neurotropic virus

嗜神经病毒
  • 文章类型: Journal Article
    大脑的三维培养模型能够在复杂的互连细胞基质的背景下研究神经感染。根据神经细胞的分化状态,存在两种模型:3D球体,也称为神经球和大脑器官。这里,我们描述了3D球体和脑类器官的制备,并对它们在研究裂谷热病毒和其他嗜神经病毒中的应用进行了展望。
    Three-dimensional culture models of the brain enable the study of neuroinfection in the context of a complex interconnected cell matrix. Depending on the differentiation status of the neural cells, two models exist: 3D spheroids also called neurospheres and cerebral organoids. Here, we describe the preparation of 3D spheroids and cerebral organoids and give an outlook on their usage to study Rift Valley fever virus and other neurotropic viruses.
    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: Case Reports
    纵向广泛的横贯性脊髓炎是由登革热感染引起的一种罕见的神经系统表现。这里,我们描述了一名24岁男性的罕见表现,该患者出现发热和斑丘疹,随后出现弛缓性四肢轻瘫伴急性尿潴留。对整个脊柱进行对比的磁共振成像显示,在脐带中发现了长段不明确的高强度信号。患者接受了静脉内类固醇和随后静脉内免疫球蛋白的保守治疗。患者定期随访,情况良好。目前,患者正在服用剂量逐渐减少的强的松龙片剂。
    Longitudinally extensive transverse myelitis is a rare neurological manifestation caused by dengue infection. Here, we describe the uncommon presentation of a 24-year-old male with fever and maculopapular rash followed by flaccid quadriparesis with acute urinary retention. Magnetic resonance imaging of the whole spine with contrast revealed a long-segment ill-defined hyperintense signal noted in the cord. The patient was managed conservatively with intravenous steroids and later intravenous immunoglobulins. The patient is on regular follow-up and doing well. Currently, the patient is on tablet prednisolone with a tapering dose.
    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
    通过与非接种眼亲神经病毒性视网膜感染的vonSzily动物模型的比较,重新评估急性后部多灶性胎盘色素上皮病(APMPPE)和无情的胎盘脉络膜视网膜炎(RPC)的潜在病理生理学。
    叙事回顾。
    APMPPE中分离的嗜神经病毒实体和血清学病毒滴度上升的文献报道支持潜在的直接感染病因。总的来说,病毒沿轴突的运输导致线粒体停滞和轴质流的破坏。APMPPE/RPC中的轴质流破坏的临床表现可能表明病毒沿神经元途径通过。从11名患者的病例系列中,我们展示了一个及时的,空间,以及视盘肿胀与APMPPE病变发生的比例关联。内部视网膜内的体征似乎先于外部视网膜病变;外核层(ONL)的急性区域高反射率似乎是多个类似轴突球体的高反射焦点合并的结果(由于轴浆破裂而发生),并跟随Henle纤维层神经元。视网膜色素上皮(RPE)高自发荧光的基础区域遵循ONL高反射率,可能表示局部感染。RPE/Bruch膜分离的明显脉络膜毛细血管灌注不足镜区域,并在上述牵引力下出现。在APMPPE/RPC和vonSzily模型中均观察到脉络膜厚度增加,病变发生和脉络膜毛细血管灌注不足的局灶性区域。
    神经营养性感染模型比现有的原发性脉络膜毛细血管缺血假说提供了显着优势,以解释在APMPPE和RPC中观察到的影像学征象范围。
    UNASSIGNED: To reassess the underlying pathophysiology of acute posterior multifocal placoid pigment epitheliopathy (APMPPE) and relentless placoid chorioretinitis (RPC) through comparison with the non-inoculated eye of the von Szily animal model of neurotropic viral retinal infection.
    UNASSIGNED: Narrative review.
