neurovirulence

神经毒力
  • 文章类型: Journal Article
    目前没有获得许可的预防性疫苗来预防由拉沙病毒(LASV)感染引起的拉沙热。新兴生物解决方案(EBS)候选疫苗,EBS-LASV,正在开发用于预防拉沙热。EBS-LASV是一种活的减毒重组水泡性口炎病毒(rVSV)载体疫苗,编码来自LASV的表面糖蛋白复合物(GPC),并具有两个减毒载体修饰:VSVN基因的基因改组和VSVG基因的缺失。进行临床前研究以评估颅内(IC)注射后EBS-LASV的神经毒力潜力,并确定小鼠肌内(IM)接种后的生物分布和载体复制。此外,EBS-LASV的潜在毒性是通过对家兔重复给药IMEBS-LASV来评估的.所有接受IC注射EBS-LASV的小鼠均存活,而施用未减毒对照载体的小鼠没有。疫苗只在注射部位的肌肉中检测到,引流淋巴结,在小鼠注射IMEBS-LASV后的第一周,没有检测到的血浆病毒血症。在接受EBS-LASV三剂量方案的兔子中未观察到毒性。这些研究表明,EBS-LASV在动物给药时是安全的,并支持首次在人剂量递增。安全,和免疫原性临床研究。
    There are currently no prophylactic vaccines licensed to protect against Lassa fever caused by Lassa virus (LASV) infection. The Emergent BioSolutions (EBS) vaccine candidate, EBS-LASV, is being developed for the prevention of Lassa fever. EBS-LASV is a live-attenuated recombinant Vesicular Stomatitis Virus (rVSV)-vectored vaccine encoding the surface glycoprotein complex (GPC) from LASV and has two attenuating vector modifications: a gene shuffle of the VSV N gene and a deletion of the VSV G gene. Preclinical studies were performed to evaluate EBS-LASV\'s neurovirulence potential following intracranial (IC) injection and to determine the biodistribution and vector replication following intramuscular (IM) inoculation in mice. In addition, the potential EBS-LASV toxicity was assessed using repeated-dose IM EBS-LASV administration to rabbits. All mice receiving the IC injection of EBS-LASV survived, while mice administered the unattenuated control vector did not. The vaccine was only detected in the muscle at the injection site, draining lymph nodes, and the spleen over the first week following IM EBS-LASV injection in mice, with no detectable plasma viremia. No toxicity was observed in rabbits receiving a three-dose regimen of EBS-LASV. These studies demonstrate that EBS-LASV is safe when administered to animals and supported a first-in-human dose-escalation, safety, and immunogenicity clinical study.
    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
    马疱疹病毒1型(EHV-1)引起呼吸道疾病,胎儿丢失,围产期死亡率,和骨髓脑病。本研究调查了ORF15对病毒感染性和神经毒力的影响。以EHV1的Ab4p神经毒力菌株为骨架制备Ab4pattB,通过BACDNA转染到RK-13细胞中的Ab4pΔORF15和Ab4pΔORF15R嵌合体。病毒生长动力学,斑块大小,转录,并在MDBK细胞中评估生长,小鼠神经元,和胎儿马脑细胞。鼻内接种雄性CBA/N1SPF小鼠后评估神经毒力,测量标志,病毒滴度,和组织病理学变化。删除EUL45(Ab4p-ΔEUL45)降低了病毒复制效率,导致释放减少和更小的斑块。EUL45缺失也上调邻近基因(EUL46和EUL44)。与野生型病毒相比,Ab4p-ΔEUL45在神经细胞中表现出降低的毒力和较差的生长。本研究揭示了EUL45在EHV-1,病毒复制中的作用,和EUL46和EUL44表达的调节,提示作为疫苗候选的潜力。
    Equine herpesvirus 1 (EHV-1) causes respiratory illness, fetal loss, perinatal mortality, and myeloencephalopathy. This study investigated ORF15\'s impact on virus infectivity and neurovirulence. The Ab4p neurovirulent strain of EHV1 was used as a backbone to create Ab4p attB, Ab4p∆ORF15, and Ab4p∆ORF15R chimeras via BAC DNA transfection into RK-13 cells. Viral growth kinetics, plaque size, transcription, and growth were assessed in MDBK cells, mouse neurons, and fetal equine brain cells. Neurovirulence was evaluated post-intranasal inoculation into male CBA/N1 SPF mice, measuring signs, virus titers, and histopathological changes. Deletion of EUL45 (Ab4p-∆EUL45) reduced viral replication efficiency, resulting in decreased release and smaller plaques. EUL45 deletion also upregulated neighbouring genes (EUL46 and EUL44). Ab4p-∆EUL45 exhibited reduced virulence and poor growth in neural cells compared to wild-type viruses. This study sheds light on EUL45\'s role in EHV-1, viral replication, and regulation of EUL46 and EUL44 expression, suggesting potential as a vaccine candidate.
