Ectromelia, Infectious

  • 文章类型: Journal Article
    Ectromelia病毒(ECTV)是鼠痘的病原体。它为研究正痘病毒的免疫生物学提供了合适的模型,包括它们与宿主细胞骨架的相互作用。作为专业的抗原呈递细胞,树突状细胞(DC)控制细胞环境,捕获抗原,并在迁移到次级淋巴器官后将其呈现给T淋巴细胞。未成熟DC的迁移是可能的,由于存在专门的粘附结构,如后体或粘着斑(FA)。由于粘性结构的组装和拆卸与DC免疫调节和迁移功能高度相关,我们评估了ECTV感染如何靶向天然宿主骨髓来源的DC(BMDC)中的足细胞体和FAs的组织和形成。我们发现ECTV在感染的早期诱导足细胞迅速溶解,伴随着比未感染的对照细胞更大和更宽的FA的发展。在感染的后期,FAs主要在长细胞延伸中观察到,由感染细胞广泛形成。在伤口愈合试验中,感染ECTV的BMDCs中的podoses的溶解与成熟和2D细胞迁移增加无关;然而,观察到ECTV感染的细胞向源自LPS条件的BMDC的上清液加速的transwell迁移。我们建议ECTV引起的DC中粘附结构的空间组织变化可能会在某些条件下改变DC的粘附性/迁移。例如,炎症。
    Ectromelia virus (ECTV) is a causative agent of mousepox. It provides a suitable model for studying the immunobiology of orthopoxviruses, including their interaction with the host cell cytoskeleton. As professional antigen-presenting cells, dendritic cells (DCs) control the pericellular environment, capture antigens, and present them to T lymphocytes after migration to secondary lymphoid organs. Migration of immature DCs is possible due to the presence of specialized adhesion structures, such as podosomes or focal adhesions (FAs). Since assembly and disassembly of adhesive structures are highly associated with DCs\' immunoregulatory and migratory functions, we evaluated how ECTV infection targets podosomes and FAs\' organization and formation in natural-host bone marrow-derived DCs (BMDC). We found that ECTV induces a rapid dissolution of podosomes at the early stages of infection, accompanied by the development of larger and wider FAs than in uninfected control cells. At later stages of infection, FAs were predominantly observed in long cellular extensions, formed extensively by infected cells. Dissolution of podosomes in ECTV-infected BMDCs was not associated with maturation and increased 2D cell migration in a wound healing assay; however, accelerated transwell migration of ECTV-infected cells towards supernatants derived from LPS-conditioned BMDCs was observed. We suggest that ECTV-induced changes in the spatial organization of adhesive structures in DCs may alter the adhesiveness/migration of DCs during some conditions, e.g., inflammation.
    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
    常规树突状细胞(cDC)是先天免疫细胞,在诱导抗病毒适应性免疫反应中起关键作用,因为它们具有非凡的能力来启动和极化原始T细胞进入不同的效应T辅助T(Th)亚群。cDCs的两个主要亚群,cDC1(小鼠中的CD8α+和人中的CD141+)和cDC2(小鼠中的CD11b+和人中的CD1c+),可以优先将T细胞极化为Th1和Th2表型,分别。在感染外胚层病毒(ECTV)期间,一种来自痘病毒科的正痘病毒,适当的Th免疫反应的时机和激活有助于近交小鼠品系对致死形式的鼠痘的抗性(Th1)或易感性(Th2)。由于cDC亚群在调节特定免疫应答质量方面的高度可塑性和多样性特性,在本研究中,我们比较了源自不同ECTV感染小鼠品系的脾cDC1和cDC2成熟的能力,激活,并使鼠痘期间的免疫反应极化。我们的结果表明,在鼠痘的早期阶段,在体内ECTV感染后,来自耐药C57BL/6和易感BALB/c小鼠的cDC亚群均被激活。这些细胞表现出升高水平的表面MHCI类和II类,和共刺激分子,并显示出增强的产生细胞因子的潜力。然而,BALB/c小鼠的cDC亚群均显示出比C57BL/6小鼠更高的成熟状态。尽管他们的激活状态较高,来自易感小鼠的cDC1和cDC2产生少量的Th1极化细胞因子,包括IL-12和IFN-γ,这些细胞刺激同种异体CD4+T细胞增殖和Th1极化的能力严重受损。相比之下,与来自BALB/c小鼠的cDC相比,来自抗性小鼠的两个cDC亚群都产生了大量的Th1极化细胞因子,并且在将同种异体T细胞分化为Th1细胞方面表现出更强的能力.总的来说,我们的结果表明,在鼠痘的早期阶段,来自抗性小鼠品系的脾cDC亚群可以比易感品系更好地引发Th1细胞介导的反应,可能有助于诱导控制病毒传播和从ECTV攻击中生存所必需的保护性免疫反应。
    Conventional dendritic cells (cDCs) are innate immune cells that play a pivotal role in inducing antiviral adaptive immune responses due to their extraordinary ability to prime and polarize naïve T cells into different effector T helper (Th) subsets. The two major subpopulations of cDCs, cDC1 (CD8α+ in mice and CD141+ in human) and cDC2 (CD11b+ in mice and CD1c+ in human), can preferentially polarize T cells toward a Th1 and Th2 phenotype, respectively. During infection with ectromelia virus (ECTV), an orthopoxvirus from the Poxviridae