    UNASSIGNED: Literature reports of isolated neurotropic viral entities and rising serological viral titers in APMPPE after presentation support a potential direct infective etiology. In general, viral transport along axons results in mitochondrial stasis and disruption of axoplasmic flow. Clinical manifestations of axoplasmic flow disruption in APMPPE/RPC may signify the passage of virus along the neuronal pathway. From a case series of 11 patients, we demonstrate a timely, spatial, and proportional association of optic disc swelling with APMPPE lesion occurrence. Signs within the inner retina appear to precede outer retinal lesions; and acute areas of outer nuclear layer (ONL) hyperreflectivity appear to be the result of coalescence of multiple hyperreflective foci resembling axonal spheroids (which occur as a consequence of axoplasmic disruption) and follow the Henle fiber layer neurons. Underlying areas of retinal pigment epithelium (RPE) hyper-autofluorescence follow ONL hyperreflectivity and may signify localized infection. Areas of apparent choriocapillaris hypoperfusion mirror areas of RPE/Bruch\'s membrane separation and appear secondary to tractional forces above. Increases in choroidal thickness with lesion occurrence and focal areas of choriocapillaris hypoperfusion are observed in both APMPPE/RPC and the von Szily model.
    UNASSIGNED: The neurotrophic infection model provides significant advantages over the existing primary choriocapillaris ischemia hypothesis to account for the range of imaging signs observed in APMPPE and RPC.
    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
    西尼罗河病毒(WNV)是一种重要的嗜神经病毒,它解释了人类虫媒病毒性脑炎和脑膜炎的出现。WNV与信号通路的相互作用在控制WNV感染中起关键作用。我们已经研究了AKT和ERK途径在支持WNV传播和调节WNV感染后的炎症反应中的作用。WNV在源自人和小鼠的神经元细胞系中建立了生产性感染。IL-11和TNF-α的表达在感染的人神经细胞中显著上调,表明在WNV感染后引发炎症反应。WNV孵育快速激活AKT(AKT-S6-4E-BP1)和ERK(MEK-ERK-p90RSK)途径的信号传导级联。用AKT抑制剂MK-2206或MEK抑制剂U0126治疗消除了WNV诱导的AKT或ERK活化。在WNV感染后24小时可以检测到AKT和ERK信号通路的强烈激活,而这种激活在感染后48小时被取消。U0126处理或敲低ERK表达显著增加WNVRNA水平和病毒滴度,并有效降低WNV诱导的IL-11产生,提示ERK通路参与WNV增殖和IL-11诱导。MK-2206处理增强WNVRNA复制,伴随IL-11产生的适度降低。这些结果表明,AKT和ERK信号通路的参与促进了病毒感染,并且可能与WNV发病机理有关。
    West Nile virus (WNV) is an important neurotropic virus that accounts for the emergence of human arboviral encephalitis and meningitis. The interaction of WNV with signaling pathways plays a key role in controlling WNV infection. We have investigated the roles of the AKT and ERK pathways in supporting WNV propagation and modulating the inflammatory response following WNV infection. WNV established a productive infection in neuronal cell lines originated from human and mouse. Expression of IL-11 and TNF-α was markedly up-regulated in the infected human neuronal cells, indicating elicitation of inflammation response upon WNV infection. WNV incubation rapidly activated signaling cascades of AKT (AKT-S6-4E-BP1) and ERK (MEK-ERK-p90RSK) pathways. Treatment with AKT inhibitor MK-2206 or MEK inhibitor U0126 abrogated WNV-induced AKT or ERK activation. Strong activation of AKT and ERK signaling pathways could be detectable at 24 h after WNV infection, while such activation was abolished at 48 h post infection. U0126 treatment or knockdown of ERK expression significantly increased WNV RNA levels and viral titers and efficiently decreased IL-11 production induced by WNV, suggesting the involvement of ERK pathway in WNV propagation and IL-11 induction. MK-2206 treatment enhanced WNV RNA replication accompanied with a moderate decrease in IL-11 production. These results demonstrate that engagement of AKT and ERK signaling pathways facilitates viral infection and may be implicated in WNV 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
