凸显了其在许多人类疾病中的重要性。因为这个紧密组织的过程的干扰最终会造成有害的影响。有趣的是,在自噬活动的基础水平的任何改变的存在下,神经元特别容易受到损害;这可能是由于它们的高代谢需求,有丝分裂后的性质,以及自噬在神经元不同基本功能中的作用。在这里,我们报道了自噬在不同的中枢神经系统疾病如帕金森病中的作用,老年痴呆症,亨廷顿病,癫痫,除了靶向自噬的药物。由于自噬在许多疾病的发病机制中的重要作用,开发有效的方法来检测这种动态过程至关重要。在这一章中,我们总结了研究和检测自噬最常用的技术,包括电子显微镜,荧光显微镜,西方印迹,细胞内蛋白质降解,和隔离试验。
    The exquisite balance between cellular prosurvival and death pathways is extremely necessary for homeostasis. Different forms of programmed cell death have been widely studied and reported such as apoptosis, necroptosis, pyroptosis, and autophagy. Autophagy is a catabolic process important for normal cellular functioning. The main aim of this machinery is to degrade the misfolded or damaged proteins, unuseful organelles, and pathogens, which invade the cells, thereby maintaining cellular homeostasis and assuring the regular renewal of cell components. This prosurvival function of autophagy highlights its importance in many human diseases, as the disturbance of this tightly organized process ultimately causes detrimental effects. Interestingly, neurons are particularly susceptible to damage upon the presence of any alteration in the basal level of the autophagic activity; this could be due to their high metabolic demand, post-mitotic nature, and the contribution of autophagy in the different fundamental functions of neurons. Herein, we have reported the role of autophagy in different CNS disorders such as Parkinson\'s disease, Alzheimer\'s disease, Huntington\'s disease, and epilepsy, besides the pharmacological agents targeting autophagy. Due to the significant contribution of autophagy in the pathogenesis of many diseases, it is crucial to develop effective methods to detect this dynamic process. In this chapter, we have summarized the most frequently employed techniques in studying and detecting autophagy including electron microscopy, fluorescence microscopy, Western blotting, intracellular protein degradation, and sequestration assay.
    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
    HIV感染由于其致病性和神经致病性特征而损害外周和中枢免疫系统。驱动HIV-1发病机制和神经发病机制涉及一系列事件,包括代谢失调.此外,HIV亚型特异性变异,特别是关键病毒蛋白的氨基酸序列的改变,已知会影响HIV感染者临床结局的严重程度。然而,特定病毒蛋白中氨基酸序列变异的影响,如病毒蛋白R(Vpr),对HIV感染者中色氨酸(Trp)-犬尿氨酸(Kyn)途径内的代谢物仍不清楚。我们的研究旨在探索Vpr氨基酸序列的变异(特别是在22、41、45和55位,因为这些先前与神经认知功能有关)与外周Trp-Kyn代谢物之间的关系。此外,我们试图通过检查Trp-Kyn代谢与外周炎症之间的联系来阐明Vpr序列变异的系统生物学,作为一种神经致病机制。在这项初步研究中,我们分析了32例(n=32)南非cART初治HIV感染者的独特队列.我们采用Sanger测序来确定血液来源的Vpr氨基酸序列变异,并采用靶向LC-MS/MS代谢组学平台来评估Trp-Kyn代谢产物。比如Trp,Kyn,犬尿氨酸(KA),和喹啉酸(QUIN)。粒子增强比浊法和酶联免疫吸附试验用于测量免疫标志物,hsCRP,IL-6suPAR,NGAL和sCD163。在应用Bonferroni校正(p=0.05/3)并调整协变量(年龄和性别)后,与VprS41和T55组相比,只有VprG41和A55组的QUIN水平接近显著性,分别(所有p=0.023)。多元回归结果显示,41位的Vpr氨基酸变异(adjR2=0.049,β=0.505;p=0.023),55(adjR2=0.126,β=0.444;p=0.023)在调整年龄和性别后显示与QUIN的显着关联。最后,发现在VprG41组中观察到的较高QUIN水平与suPAR相关(r=.588,p=.005)。这些结果共同强调了特定Vpr氨基酸取代在影响QUIN和炎症(特别是suPAR水平)方面的重要性。可能有助于我们理解它们在HIV-1发病机制和神经发病机制中的作用。
    HIV infection compromises both the peripheral and central immune systems due to its pathogenic and neuropathogenic features. The mechanisms driving HIV-1 pathogenesis and neuropathogenesis involve a series of events, including metabolic dysregulation. Furthermore, HIV-subtype-specific variations, particularly alterations in the amino acid sequences of key viral proteins, are known to influence the severity of clinical outcomes in people living with HIV. However, the impact of amino acid sequence variations in specific viral proteins, such as Viral protein R (Vpr), on metabolites within the Tryptophan (Trp)-kynurenine (Kyn) pathway in people living with HIV remains unclear. Our research aimed to explore the relationship between variations in the Vpr amino acid sequence (specifically at positions 22, 41, 45, and 55, as these have been previously linked to neurocognitive function) and peripheral Trp-Kyn metabolites. Additionally, we sought to clarify the systems biology of Vpr sequence variation by examining the link between Trp-Kyn metabolism and peripheral inflammation, as a neuropathogenic mechanism. In this preliminary study, we analyzed a unique cohort of thirty-two (n = 32) South African cART naïve people living with HIV. We employed Sanger sequencing to ascertain blood-derived Vpr amino acid sequence variations and a targeted LC-MS/MS metabolomics platform to assess Trp-Kyn metabolites, such as Trp, Kyn, kynurenic acid (KA), and quinolinic acid (QUIN). Particle-enhanced turbidimetric assay and Enzyme-linked immunosorbent assays were used to measure immune markers, hsCRP, IL-6, suPAR, NGAL and sCD163. After applying Bonferroni corrections (p =.05/3) and adjusting for covariates (age and sex), only the Vpr G41 and A55 groups was nearing significance for higher levels of QUIN compared to the Vpr S41 and T55 groups, respectively (all p =.023). Multiple regression results revealed that Vpr amino acid variations at position 41 (adj R2 = 0.049, β = 0.505; p =.023), and 55 (adj R2 = 0.126, β = 0.444; p =.023) displayed significant associations with QUIN after adjusting for age and sex. Lastly, the higher QUIN levels observed in the Vpr G41 group were found to be correlated with suPAR (r =.588, p =.005). These results collectively underscore the importance of specific Vpr amino acid substitutions in influencing QUIN and inflammation (specifically suPAR levels), potentially contributing to our understanding of their roles in the pathogenesis and neuropathogenesis of HIV-1.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号