family, the timing and activation of an appropriate Th immune response contributes to the resistance (Th1) or susceptibility (Th2) of inbred mouse strains to the lethal form of mousepox. Due to the high plasticity and diverse properties of cDC subpopulations in regulating the quality of a specific immune response, in the present study we compared the ability of splenic cDC1 and cDC2 originating from different ECTV-infected mouse strains to mature, activate, and polarize the Th immune response during mousepox. Our results demonstrated that during early stages of mousepox, both cDC subsets from resistant C57BL/6 and susceptible BALB/c mice were activated upon in vivo ECTV infection. These cells exhibited elevated levels of surface MHC class I and II, and co-stimulatory molecules and showed enhanced potential to produce cytokines. However, both cDC subsets from BALB/c mice displayed a higher maturation status than that of their counterparts from C57BL/6 mice. Despite their higher activation status, cDC1 and cDC2 from susceptible mice produced low amounts of Th1-polarizing cytokines, including IL-12 and IFN-γ, and the ability of these cells to stimulate the proliferation and Th1 polarization of allogeneic CD4+ T cells was severely compromised. In contrast, both cDC subsets from resistant mice produced significant amounts of Th1-polarizing cytokines and demonstrated greater capability in differentiating allogeneic T cells into Th1 cells compared to cDCs from BALB/c mice. Collectively, our results indicate that in the early stages of mousepox, splenic cDC subpopulations from the resistant mouse strain can better elicit a Th1 cell-mediated response than the susceptible strain can, probably contributing to the induction of the protective immune responses necessary for the control of virus dissemination and for survival from ECTV challenge.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: English Abstract
    BACKGROUND: The mouse-specific orthopoxvirus, ectromelia virus, is one of the best models that can be used to study key issues of pathogenesis, prevention, and treatment of smallpox, and to develop measures to increase virulence, transmissibility, or the ability to overcome vaccine immunity. The aim of the work is to screen the antiviral activity of samples from Inonotus obliquus chaga and humic acid from brown coal in vitro against ectromelia virus.
    METHODS: We used ectromelia virus, strain K-1 (reg. No V-142), obtained from the State Collection of Pathogens of Viral Infections and Rickettsioses of the State Scientific Center of Virology and Biotechnology \"Vector\"; Vero Е6 cell culture (No 70) from the Collection of cell cultures of the State Scientific Center of Virology and Biotechnology \"Vector\". Nine samples from chaga I. obliquus and humic acid from brown coal were used to evaluate the changes in the infectivity of the ectromelia virus on cell culture using 2 schemes of application of drugs and virus (preventive and therapeutic schemes), and to assess their cytotoxicity and antiviral activity.
    RESULTS: 50% cytotoxic concentration, 50% virus-inhibiting concentrations and selectivity index were determined for all samples. The studied samples were shown to be non-toxic to the monolayer of Vero cell culture in a dilution of 300 and more micrograms/ml, while demonstrated high antiviral activity against strain K-1 of ectromelia virus in two application schemes - preventive and curative.