    裂谷热病毒(RVFV)是一种新兴的具有大流行潜力的虫媒病毒病。虽然感染通常是自限性的,一部分人可能发展为迟发性脑炎,占重症病例的20%。重要的是,显示神经系统疾病的个体有高达53%的病死率,然而,RVFV感染的神经发病机制仍未得到充分研究。在这项研究中,我们评估了离体出生后大鼠脑切片培养物(BSC)是否可用于评估中枢神经系统的RVFV感染。BSC在切片后出现炎症反应,随着时间的推移,它们在培养中存活至少12天。用致病性RVFV菌株ZH501感染大鼠BSC在48小时内诱导组织损伤和凋亡。BSC中的病毒复制达到1×107p.f.u.当量/ml,取决于接种剂量。清除切片的共聚焦免疫荧光显微镜证实了神经元的直接感染以及小胶质细胞和星形胶质细胞的激活。Further,RVFV感染的大鼠BSC产生抗病毒细胞因子和趋化因子,包括MCP-1和GRO/KC。这项研究表明,大鼠BSC支持RVFV的复制,用于神经发病机理的离体研究。这允许在离体出生后脑切片培养形式中对RVFV感染进行持续和补充的研究。
    Rift Valley fever virus (RVFV) is an emerging arboviral disease with pandemic potential. While infection is often self-limiting, a subset of individuals may develop late-onset encephalitis, accounting for up to 20 % of severe cases. Importantly, individuals displaying neurologic disease have up to a 53 % case fatality rate, yet the neuropathogenesis of RVFV infection remains understudied. In this study, we evaluated whether ex vivo postnatal rat brain slice cultures (BSCs) could be used to evaluate RVFV infection in the central nervous system. BSCs mounted an inflammatory response after slicing, which resolved over time, and they were viable in culture for at least 12 days. Infection of rat BSCs with pathogenic RVFV strain ZH501 induced tissue damage and apoptosis over 48 h. Viral replication in BSCs reached up to 1×107 p.f.u. equivalents/ml, depending on inoculation dose. Confocal immunofluorescent microscopy of cleared slices confirmed direct infection of neurons as well as activation of microglia and astrocytes. Further, RVFV-infected rat BSCs produced antiviral cytokines and chemokines, including MCP-1 and GRO/KC. This study demonstrates that rat BSCs support replication of RVFV for ex vivo studies of neuropathogenesis. This allows for continued and complementary investigation into RVFV infection in an ex vivo postnatal brain slice culture format.
    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
    病毒神经感染代表了需要开发抗病毒药物的主要健康负担。针对脑部感染的后果(对症治疗)而不是原因(直接作用的抗病毒药物)的抗病毒化合物构成了一种有希望的缓解策略,需要在相关模型中进行研究。然而,缺乏模仿成年人皮质的生理替代品,限制了我们对病毒引起的神经系统疾病相关机制的理解。这里,我们优化了死后成人皮质脑外植体(OPAB)的器官型培养,作为人工智能(AI)驱动的抗病毒研究的临床前平台。OPAB在几周内显示出强大的生存能力,保存完好的3D细胞结构,病毒的宽容,和自发局部场电位(LFP)。使用LFP作为神经健康的替代品,我们开发了一个机器学习框架来高置信度预测OPAB的感染状态。作为一个概念证明,我们表明,抗病毒治疗的OPAB可以以供体依赖性方式部分恢复感染的OPAB的基于LFP的电活动。一起,我们提出OPAB作为一个生理相关的和通用的模型来研究神经感染和超越,为临床前药物发现提供平台。
    Viral neuroinfections represent a major health burden for which the development of antivirals is needed. Antiviral compounds that target the consequences of a brain infection (symptomatic treatment) rather than the cause (direct-acting antivirals) constitute a promising mitigation strategy that requires to be investigated in relevant models. However, physiological surrogates mimicking an adult human cortex are lacking, limiting our understanding of the mechanisms associated with viro-induced neurological disorders. Here, we optimized the Organotypic culture of Post-mortem Adult human cortical Brain explants (OPAB) as a preclinical platform for Artificial Intelligence (AI)-driven antiviral studies. OPAB shows robust viability over weeks, well-preserved 3D cytoarchitecture, viral permissiveness, and spontaneous local field potential (LFP). Using LFP as a surrogate for neurohealth, we developed a machine learning framework to predict with high confidence the infection status of OPAB. As a proof-of-concept, we showed that antiviral-treated OPAB could partially restore LFP-based electrical activity of infected OPAB in a donor-dependent manner. Together, we propose OPAB as a physiologically relevant and versatile model to study neuroinfections and beyond, providing a platform for preclinical drug discovery.