    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
    Powassan病毒(POWV)是一种新兴的蜱传黄病毒,可引起老年人的致命脑炎和幸存者的长期神经系统后遗症。年龄如何导致严重的POWV脑炎仍然是一个谜,并且没有动物模型评估年龄依赖性POWV神经病理学。用目前在Ixodes蜱中循环的POWV毒株(LI9)接种C57BL/6小鼠导致年龄依赖性POWV致死率10-20dpi。50周龄小鼠的POWV感染致死性为82%,10周龄小鼠的致死率随年龄依次降低至7.1%。POWVLI9对所有年龄的小鼠都有神经侵袭性,导致急性海绵状CNS病理和反应性神经胶质增生5-15dpi,持续存在于幸存者30dpi中。在所有10dpi小鼠中均发现高CNS病毒载量。然而,15dpi,10至40周龄小鼠的病毒载量减少了2-4个日志,而在50周龄小鼠中保持高水平。CNS病毒载量15dpi的年龄依赖性差异伴随着CNS细胞因子反应的显着变化。在50周龄小鼠的中枢神经系统中,POWV诱导的Th1型细胞因子(IFNγ,IL-2,IL-12,IL-4,TNFα,IL-6),提示神经退行性促炎M1小胶质细胞程序。相比之下,在10周大的老鼠身上,POWV诱导的Th2型细胞因子(IL-10,TGFβ,IL-4)与神经保护性M2小胶质细胞表型一致。这些发现将年龄依赖性CNS细胞因子反应和病毒载量与POWV致死率相关联,并提示了潜在的神经炎性治疗靶标。我们的结果在小鼠模型中建立了POWV的年龄依赖性致死率,该模型反映了老年人的人类POWV严重程度和长期CNS病理。
    目的:Powassan病毒是一种新兴的蜱传黄病毒,在老年人中引起致死性脑炎。我们揭示了一个年龄依赖性POWV小鼠模型,该模型反映了老年人的人类POWV脑炎和长期中枢神经系统损害。我们发现POWV是神经侵袭性的,并在所有年龄的小鼠中引导反应性神经胶质增生,但在急性期,在50周龄小鼠中选择性诱导促炎Th1细胞因子反应,在10周龄小鼠中诱导神经保护性Th2细胞因子反应。我们的发现将CNS病毒载量和不同的细胞因子反应与年龄依赖性POWV致死率和生存结果相关联。幼鼠的反应提示了预防严重POWV脑炎的潜在治疗靶标和方法,可广泛适用于其他神经退行性疾病。我们的年龄依赖性小鼠POWV模型允许分析预防POWV致死性的疫苗,和解决严重POWV脑炎的治疗方法。
    Powassan virus (POWV) is an emergent tick-borne flavivirus that causes fatal encephalitis in the elderly and long-term neurologic sequelae in survivors. How age contributes to severe POWV encephalitis remains an enigma, and no animal models have assessed age-dependent POWV neuropathology. Inoculating C57BL/6 mice with a POWV strain (LI9) currently circulating in Ixodes ticks resulted in age-dependent POWV lethality 10-20 dpi. POWV infection of 50-week-old mice was 82% fatal with lethality sequentially reduced by age to 7.1% in 10-week-old mice. POWV LI9 was neuroinvasive in mice of all ages, causing acute spongiform CNS pathology and reactive gliosis 5-15 dpi that persisted in survivors 30 dpi. High CNS viral loads were found in all mice 10 dpi. However, by 15 dpi, viral loads decreased by 2-4 logs in 10- to 40-week-old mice, while remaining at high levels in 50-week-old mice. Age-dependent differences in CNS viral loads 15 dpi occurred concomitantly with striking changes in CNS cytokine responses. In the CNS of 50-week-old mice, POWV induced Th1-type cytokines (IFNγ, IL-2, IL-12, IL-4, TNFα, IL-6), suggesting a neurodegenerative pro-inflammatory M1 microglial program. By contrast, in 10-week-old mice, POWV-induced Th2-type cytokines (IL-10, TGFβ, IL-4) were consistent with a neuroprotective M2 microglial phenotype. These findings correlate age-dependent CNS cytokine responses and viral loads with POWV lethality and suggest potential neuroinflammatory therapeutic targets. Our results establish the age-dependent lethality of POWV in a murine model that mirrors human POWV severity and long-term CNS pathology in the elderly.
