herpes simplex virus type 1 (HSV-1)

  • 文章类型: Journal Article
    单纯疱疹病毒1型(HSV-1),一种嗜神经DNA病毒,在神经组织中建立潜伏期,再激活会导致严重的后果,比如脑炎。新出现的证据将HSV-1感染与慢性神经炎症和神经退行性疾病联系起来。小胶质细胞,中枢神经系统(CNS)免疫前哨,表达不同的受体,包括α7烟碱乙酰胆碱受体(α7nAChRs),对免疫调节至关重要。最近的研究表明,α7nAChR激活可以防止病毒感染。这里,我们显示α7nAChR激动剂,胆碱和PNU-282987显著抑制小胶质细胞BV2中HSV-1的复制。值得注意的是,这种抑制作用独立于传统的离子型nAChR信号通路。mRNA分析显示,胆碱刺激抗病毒因子的表达,IL-1β和Nos2下调BV2细胞凋亡基因和A型Lamins。这些发现提示了小胶质细胞α7nAChRs通过调节先天免疫应答来限制病毒感染的新机制。
    Herpes simplex virus type 1 (HSV-1), a neurotropic DNA virus, establishes latency in neural tissues, with reactivation causing severe consequences like encephalitis. Emerging evidence links HSV-1 infection to chronic neuroinflammation and neurodegenerative diseases. Microglia, the central nervous system\'s (CNS) immune sentinels, express diverse receptors, including α7 nicotinic acetylcholine receptors (α7 nAChRs), critical for immune regulation. Recent studies suggest α7 nAChR activation protects against viral infections. Here, we show that α7 nAChR agonists, choline and PNU-282987, significantly inhibit HSV-1 replication in microglial BV2 cells. Notably, this inhibition is independent of the traditional ionotropic nAChR signaling pathway. mRNA profiling revealed that choline stimulates the expression of antiviral factors, IL-1β and Nos2, and down-regulates the apoptosis genes and type A Lamins in BV2 cells. These findings suggest a novel mechanism by which microglial α7 nAChRs restrict viral infections by regulating innate immune responses.
    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
    单纯疱疹病毒1型(HSV-1)被认为是一种常见的病毒病原体,可以感染身体的多个部位,导致各种临床表现。根据这种多样化的表现,证明了许多细胞类型中的HSV-1感染。除了HSV-1细胞嗜性,例如,成纤维细胞,上皮,粘膜细胞,和神经元,HSV-1感染可发生在人类T淋巴细胞中,特别是在活化的T细胞中。此外,一些研究发现,肌动蛋白聚合和丝足形成支持不同细胞类型的HSV-1感染。因此,这篇综述的目的是探讨HSV-1感染涉及丝状伪足形成的各种类型细胞的机制,并强调HSV-1进入相关研究的潜在未来方向。此外,这篇综述涵盖了几种可能的抗HSV药物的策略,重点是进入步骤,提供潜在治疗干预措施的见解。
    Herpes simplex virus type 1 (HSV-1) has been known as a common viral pathogen that can infect several parts of the body, leading to various clinical manifestations. According to this diverse manifestation, HSV-1 infection in many cell types was demonstrated. Besides the HSV-1 cell tropism, e.g., fibroblast, epithelial, mucosal cells, and neurons, HSV-1 infections can occur in human T lymphocyte cells, especially in activated T cells. In addition, several studies found that actin polymerization and filopodia formation support HSV-1 infection in diverse cell types. Hence, the goal of this review is to explore the mechanism of HSV-1 infection in various types of cells involving filopodia formation and highlight potential future directions for HSV-1 entry-related research. Moreover, this review covers several strategies for possible anti-HSV drugs focused on the entry step, offering insights into potential 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