    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
    嗜神经病毒可以通过各种机制通过血脑屏障(BBB)渗入CNS,包括细胞旁,跨细胞,和白细胞透析期间的“特洛伊木马”机制。这些病毒属于几个家族,包括逆转录病毒;人类免疫缺陷病毒1型(HIV-1),黄病毒;日本脑炎(JEV);和疱疹病毒;单纯疱疹病毒1型(HSV-1),EB病毒(EBV)和小鼠腺病毒1(MAV-1)。为了进入大脑,病毒蛋白作用于脑微血管内皮细胞(BMEC)之间的紧密连接(TJs)。例如,HIV-1蛋白,如糖蛋白120,Nef,Vpr,还有Tat,破坏BBB并产生神经毒性作用。重组Tat通过降低TJ蛋白如claudin-1、claudin-5和cloadens-1(ZO-1)的表达来触发BBB中的修正。因此,在包括多发性硬化症(MS)在内的许多神经系统疾病中,已经报道了BBB的破坏。嗜神经病毒还表现出与几种髓鞘蛋白的分子模拟,即,发现抗EBV核抗原1(EBNA1)aa411-426与MBP和EBNA1aa385-420交叉反应的抗体与MS风险单倍型HLA-DRB1*150相关.值得注意的是,髓磷脂蛋白表位(PLP139-151,MOG35-55和MBP87-99)用于生成MS的模型系统,例如实验性自身免疫性脑脊髓炎(EAE),以了解疾病机制和治疗方法。如Theiler鼠脑脊髓炎病毒(TMEV)的病毒也通常用于产生EAE。总之,这篇综述提供了有关病毒与BBB渗漏和MS相关的见解,以及可能用于治疗的机制细节。
    Neurotropic viruses can infiltrate the CNS by crossing the blood-brain barrier (BBB) through various mechanisms including paracellular, transcellular, and \"Trojan horse\" mechanisms during leukocyte diapedesis. These viruses belong to several families, including retroviruses; human immunodeficiency virus type 1 (HIV-1), flaviviruses; Japanese encephalitis (JEV); and herpesviruses; herpes simplex virus type 1 (HSV-1), Epstein-Barr virus (EBV), and mouse adenovirus 1 (MAV-1). For entering the brain, viral proteins act upon the tight junctions (TJs) between the brain microvascular endothelial cells (BMECs). For instance, HIV-1 proteins, such as glycoprotein 120, Nef, Vpr, and Tat, disrupt the BBB and generate a neurotoxic effect. Recombinant-Tat triggers amendments in the BBB by decreasing expression of the TJ proteins such as claudin-1, claudin-5, and zona occludens-1 (ZO-1). Thus, the breaching of BBB has been reported in myriad of neurological diseases including multiple sclerosis (MS). Neurotropic viruses also exhibit molecular mimicry with several myelin sheath proteins, i.e., antibodies against EBV nuclear antigen 1 (EBNA1) aa411-426 cross-react with MBP and EBNA1 aa385-420 was found to be associated with MS risk haplotype HLA-DRB1*150. Notably, myelin protein epitopes (PLP139-151, MOG35-55, and MBP87-99) are being used to generate model systems for MS such as experimental autoimmune encephalomyelitis (EAE) to understand the disease mechanism and therapeutics. Viruses like Theiler\'s murine encephalomyelitis virus (TMEV) are also commonly used to generate EAE. Altogether, this review provide insights into the viruses\' association with BBB leakiness and MS along with possible mechanistic details which could potentially use for therapeutics.
    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
    α-疱疹病毒感染(α-HVs)广泛存在,影响了70%以上的成年人。通常,感染开始于粘膜上皮,病毒颗粒侵入周围神经系统的轴突。在外周神经节的核中,α-HV建立终身潜伏期并最终经历多个再激活周期。重新激活后,病毒后代可以进入神经,回到他们进入有机体的外围,或者它们可以向中枢神经系统(CNS)移动。这种潜伏期-再激活循环是由宿主的内在和先天性免疫反应的复杂作用控制的,并被病毒蛋白精细抵消,努力在人群中共存。如果这种阴阳或纳什平衡样平衡状态由于免疫抑制或宿主反应因子特别是中枢神经系统的基因突变而被打破,或者其他致病刺激的存在,α-HV再激活可能导致危及生命的病理。在这次审查中,我们将总结从粘膜上皮感染开始的分子病毒-宿主相互作用,导致PNS潜伏期的建立和α-HVs可能的CNS侵袭,强调与NS中不受控制的病毒复制相关的病理。
    Alpha herpesvirus infections (α-HVs) are widespread, affecting more than 70% of the adult human population. Typically, the infections start in the mucosal epithelia, from which the viral particles invade the axons of the peripheral nervous system. In the nuclei of the peripheral ganglia, α-HVs establish a lifelong latency and eventually undergo multiple reactivation cycles. Upon reactivation, viral progeny can move into the nerves, back out toward the periphery where they entered the organism, or they can move toward the central nervous system (CNS). This latency-reactivation cycle is remarkably well controlled by the intricate actions of the intrinsic and innate immune responses of the host, and finely counteracted by the viral proteins in an effort to co-exist in the population. If this yin-yang- or Nash-equilibrium-like balance state is broken due to immune suppression or genetic mutations in the host response factors particularly in the CNS, or the presence of other pathogenic stimuli, α-HV reactivations might lead to life-threatening pathologies. In this review, we will summarize the molecular virus-host interactions starting from mucosal epithelia infections leading to the establishment of latency in the PNS and to possible CNS invasion by α-HVs, highlighting the pathologies associated with uncontrolled virus replication in the NS.