    OBJECTIVE: Powassan virus is an emerging tick-borne flavivirus causing lethal encephalitis in aged individuals. We reveal an age-dependent POWV murine model that mirrors human POWV encephalitis and long-term CNS damage in the elderly. We found that POWV is neuroinvasive and directs reactive gliosis in all age mice, but at acute stages selectively induces pro-inflammatory Th1 cytokine responses in 50-week-old mice and neuroprotective Th2 cytokine responses in 10-week-old mice. Our findings associate CNS viral loads and divergent cytokine responses with age-dependent POWV lethality and survival outcomes. Responses of young mice suggest potential therapeutic targets and approaches for preventing severe POWV encephalitis that may be broadly applicable to other neurodegenerative diseases. Our age-dependent murine POWV model permits analysis of vaccines that prevent POWV lethality, and therapeutics that resolve severe POWV encephalitis.
    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
    α-突触核蛋白(α-syn),以其在帕金森病中的关键作用而闻名,最近已成为嗜神经RNA病毒感染的重要参与者。已发现在各种病毒感染中α-syn的上调通过调节神经递质合成来影响神经保护功能。囊泡贩运,和突触小泡循环。这篇综述着重于α-syn在通过调节对小胶质细胞的化学引诱物特性来控制病毒复制中的多方面作用。病毒诱导的ER应激信号,抗氧化蛋白表达。此外,本文强调了α-syn介导的干扰素刺激基因的调节。该综述可能有助于提出潜在的治疗途径,通过利用α-syn神经保护生物学来减轻RNA病毒对中枢神经系统的影响。
    Alpha-synuclein (α-syn), known for its pivotal role in Parkinson\'s disease, has recently emerged as a significant player in neurotropic RNA virus infections. Upregulation of α-syn in various viral infections has been found to impact neuroprotective functions by regulating neurotransmitter synthesis, vesicle trafficking, and synaptic vesicle recycling. This review focuses on the multifaceted role of α-syn in controlling viral replication by modulating chemoattractant properties towards microglial cells, virus-induced ER stress signaling, anti-oxidative proteins expression. Furthermore, the text underlines the α-syn-mediated regulation of interferon-stimulated genes. The review may help suggest potential therapeutic avenues for mitigating the impact of RNA viruses on the central nervous system by exploiting α-syn neuroprotective biology.