    单纯疱疹性角膜炎(HSK)是由单纯疱疹病毒1型(HSV-1)感染引起的常见致盲性角膜疾病。抗病毒药物和皮质类固醇没有显示出足够的治疗效果。在HSV-1感染的早期阶段,巨噬细胞是第一道防线。特别是,CD169+巨噬细胞在吞噬作用和抗原呈递中起重要作用。因此,我们建造了GM-gD-lip,神经节苷脂GM1脂质体疫苗封装HSV-1糖蛋白D并靶向CD169+巨噬细胞。结膜下注射疫苗后,我们评估了HSK小鼠的存活率和眼表病变,以及泪液中的病毒水平,角膜,和三叉神经节.我们发现GM-gD-lip降低HSV-1病毒载量并减轻HSK的临床严重程度。GM-gD-lip也增加了角膜浸润巨噬细胞的数量,尤其是CD169+巨噬细胞,并将它们极化为M1。此外,眼表引流淋巴结中树突状细胞(DCs)和CD8+T细胞的数量显著增加。这些发现表明,GM-gD-lip将CD169+巨噬细胞极化为M1以消除病毒,同时通过DC将抗原交叉呈递至CD8+T细胞以激活适应性免疫,最终减轻HSK的严重程度。使用GM-gD-lip作为治疗HSK的免疫治疗方法具有重要意义。
    Herpes simplex keratitis (HSK) is a common blinding corneal disease caused by herpes simplex virus type 1 (HSV-1) infection. Antiviral drugs and corticosteroids haven\'t shown adequate therapeutic efficacy. During the early stage of HSV-1 infection, macrophages serve as the first line of defense. In particular, CD169+ macrophages play an important role in phagocytosis and antigen presentation. Therefore, we constructed GM-gD-lip, a ganglioside GM1 liposome vaccine encapsulating HSV-1 glycoprotein D and targeting CD169+ macrophages. After subconjunctival injection of the vaccine, we evaluated the survival rate and ocular surface lesions of the HSK mice, as well as the virus levels in the tear fluid, corneas, and trigeminal ganglia. We discovered that GM-gD-lip reduced HSV-1 viral load and alleviated the clinical severity of HSK. The GM-gD-lip also increased the number of corneal infiltrating macrophages, especially CD169+ macrophages, and polarized them toward M1. Furthermore, the number of dendritic cells (DCs) and CD8+ T cells in the ocular draining lymph nodes was significantly increased. These findings demonstrated that GM-gD-lip polarized CD169+ macrophages toward M1 to eliminate the virus while cross-presenting antigens to CD8+ T cells via DCs to activate adaptive immunity, ultimately attenuating the severity of HSK. The use of GM-gD-lip as an immunotherapeutic method for the treatment of HSK has significant implications.
    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
    人疱疹病毒是具有在人群中高度流行的双链线性DNA基因组的包膜病毒。这些病毒被细分为三个亚家族,即α疱疹病毒(单纯疱疹病毒1型,HSV-1;单纯疱疹病毒2型,HSV-2;和水痘-带状疱疹病毒,VZV),betaherpesvirinae(人巨细胞病毒,HCMV;人类疱疹病毒6,HHV-6;和人类疱疹病毒7,HHV-7)和γ-疱疹病毒(EB病毒,EBV和卡波西肉瘤相关疱疹病毒,KSHV)。除了编码许多分子决定子逃避宿主的抗病毒反应,这些病毒还调节细胞代谢过程以促进其复制。这里,我们回顾并讨论了描述碳水化合物代谢和疱疹病毒复制周期之间相互作用的现有研究,基于这些可用于阻断疱疹病毒感染的相互作用,共同突出潜在的新分子靶标。