    CONCLUSIONS: All samples tested for ectromelia virus in vitro can be considered promising for further development of drugs against diseases caused by orthopoxviruses.
    Введение. Специфичный для мыши ортопоксвирус – вирус эктромелии – является одной из лучших моделей, которую можно использовать для изучения основных вопросов патогенеза, профилактики и лечения оспы, а также разработки мер для повышения вирулентности, трансмиссивности или способности преодолевать вакцинный иммунитет. Целью работы является скрининг противовирусной активности образцов из чаги Inonotus obliquus и гуминовой кислоты из бурого угля in vitro в отношении вируса эктромелии. Материалы и методы. В работе использовали вирус эктромелии, штамм К-1 (рег. номер V-142), полученный из Государственной коллекции возбудителей вирусных инфекций и риккетсиозов ГНЦ ВБ «Вектор» Роспотребнадзора; культуру клеток Vero Е6 (шифр коллекционный 70), полученную из Коллекции культур клеток ГНЦ ВБ «Вектор» Роспотребнадзора. Для 9 образцов из чаги I. obliquus и гуминовой кислоты из бурого угля была проведена оценка изменения инфекционности вируса эктромелии на культуре клеток при использовании двух схем внесения препаратов и вируса (профилактической и лечебной), а также определение их цитотоксичности и противовирусной активности. Результаты. Для всех образцов были определены 50% цитотоксическая концентрация, 50% эффективные дозы и химиотерапевтические индексы образцов. Было показано, что исследуемые образцы не являются токсичными для монослоя культуры клеток Vero Е6 в разведении 300 мкг/мл и более, продемонстрировали высокую противовирусную активность в отношении штамма К-1 вируса эктромелии в двух схемах применения – профилактической и лечебной. Заключение. Все образцы, проверенные в отношении вируса эктромелии in vitro, могут рассматриваться как перспективные для дальнейшей разработки препаратов против заболеваний, вызываемых ортопоксвирусами.
    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
    最近猴痘病毒在人类中的传播加剧了对正痘病毒感染的担忧。因此,对猴痘病毒的免疫生物学进行全面研究对于开发有效的疗法至关重要。Ectromelia病毒(ECTV)与猴痘病毒的遗传和疾病特征非常相似,使其成为研究正痘病毒与宿主相互作用的有价值的研究工具。鸟苷酸结合蛋白(GBPs),高表达干扰素刺激基因(ISGs),对各种胞内病原微生物有拮抗作用。我们先前的研究表明,GBP2对ECTV感染具有轻度但统计学上显着的抑制作用。痘病毒基因组中编码宿主免疫应答的大量分子的存在引发了关于它是否还包括抵消GBP2抗病毒活性的蛋白质的问题。使用IP/MS和co-IP技术,我们发现ECTV的poly(A)聚合酶催化亚基(PAPL)蛋白是与GBP2相互作用的病毒调节分子。进一步的研究表明,PAPL通过降低GBP2的蛋白质水平来拮抗GBP2的抗病毒活性。用CRISPR/Cas9系统敲除ECTV的PAPL基因会显著降低病毒的复制能力,说明了PAPL在ECTV复制过程中不可或缺的作用。总之,我们的研究提供了初步证据支持PAPL作为可与GBP2相互作用的毒力因子的重要性.
    The recent spread of the monkeypox virus among humans has heightened concerns regarding orthopoxvirus infections. Consequently, conducting a comprehensive study on the immunobiology of the monkeypox virus is imperative for the development of effective therapeutics. Ectromelia virus (ECTV) closely resembles the genetic and disease characteristics of monkeypox virus, making it a valuable research tool for studying orthopoxvirus-host interactions. Guanylate-binding proteins (GBPs), highly expressed interferon-stimulated genes (ISGs), have antagonistic effects against various intracellular pathogenic microorganisms. Our previous research has shown that GBP2 has a mild but statistically significant inhibitory effect on ECTV infection. The presence of a significant number of molecules in the poxvirus genome that encode the host immune response raises questions about whether it also includes proteins that counteract the antiviral activity of GBP2. Using IP/MS and co-IP technology, we discovered that the poly(A) polymerase catalytic subunit (PAPL) protein of ECTV is a viral regulatory molecule that interacts with GBP2. Further studies have shown that PAPL antagonizes the antiviral activity of GBP2 by reducing its protein levels. Knocking out the PAPL gene of ECTV with the CRISPR/Cas9 system significantly diminishes the replication ability of the virus, indicating the indispensable role of PAPL in the replication process of ECTV. In conclusion, our study presents preliminary evidence supporting the significance of PAPL as a virulence factor that can interact with GBP2.