    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
    肠道病毒D68(EV-D68)是一种新兴的病原体,可引起严重的呼吸道和神经系统疾病[急性弛缓性脊髓炎(AFM)]。用US/IL/14-18952(IL52)肌内(IM)注射新生SwissWebster(SW)小鼠,2014年EV-D68流行的临床分离株,导致人类AFM的许多致病特征,包括脊髓的病毒感染,运动神经元死亡,并导致进行性瘫痪。在区别上,CA/14-4231(CA4231),2014年EV-D68爆发的另一种临床分离株,不会导致老鼠瘫痪,不会在脊髓中生长,并且在IM注射后不会导致运动神经元丢失。一组含有来自IL52和CA4231的序列的嵌合病毒用于证明VP1是IM注射新生SW小鼠后EV-D68神经毒力的主要决定因素。VP1含有IL52和CA4231之间的四个氨基酸差别。导致取代这四个氨基酸(CA4231残基进入IL52多蛋白)的突变完全消除了神经毒力。相反,导致将VP1IL52氨基酸残基替换为CA4231多蛋白的突变产生了一种与IL52相同程度的诱导瘫痪的病毒。用亲本和嵌合病毒感染新生SW小鼠后的神经毒力与脊髓中的病毒生长有关。
    目的:新兴病毒允许我们研究导致疾病严重程度增加的突变。肠道病毒D68(EV-D68),曾经是罕见呼吸道疾病的原因,最近获得了引起严重呼吸道和神经系统疾病的能力。嵌合病毒用于证明病毒结构蛋白VP1决定脊髓的生长,运动神经元丢失,用EV-D68肌内(IM)注射新生SwissWebster(SW)小鼠后的瘫痪。这些结果对于预测未来EV-D68流行病的临床结果以及靶向逆行运输作为治疗病毒诱导的神经系统疾病的潜在策略具有相关性。
    Enterovirus D68 (EV-D68) is an emerging pathogen that can cause severe respiratory and neurologic disease [acute flaccid myelitis (AFM)]. Intramuscular (IM) injection of neonatal Swiss Webster (SW) mice with US/IL/14-18952 (IL52), a clinical isolate from the 2014 EV-D68 epidemic, results in many of the pathogenic features of human AFM, including viral infection of the spinal cord, death of motor neurons, and resultant progressive paralysis. In distinction, CA/14-4231 (CA4231), another clinical isolate from the 2014 EV-D68 outbreak, does not cause paralysis in mice, does not grow in the spinal cord, and does not cause motor neuron loss following IM injection. A panel of chimeric viruses containing sequences from IL52 and CA4231 was used to demonstrate that VP1 is the main determinant of EV-D68 neurovirulence following IM injection of neonatal SW mice. VP1 contains four amino acid differences between IL52 and CA4231. Mutations resulting in substituting these four amino acids (CA4231 residues into the IL52 polyprotein) completely abolished neurovirulence. Conversely, mutations resulting in substituting VP1 IL52 amino acid residues into the CA4231 polyprotein created a virus that induced paralysis to the same degree as IL52. Neurovirulence following infection of neonatal SW mice with parental and chimeric viruses was associated with viral growth in the spinal cord.
    OBJECTIVE: Emerging viruses allow us to investigate mutations leading to increased disease severity. Enterovirus D68 (EV-D68), once the cause of rare cases of respiratory illness, recently acquired the ability to cause severe respiratory and neurologic disease. Chimeric viruses were used to demonstrate that viral structural protein VP1 determines growth in the spinal cord, motor neuron loss, and paralysis following intramuscular (IM) injection of neonatal Swiss Webster (SW) mice with EV-D68. These results have relevance for predicting the clinical outcome of future EV-D68 epidemics as well as targeting retrograde transport as a potential strategy for treating virus-induced neurologic disease.
    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