    Human herpesviruses are enveloped viruses with double-stranded linear DNA genomes highly prevalent in the human population. These viruses are subdivided into three subfamilies, namely alphaherpesvirinae (herpes simplex virus type 1, HSV-1; herpes simplex virus type 2, HSV-2; and varicella-zoster virus, VZV), betaherpesvirinae (human cytomegalovirus, HCMV; human herpesvirus 6, HHV-6; and human herpesvirus 7, HHV-7) and gammaherpesvirinae (Epstein-Barr virus, EBV; and Kaposi\'s sarcoma-associated herpesvirus, KSHV). Besides encoding numerous molecular determinants to evade the host antiviral responses, these viruses also modulate cellular metabolic processes to promote their replication. Here, we review and discuss existing studies describing an interplay between carbohydrate metabolism and the replication cycle of herpesviruses, altogether highlighting potentially new molecular targets based on these interactions that could be used to block herpesvirus 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
    人类免疫缺陷病毒(HIV)引起的艾滋病每年在全球范围内引起大量感染和死亡,仍然没有疫苗可以预防感染。基于编码其他病原体靶蛋白的重组1型单纯疱疹病毒(HSV-1)载体的疫苗已广泛用于疾病控制。这里,HIV-1gp160基因整合到内部反向(IR)区域缺失的HSV-1载体(HSV-BAC)中的重组病毒,通过细菌人工染色体(BAC)技术获得,及其在BALB/c小鼠中的免疫原性研究。结果表明,基于HSV-BAC的重组病毒的复制能力与野生型相似。此外,体液和细胞免疫反应显示出腹膜内(IP)给药的优越性,与鼻内(IN)相比,皮下(SC)和肌肉内(IM),这通过显著的抗体和T细胞应答的产生来证明。更重要的是,在prime-boost组合研究小鼠模型中,与类似疫苗接种方案中的单一病毒或蛋白质疫苗接种相比,重组病毒引发后HIV-1VLP加强可诱导更强和更广泛的免疫应答.抗体生产是足够的,具有巨大的潜在的病毒清除,以及有效的T细胞激活,通过酶联免疫吸附测定(ELISA)和流式细胞术(FC)进行评估。总的来说,这些发现揭示了组合不同疫苗载体和方式以提高针对不同HIV-1抗原的免疫原性和广度的价值.
    Human immunodeficiency virus (HIV) induced AIDS causes a large number of infections and deaths worldwide every year, still no vaccines are available to prevent infection. Recombinant herpes simplex virus type 1 (HSV-1) vector-based vaccines coding the target proteins of other pathogens have been widely used for disease control. Here, a recombinant virus with HIV-1 gp160 gene integration into the internal reverse (IR) region-deleted HSV-1 vector (HSV-BAC), was obtained by bacterial artificial chromosome (BAC) technology, and its immunogenicity investigated in BALB/c mice. The result showed similar replication ability of the HSV-BAC-based recombinant virus and wild type. Furthermore, humoral and cellular immune response showed superiority of intraperitoneal (IP) administration, compared to intranasally (IN), subcutaneous (SC) and intramuscularly (IM), that evidenced by production of significant antibody and T cell responses. More importantly, in a prime-boost combination study murine model, the recombinant viruses prime followed by HIV-1 VLP boost induced stronger and broader immune responses than single virus or protein vaccination in a similar vaccination regimen. Antibody production was sufficient with huge potential for viral clearance, along with efficient T-cell activation, which were evaluated by the enzyme-linked immunosorbent assay (ELISA) and flow cytometry (FC). Overall, these findings expose the value of combining different vaccine vectors and modalities to improve immunogenicity and breadth against different HIV-1 antigens.