    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
    神经退行性疾病(ND)构成一组以神经系统功能进行性恶化为特征的疾病。目前,造成NDS的确切病因仍未完全阐明,虽然很可能是衰老的组合,遗传易感性,环境压力源参与了这一过程。越来越多的证据表明病毒感染,尤其是嗜神经病毒,可能有助于ND的发作和进展。在这次审查中,总结了支持病毒感染与ND之间关联的新证据,讨论了病毒感染介导的自噬途径如何引起与各种ND相关的细胞蛋白的病理性聚集。此外,分析了参与单纯疱疹病毒(HSV-1)感染和ND的自噬相关基因(ARGs),并讨论了这些基因是否可以将HSV-1感染与ND联系起来。阐明NDs的潜在机制对于开发预防NDs发作和减缓其进展的靶向治疗方法至关重要。
    Neurodegenerative diseases (NDs) constitute a group of disorders characterized by the progressive deterioration of nervous system functionality. Currently, the precise etiological factors responsible for NDs remain incompletely elucidated, although it is probable that a combination of aging, genetic predisposition, and environmental stressors participate in this process. Accumulating evidence indicates that viral infections, especially neurotropic viruses, can contribute to the onset and progression of NDs. In this review, emerging evidence supporting the association between viral infection and NDs is summarized, and how the autophagy pathway mediated by viral infection can cause pathological aggregation of cellular proteins associated with various NDs is discussed. Furthermore, autophagy-related genes (ARGs) involved in Herpes simplex virus (HSV-1) infection and NDs are analyzed, and whether these genes could link HSV-1 infection to NDs is discussed. Elucidating the mechanisms underlying NDs is critical for developing targeted therapeutic approaches that prevent the onset and slow the progression of NDs.
    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
    中枢神经系统(CNS)的病毒感染会导致从急性到严重的神经系统后遗症的不同结果,并增加发病率和死亡率。病毒神经侵入通过免疫应答失调直接或间接诱导脑炎,并有助于神经元功能的改变和神经元细胞的退化。本文综述了病毒诱导神经变性的细胞和分子机制。嗜神经病毒感染影响神经元功能障碍的许多方面,包括促进慢性炎症,诱导细胞氧化应激,损害线粒体自噬,遇到线粒体动力学,增强新陈代谢重新布线,改变神经递质系统,并诱导与神经退行性疾病相关的错误折叠和聚集的病理蛋白。这些发病机制造成大脑的多维损伤,导致特定的神经元和大脑功能障碍。了解与病毒感染的神经变性相关的神经发生的分子机制可能会强调预防策略,保护,和治疗中枢神经系统的病毒感染。
    Viral infections of the central nervous system (CNS) cause variable outcomes from acute to severe neurological sequelae with increased morbidity and mortality. Viral neuroinvasion directly or indirectly induces encephalitis via dysregulation of the immune response and contributes to the alteration of neuronal function and the degeneration of neuronal cells. This review provides an overview of the cellular and molecular mechanisms of virus-induced neurodegeneration. Neurotropic viral infections influence many aspects of neuronal dysfunction, including promoting chronic inflammation, inducing cellular oxidative stress, impairing mitophagy, encountering mitochondrial dynamics, enhancing metabolic rewiring, altering neurotransmitter systems, and inducing misfolded and aggregated pathological proteins associated with neurodegenerative diseases. These pathogenetic mechanisms create a multidimensional injury of the brain that leads to specific neuronal and brain dysfunction. The understanding of the molecular mechanisms underlying the neurophathogenesis associated with neurodegeneration of viral infection may emphasize the strategies for prevention, protection, and treatment of virus infection of the CNS.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号