    虽然单纯疱疹病毒1型(HSV-1)基因组被认为含有大约80种不同的蛋白质编码序列(CDS),最近的多组学分析报道HSV-1编码超过200个潜在的CDS。然而,在HSV-1感染的细胞中,几乎没有新鉴定的CDS被证实在肽或蛋白质水平上表达。此外,它们编码的蛋白质对HSV-1感染的影响在很大程度上是未知的.这项研究集中在一种新发现的CDS上,UL31.6.我们先前的化学蛋白质组学数据的再分析证实UL31.6在HSV-1感染的细胞中以肽水平表达。针对UL31.6(pUL31.6)编码的病毒蛋白产生的抗血清与在感染了三种HSV-1菌株的Vero细胞的裂解物中分子量约为37kDa的蛋白质反应。pUL31.6在高盐溶液中有效地从病毒体中解离。UL31.6无效突变对HSV-1基因表达的影响很小,复制,细胞间扩散,和Vero细胞的形态发生;相反,它显着降低了HSV-1在三种神经细胞中的细胞间扩散,但在包括Vero细胞在内的四种非神经细胞中没有。UL31.6-null突变还显著降低了颅内感染后小鼠大脑中的死亡率和病毒复制,但对眼睛内外的致病表现影响很小,眼感染后在小鼠泪膜中检测到病毒复制。这些结果表明,pUL31.6是一种皮膜蛋白,并通过潜在地促进中枢神经系统中神经元细胞之间的病毒传播而特异性地充当神经毒力因子。重要信息最近的多组学分析报道了1型单纯疱疹病毒(HSV-1)基因组编码额外数量的潜在编码序列(CDS)。然而,这些CDS在肽或蛋白质水平上的表达以及这些CDS对HSV-1感染的生物学效应仍在很大程度上未知。这项研究注释了一个神秘的孤儿CDS,被称为UL31.6,一种HSV-1基因,编码分子量约为37kDa的被膜蛋白,它特别充当神经毒力因子。我们的研究表明,对于病毒发病机理重要的HSV-1蛋白仍有待鉴定,对HSV-1发病机理的全面了解不仅需要使用新兴技术鉴定隐匿孤儿CDS,而且还需要逐步和深入地分析每个隐匿孤儿CDS。
    Although the herpes simplex virus type 1 (HSV-1) genome was thought to contain approximately 80 different protein coding sequences (CDSs), recent multi-omics analyses reported HSV-1 encodes more than 200 potential CDSs. However, few of the newly identified CDSs were confirmed to be expressed at the peptide or protein level in HSV-1-infected cells. Furthermore, the impact of the proteins they encode on HSV-1 infection is largely unknown. This study focused on a newly identified CDS, UL31.6. Re-analyzation of our previous chemical proteomics data verified that UL31.6 was expressed at the peptide level in HSV-1-infected cells. Antisera raised against a viral protein encoded by UL31.6 (pUL31.6) reacted with a protein with an approximate molecular mass of 37  kDa in lysates of Vero cells infected with each of three HSV-1 strains. pUL31.6 was efficiently dissociated from virions in high-salt solution. A UL31.6-null mutation had a minimal effect on HSV-1 gene expression, replication, cell-to-cell spread, and morphogenesis in Vero cells; in contrast, it significantly reduced HSV-1 cell-to-cell spread in three neural cells but not in four non-neural cells including Vero cells. The UL31.6-null mutation also significantly reduced the mortality and viral replication in the brains of mice after intracranial infection, but had minimal effects on pathogenic manifestations in and around the eyes, and viral replication detected in the tear films of mice after ocular infection. These results indicated that pUL31.6 was a tegument protein and specifically acted as a neurovirulence factor by potentially promoting viral transmission between neuronal cells in the central nervous system.IMPORTANCERecent multi-omics analyses reported the herpes simplex virus type 1 (HSV-1) genome encodes an additional number of potential coding sequences (CDSs). However, the expressions of these CDSs at the peptide or protein levels and the biological effects of these CDSs on HSV-1 infection remain largely unknown. This study annotated a cryptic orphan CDS, termed UL31.6, an HSV-1 gene that encodes a tegument protein with an approximate molecular mass of 37  kDa, which specifically acts as a neurovirulence factor. Our study indicates that HSV-1 proteins important for viral pathogenesis remain to be identified and a comprehensive understanding of the pathogenesis of HSV-1 will require not only the identification of cryptic orphan CDSs using emerging technologies but also step-by-step and in-depth analyses of each of the cryptic orphan CDSs.