    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
    单纯疱疹病毒1型(HSV-1)导致全球终身感染,目前还没有有效的治疗方法或疫苗。HSV-1衍生工具,如神经元回路示踪剂和溶瘤病毒,已被广泛使用;然而,HSV-1的进一步基因工程受到其复杂基因组结构的阻碍。在本研究中,我们设计并构建了基于H129-G4的HSV-1合成平台。在酵母中使用转化相关重组(TAR)通过3轮合成从10个片段构建完整的基因组,被命名为H129-Syn-G2.H129-Syn-G2基因组含有两个拷贝的gfp基因,并被转染到细胞中以拯救病毒。根据生长曲线测定和电镜结果,与亲本病毒相比,合成病毒表现出更优化的生长特性和相似的形态发生。这个合成平台将有助于进一步操纵HSV-1基因组,以开发神经元回路示踪剂。溶瘤病毒,和疫苗。
    Herpes simplex virus type 1 (HSV-1) causes lifelong infections worldwide, and currently there is no efficient cure or vaccine. HSV-1-derived tools, such as neuronal circuit tracers and oncolytic viruses, have been used extensively; however, further genetic engineering of HSV-1 is hindered by its complex genome structure. In the present study, we designed and constructed a synthetic platform for HSV-1 based on H129-G4. The complete genome was constructed from 10 fragments through 3 rounds of synthesis using transformation-associated recombination (TAR) in yeast, and was named H129-Syn-G2. The H129-Syn-G2 genome contained two copies of the gfp gene and was transfected into cells to rescue the virus. According to growth curve assay and electron microscopy results, the synthetic viruses exhibited more optimized growth properties and similar morphogenesis compared to the parental virus. This synthetic platform will facilitate further manipulation of the HSV-1 genome for the development of neuronal circuit tracers, oncolytic viruses, and vaccines.
    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)是一种严重的,由单纯疱疹病毒1型(HSV-1)感染引起的感染性角膜疾病。阿昔洛韦耐药的患病率越来越高,激素类药物的副作用,手术后复发的容易性使得开发治疗HSK的新方法变得至关重要。HSV-1通过各种机制逃避宿主免疫应答。因此,我们探讨了免疫原性细胞死亡诱导因子PKHB1肽在HSK中的作用。结膜下注射PKHB1肽后,我们观察了HSK小鼠的眼表病变和存活,并检测了泪液中的病毒水平,角膜,和三叉神经节.我们发现PKHB1肽降低了眼中的HSV-1水平并减轻了HSK的严重程度。此外,它增加了角膜浸润抗原呈递细胞(APC)的数量,如巨噬细胞和树突状细胞,和眼引流淋巴结中的CD8+T细胞。我们进一步观察到PKHB1肽促进钙网蛋白的暴露,以及体外HSV-1感染细胞中ATP和高迁移率族蛋白1的释放。我们的发现表明,PKHB1肽通过诱导感染细胞释放大量与损伤相关的分子模式来促进APC的募集和成熟。然后APC吞噬抗原物质并转移到淋巴结,引发细胞毒性T淋巴细胞依赖性免疫反应,最终缓解HSK。
    Herpes simplex keratitis (HSK) is a severe, infectious corneal disease caused by herpes simplex virus type 1 (HSV-1) infection. The increasing prevalence of acyclovir resistance, the side effects of hormonal drugs, and the ease of recurrence after surgery have made it crucial to develop new methods of treating HSK. HSV-1 evades the host immune response through various mechanisms. Therefore, we explored the role of the immunogenic cell death inducer PKHB1 peptide in HSK. After subconjunctival injection of PKHB1 peptide, we observed the ocular surface lesions and survival of HSK mice and detected the virus levels in tear fluid, corneas, and trigeminal ganglions. We found that PKHB1 peptide reduced HSV-1 levels in the eye and alleviated the severity of HSK. Moreover, it increased the number of corneal infiltrating antigen-presenting cells (APCs), such as macrophages and dendritic cells, and CD8+ T cells in ocular draining lymph nodes. We further observed that PKHB1 peptide promoted the exposure of calreticulin, as well as the release of ATP and high-mobility group box 1 in HSV-1-infected cells in vitro. Our findings suggested that PKHB1 peptide promoted the recruitment and maturation of APCs by inducing the release of large amounts of damage-associated molecular patterns from infected cells. APCs then phagocytized antigenic materials and translocated to the lymph nodes, triggering a cytotoxic T lymphocyte-dependent immune response that ultimately alleviated HSK.
    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
    单纯疱疹病毒1型(HSV-1)是唯一被FDA和EMA批准的溶瘤病毒,因此,许多潜在的溶瘤HSV(oHSV)正在临床开发中。利用的oHSV亲本菌株是,然而,主要基于实验室参考菌株,与HSV-1的循环菌株相比,其可能具有受损的细胞溶解能力。这里,我们评估了来自芬兰的36种循环HSV-1菌株的表型,以揭示它们作为oHSV骨架的潜力.首先,我们确定了它们的细胞间扩散能力和细胞间扩散能力,找到具有对每种应用有利的复制谱的菌株。第二,来展现差异,我们研究了两种相关病毒糖蛋白(gB/UL27,gI/US7)的遗传多样性。第三,我们检查了代表神经胶质瘤的细胞中菌株的溶瘤潜力,淋巴瘤和结直肠腺癌。我们的结果表明,循环分离株的表型,包括溶瘤潜能,与宿主细胞类型高度相关。然而,我们在许多或所有研究的癌细胞类型中鉴定出与参考病毒相比具有更高的溶瘤潜能的分离株.我们的研究强调在早期载体设计中需要仔细选择骨干病毒,并强调了临床分离株在oHSV发展中作为骨干的潜力。
    Herpes simplex virus type 1 (HSV-1) is the only FDA- and EMA- approved oncolytic virus, and accordingly, many potential oncolytic HSVs (oHSV) are in clinical development. The utilized oHSV parental strains are, however, mostly based on laboratory reference strains, which may possess a compromised cytolytic capacity in contrast to circulating strains of HSV-1. Here, we assess the phenotype of thirty-six circulating HSV-1 strains from Finland to uncover their potential as oHSV backbones. First, we determined their capacity for cell-to-cell versus extracellular spread, to find strains with replication profiles favorable for each application. Second, to unfold the differences, we studied the genetic diversity of two relevant viral glycoproteins (gB/UL27, gI/US7). Third, we examined the oncolytic potential of the strains in cells representing glioma, lymphoma, and colorectal adenocarcinoma. Our results suggest that the phenotype of a circulating isolate, including the oncolytic potential, is highly related to the host cell type. Nevertheless, we identified isolates with increased oncolytic potential in comparison with the reference viruses across many or all of the studied cancer cell types. Our research emphasizes the need for careful selection of the backbone virus in early vector design, and it highlights the potential of clinical isolates as backbones in oHSV development.
    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
    皮质类固醇拮抗剂损害单纯疱疹病毒1(HSV-1)生产性感染和外植体诱导的潜伏期再激活,提示皮质类固醇和糖皮质激素受体(GR)介导这些复杂的病毒-宿主相互作用的某些方面。GR-激素复合物正向和负向调节转录,在某种程度上,通过结合GR响应元件(GREs)。最近的研究显示感染的细胞蛋白0(ICP0),ICP4和ICP27启动子/顺式调节模块(CRM)被GR和Krüppel样因子15(KLF15)协同激活,形成前馈转录环。我们假设ICP0启动子含有被GR反式激活的独立CRMs,KLF15和合成皮质类固醇地塞米松(DEX)。这一假设是基于ICP0启动子含有多个转录因子结合位点的发现,GR和KLF15共同反式激活全长ICP0启动子。跨越-800至-635(片段A)的ICP0启动子序列被GR有效地反式激活,猴肾细胞(Vero)中的KLF15和DEX,而GR和DEX显著增强小鼠神经母细胞瘤细胞(Neuro-2A)中的启动子活性。此外,ICP0片段B(-458至-635)被GR有效地反式激活,KLF15和DEX在Vero细胞中,但不是Neuro-2A细胞.最后,片段D(-232至-24)在Vero细胞中被GR显著激活,KLF15和DEX,而KLF15和DEX足以在Neuro-2A细胞中进行反式激活。总的来说,这些研究揭示了GR对ICP0启动子内三个独立的CRM的有效反式激活,KLF15和/或DEX。最后,含有特异性蛋白1(Sp1)结合位点的富含GC的序列对于反式激活至关重要。
    A corticosteroid antagonist impairs Herpes Simplex Virus 1 (HSV-1) productive infection and explant-induced reactivation from latency, suggesting corticosteroids and the glucocorticoid receptor (GR) mediate certain aspects of these complex virus-host interactions. GR-hormone complexes regulate transcription positively and negatively, in part, by binding GR response elements (GREs). Recent studies revealed infected cell protein 0 (ICP0), ICP4, and ICP27 