    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:亲神经病毒感染积极操纵宿主细胞代谢以增强病毒神经毒力。尽管高血糖在严重感染期间很常见,其具体作用尚不清楚。这项研究调查了高血糖对肠道病毒71(EV71)的神经毒力的影响,一种依赖于内部核糖体进入位点(IRES)介导的翻译进行复制的神经毒性病毒。方法:利用hSCARB2转基因小鼠,我们探讨了高血糖在EV71感染中的作用,并阐明了其潜在机制.结果:值得注意的是,在hSCARB2转基因小鼠中单独施用胰岛素以减少高血糖导致脑干脑炎和病毒载量的减少.相反,诱导的高血糖会加剧神经发病机制,强调高血糖在神经毒力中的关键作用。值得注意的是,miR-206是由病毒感染诱导的关键介质,其表达因高血糖而进一步增强,同时被胰岛素抑制。使用antagomiR-206可有效缓解EV71诱导的脑干脑炎并降低病毒载量。机械上,miR-206通过抑制应激颗粒蛋白G3BP2促进IRES驱动的病毒复制。结论:针对严重EV71感染的新型治疗方法涉及控制高血糖和靶向miR-206应激颗粒途径以调节病毒IRES活性。
    Background: Neurotropic virus infections actively manipulate host cell metabolism to enhance virus neurovirulence. Although hyperglycemia is common during severe infections, its specific role remains unclear. This study investigates the impact of hyperglycemia on the neurovirulence of enterovirus 71 (EV71), a neurovirulent virus relying on internal ribosome entry site (IRES)-mediated translation for replication. Methods: Utilizing hSCARB2-transgenic mice, we explore the effects of hyperglycemia in EV71 infection and elucidate the underlying mechanisms. Results: Remarkably, administering insulin alone to reduce hyperglycemia in hSCARB2-transgenic mice results in a decrease in brainstem encephalitis and viral load. Conversely, induced hyperglycemia exacerbates neuropathogenesis, highlighting the pivotal role of hyperglycemia in neurovirulence. Notably, miR-206 emerges as a crucial mediator induced by viral infection, with its expression further heightened by hyperglycemia and concurrently repressed by insulin. The use of antagomiR-206 effectively mitigates EV71-induced brainstem encephalitis and reduces viral load. Mechanistically, miR-206 facilitates IRES-driven virus replication by repressing the stress granule protein G3BP2. Conclusions: Novel therapeutic approaches against severe EV71 infections involve managing hyperglycemia and targeting the miR-206-stress granule pathway to modulate virus IRES activity.
    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
    猴痘病毒(MPXV)在人类中的感染会引起神经系统疾病,而对MPXV感染的动物的研究表明该病毒会穿透大脑。焦亡是一种炎症类型的调节细胞死亡,由于裂解的气体蛋白的低聚化导致膜孔形成,导致质膜破裂(PMR)。在这里,我们调查了MPXV与另一种正痘病毒相比的人类神经细胞嗜性,痘苗病毒(VACV),以及它对免疫反应和细胞死亡的影响。星形胶质细胞最允许MPXV(和VACV)感染,其次是小胶质细胞和少突胶质细胞,基于斑块测定的神经元感染最少。在MPXV感染的星形胶质细胞中,异常的形态学变化很明显,伴随着病毒蛋白(I3)的免疫标记,并通过质谱法在细胞裂解物中检测到超过125种MPXV编码的蛋白。MPXV和VACV感染的星形胶质细胞显示免疫基因转录本(IL12,IRF3,IL1B,TNFA,CASP1和GSDMB)。然而,星形胶质细胞的MPXV感染特异性诱导gasderminB(GSDMB)(50kDa)的蛋白水解裂解,通过切割的N-末端-GSDMB(30kDa)和C-末端-GSDMB(18kDa)片段的出现证明。GSDMB裂解与乳酸脱氢酶的释放和细胞核酸染色增加有关,指示PMR。用富马酸二甲酯预处理减少了MPXV感染的星形胶质细胞中GSDMB和相关PMR的裂解。人星形胶质细胞支持生产性MPXV感染,导致伴随GSDMB介导的焦亡的炎症基因诱导。这些发现阐明了最近公认的MPXV在人类中的神经致病作用,同时也提供了潜在的治疗选择。
    Monkeypox virus (MPXV) infections in humans cause neurological disorders while studies of MPXV-infected animals indicate that the virus penetrates the brain. Pyroptosis is an inflammatory type of regulated cell death, resulting from plasma membrane rupture (PMR) due to oligomerization of cleaved gasdermins to cause membrane pore formation. Herein, we investigated the human neural cell tropism of MPXV compared to another orthopoxvirus, vaccinia virus (VACV), as well as its effects on immune responses and cell death. Astrocytes were most permissive to MPXV (and VACV) infections, followed by microglia and oligodendrocytes, with minimal infection of neurons based on plaque assays. Aberrant morphological changes were evident in MPXV-infected astrocytes that were accompanied with viral protein (I3) immunolabelling and detection of over 125 MPXV-encoded proteins in cell lysates by mass spectrometry. MPXV- and VACV-infected astrocytes showed increased expression of immune gene transcripts (IL12, IRF3, IL1B, TNFA, CASP1, and GSDMB). However, MPXV infection of astrocytes specifically induced proteolytic cleavage of gasdermin B (GSDMB) (50 kDa), evident by the appearance of cleaved N-terminal-GSDMB (30 kDa) and C-terminal- GSDMB (18 kDa) fragments. GSDMB cleavage was associated with release of lactate dehydrogenase and increased cellular nucleic acid staining, indicative of PMR. Pre-treatment with dimethyl fumarate reduced cleavage of GSDMB and associated PMR in MPXV-infected astrocytes. Human astrocytes support productive MPXV infection, resulting in inflammatory gene induction with accompanying GSDMB-mediated pyroptosis. These findings clarify the recently recognized neuropathogenic effects of MPXV in humans while also offering potential therapeutic options.
    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:单纯疱疹病毒(HSV)脑炎(HSE)是一种严重且可能危及生命的疾病,影响成人和新生儿。目前的大脑模型的局限性阻碍了对新生儿HSE涉及的病毒和宿主因素的理解,这些模型不能完全概括健康和疾病中发育中的人脑的组织结构和细胞组成。这里,我们开发了人胎儿器官型脑切片培养(hfOBSC)模型,并确定了其在体外模拟HSE神经病理学中的价值。
    方法:通过上清液中的乳酸脱氢酶释放和免疫组织学(IHC)分析确定细胞活力和组织完整性。用表达HSV-1和HSV-2的绿色荧光蛋白(GFP-)感染脑切片。通过共聚焦显微镜确定病毒的复制和传播,PCR和病毒培养。通过PCR检测促炎细胞因子和趋化因子的表达。通过IHC分析确定细胞嗜性和HSV诱导的神经病理学。最后,将HSV感染的hfOBSC的原位数据与人类HSE脑切片中检测到的神经病理学进行比较.
    结果:优化切片和无血清培养条件以在37°C下在CO2培养箱中维持离体人胎儿脑切片的活力和组织结构至少14天。hfOBSC支持生产性HSV-1和HSV-2感染,主要涉及神经元和星形胶质细胞的感染,导致促炎细胞因子和趋化因子的表达。两种病毒在感染后的较晚时间点在感染的脑切片中诱导程序性细胞死亡-尤其是坏死。病毒传播,细胞嗜性和程序性细胞死亡在HSV诱导的细胞死亡中的作用类似于HSE的神经病理学。
    结论:我们开发了一种新型的人脑培养模型,其中包括神经元在内的主要大脑驻留细胞的活力,小胶质细胞,星形胶质细胞和少突胶质细胞-并且在无血清培养条件下在体外维持组织结构至少2周。细胞嗜性非常相似,HSV-1和HSV-2在具有人类HSE病例的神经病理学特征的hfOBSC模型中的传播和神经毒力强调了其在详述其他嗜神经病毒的病理生理学方面的潜力,以及作为测试新型治疗干预措施的临床前模型的潜力.
    BACKGROUND: Herpes simplex virus (HSV) encephalitis (HSE) is a serious and potentially life-threatening disease, affecting both adults and newborns. Progress in understanding the virus and host factors involved in neonatal HSE has been hampered by the limitations of current brain models that do not fully recapitulate the tissue structure and cell composition of the developing human brain in health and disease. Here, we developed a human fetal organotypic brain slice culture (hfOBSC) model and determined its value in mimicking the HSE neuropathology in vitro.
    METHODS: Cell viability and tissues integrity were determined by lactate dehydrogenase release in supernatant and immunohistological (IHC) analyses. Brain slices were infected with green fluorescent protein (GFP-) expressing HSV-1 and HSV-2. Virus replication and spread were determined by confocal microscopy, PCR and virus culture. Expression of pro-inflammatory cytokines and chemokines were detected by PCR. Cell tropism and HSV-induced neuropathology were determined by IHC analysis. Finally, the in situ data of HSV-infected hfOBSC were compared to the neuropathology detected in human HSE brain sections.