promoter/cis-regulatory modules (CRMs) are cooperatively transactivated by GR and Krüppel-like factor 15 (KLF15), which forms a feed-forward transcription loop. We hypothesized the ICP0 promoter contains independent CRMs that are transactivated by GR, KLF15, and the synthetic corticosteroid dexamethasone (DEX). This hypothesis is based on the finding that the ICP0 promoter contains multiple transcription factor binding sites, and GR and KLF15 cooperatively transactivate the full-length ICP0 promoter. ICP0 promoter sequences spanning -800 to -635 (fragment A) were efficiently transactivated by GR, KLF15, and DEX in monkey kidney cells (Vero), whereas GR and DEX significantly enhanced promoter activity in mouse neuroblastoma cells (Neuro-2A). Furthermore, ICP0 fragment B (-458 to -635) was efficiently transactivated by GR, KLF15, and DEX in Vero cells, but not Neuro-2A cells. Finally, fragment D (-232 to -24) was transactivated significantly in Vero cells by GR, KLF15, and DEX, whereas KLF15 and DEX were sufficient for transactivation in Neuro-2A cells. Collectively, these studies revealed efficient transactivation of three independent CRMs within the ICP0 promoter by GR, KLF15, and/or DEX. Finally, GC-rich sequences containing specificity protein 1 (Sp1) binding sites were essential for transactivation.
    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
    单纯疱疹病毒1型(HSV-1)是一种人类病毒,可在全世界范围内引起终生感染。HSV-1从三叉神经节(TG)潜伏期的复发是该病毒所见发病率的触发因素。除了引起发热水泡和唇疱疹,偶尔,这种病毒也会引起角膜损伤,导致未经治疗的人失明。几种宿主细胞蛋白在眼HSV-1感染中起重要作用。HSV-1通过其与细胞表面受体的相互作用进入角膜上皮细胞。并行,Toll样受体感知病毒入侵并激活防御机制以抵抗感染。新数据显示,Optineurin,宿主自噬受体也被激活以降解病毒颗粒。相比之下,乙酰肝素酶的激活,一种宿主酶,诱导免疫炎症反应,这触发了促炎和促血管生成的环境,并最终导致许多与角膜HSV-1感染有关的临床特征。很少,HSV-1也可以传播到中枢神经系统,引起严重的疾病。在这次审查中,我们总结了有关促进眼疱疹病理生理方面的宿主分子的最新知识。
    Herpes simplex virus type-1 (HSV-1) is a human virus that causes lifelong infections in a large population worldwide. Recurrence of HSV-1 from latency in trigeminal ganglion (TG) is the trigger of the morbidities seen with this virus. In addition to causing fever blisters and cold sores, occasionally the virus can also cause corneal lesions resulting in blindness in untreated individuals. Several host cell proteins play important roles in HSV-1 infection of the eye. HSV-1 enters into the corneal epithelial cells via its interactions with cell surface receptors. In parallel, the Toll-like receptors sense viral invasion and activate defense mechanisms to fight the infection. New data shows that Optineurin, a host autophagy receptor is also activated to degrade viral particles. In contrast, activation of heparanase, a host enzyme, induces an immune-inflammatory response, which triggers pro-inflammatory and pro-angiogenic environment and ultimately results in many of the clinical features seen with HSV-1 infection of the cornea. Rarely, HSV-1 can also spread to the central nervous system causing serious diseases. In this review, we summarize the latest knowledge on host molecules that promote pathophysiological aspects of ocular herpes.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号