    RESULTS: Slicing and serum-free culture conditions were optimized to maintain the viability and tissue architecture of ex vivo human fetal brain slices for at least 14 days at 37 °C in a CO2 incubator. The hfOBSC supported productive HSV-1 and HSV-2 infection, involving predominantly infection of neurons and astrocytes, leading to expression of pro-inflammatory cytokines and chemokines. Both viruses induced programmed cell death-especially necroptosis-in infected brain slices at later time points after infection. The virus spread, cell tropism and role of programmed cell death in HSV-induced cell death resembled the neuropathology of HSE.
    CONCLUSIONS: We developed a novel human brain culture model in which the viability of the major brain-resident cells-including neurons, microglia, astrocytes and oligodendrocytes-and the tissue architecture is maintained for at least 2 weeks in vitro under serum-free culture conditions. The close resemblance of cell tropism, spread and neurovirulence of HSV-1 and HSV-2 in the hfOBSC model with the neuropathological features of human HSE cases underscores its potential to detail the pathophysiology of other neurotropic viruses and as preclinical model to test novel therapeutic interventions.
    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:基孔肯雅病毒(CHIKV)已重新成为主要的公共卫生问题,导致基孔肯雅热,病例增加和神经系统并发症。
    方法:在本研究中,我们调查了CHIKV株LK(EH)CH6708的东/中/南非(ECSA)系的低传代人类分离株,该分离株表现出混合有小的和大的病毒噬斑.小斑块和大斑块变体被分离并命名为CHIKV-SP和CHIKV-BP,分别。在体外和体内表征CHIKV-SP和CHIKV-BP以比较它们的病毒产生和毒力。此外,全病毒基因组分析和反向遗传学用于鉴定基因组毒力因子.
    结果:CHIKV-SP在哺乳动物细胞中表现出较低的病毒产生,并且在鼠模型中表现出减弱的毒力。另一方面,CHIKV-BP诱导更高的促炎细胞因子水平,损害了血脑屏障的完整性,并导致小鼠大脑中的星形胶质细胞感染。此外,CHIKV-SP变种在白纹伊蚊中的传播潜力有限,可能是由于传播受限。全病毒基因组分析揭示了CHIKV-SP变异中的多种基因突变,包括在病毒E2糖蛋白的第55位的甘氨酸(G)到精氨酸(R)突变。反向遗传学实验证实,单独的E2-G55R突变足以降低体外病毒产生和小鼠的毒力。
    结论:这些发现强调了E2-G55R突变对CHIKV致病性和神经毒力的减弱作用,并强调了在自然感染中监测该突变的重要性。
    BACKGROUND: Chikungunya virus (CHIKV) has reemerged as a major public health concern, causing chikungunya fever with increasing cases and neurological complications.
    METHODS: In the present study, we investigated a low-passage human isolate of the East/ Central/South African (ECSA) lineage of CHIKV strain LK(EH)CH6708, which exhibited a mix of small and large viral plaques. The small and large plaque variants were isolated and designated as CHIKV-SP and CHIKV-BP, respectively. CHIKV-SP and CHIKV-BP were characterized in vitro and in vivo to compare their virus production and virulence. Additionally, whole viral genome analysis and reverse genetics were employed to identify genomic virulence factors.
    RESULTS: CHIKV-SP demonstrated lower virus production in mammalian cells and attenuated virulence in a murine model. On the other hand, CHIKV-BP induced higher pro-inflammatory cytokine levels, compromised the integrity of the blood-brain barrier, and led to astrocyte infection in mouse brains. Furthermore, the CHIKV-SP variant had limited transmission potential in Aedes albopictus mosquitoes, likely due to restricted dissemination. Whole viral genome analysis revealed multiple genetic mutations in the CHIKV-SP variant, including a Glycine (G) to Arginine (R) mutation at position 55 in the viral E2 glycoprotein. Reverse genetics experiments confirmed that the E2-G55R mutation alone was sufficient to reduce virus production in vitro and virulence in mice.
    CONCLUSIONS: These findings highlight the attenuating effects of the E2-G55R mutation on CHIKV pathogenicity and neurovirulence and emphasize the importance of monitoring this mutation in natural infections.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号