    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
    受体相互作用蛋白激酶3(RIPK3)和混合谱系激酶结构域样假激酶(MLKL)是对坏死性凋亡至关重要的蛋白质,程序性细胞死亡的机制,当细胞凋亡被抑制时被激活,并被认为是抗病毒的。这里,我们调查了RIPK3和MLKL在控制正痘病毒异位病毒(ECTV)中的作用,老鼠的天然病原体。我们发现,缺乏RIPK3(Ripk3-/-)或MLKL(Mlkl-/-)的C57BL/6(B6)小鼠在低剂量腹膜内感染后对ECTV致死率的敏感性与野生型(WT)B6小鼠一样,并且通过自然足垫途径感染ECTV后与WTB6小鼠一样具有抗性。此外,足垫感染后,Mlkl-/-老鼠,但不是Ripk3-/-小鼠,在感染后三天的引流淋巴结(dLN)和感染后七天的脾脏或肝脏中,病毒滴度低于WT小鼠。尽管病毒控制有所改善,Mlkl-/-小鼠在干扰素或干扰素刺激的基因的表达或在向dLN募集自然杀伤(NK)细胞和炎性单核细胞(iMO)方面与WT小鼠没有差异。此外,Mlkl-/-和WT小鼠的CD8T细胞反应相似,即使在Mlkl-/-小鼠的DLN中,专职抗原呈递细胞感染更严重.最后,7dpi时Mlkl-/-和WT小鼠肝脏的组织病理学没有差异。因此,Mlkl-/-小鼠增强病毒控制的机制仍有待定义。重要性RIPK3和MLKL分子是坏死性细胞死亡所必需的,这被广泛认为是一种抗病毒机制。在这里,我们表明,缺乏RIPK3或MLKL的C57BL/6(B6)小鼠在低剂量腹膜内感染后与WTB6小鼠一样容易受到ECTV致死性的影响,并且在ECTV感染后与WTB6小鼠一样具有抗性通过自然脚垫途径。缺乏MLKL的小鼠在控制各种器官中的病毒载量方面比WT小鼠更有效。这种改进的病毒控制不是由于增强的干扰素,自然杀伤细胞,或CD8T细胞反应。总的来说,数据表明,对于坏死性凋亡至关重要的分子的缺乏不一定会导致病毒感染后更差的结局,并且可能会改善病毒控制.
    Receptor-interacting protein kinase 3 (RIPK3) and mixed lineage kinase domain-like pseudokinase (MLKL) are proteins that are critical for necroptosis, a mechanism of programmed cell death that is both activated when apoptosis is inhibited and thought to be antiviral. Here, we investigated the role of RIPK3 and MLKL in controlling the Orthopoxvirus ectromelia virus (ECTV), a natural pathogen of the mouse. We found that C57BL/6 (B6) mice deficient in RIPK3 (Ripk3-/-) or MLKL (Mlkl-/-) were as susceptible as wild-type (WT) B6 mice to ECTV lethality after low-dose intraperitoneal infection and were as resistant as WT B6 mice after ECTV infection through the natural footpad route. Additionally, after footpad infection, Mlkl-/- mice, but not Ripk3-/- mice, endured lower viral titers than WT mice in the draining lymph node (dLN) at three days postinfection and in the spleen or in the liver at seven days postinfection. Despite the improved viral control, Mlkl-/- mice did not differ from WT mice in the expression of interferons or interferon-stimulated genes or in the recruitment of natural killer (NK) cells and inflammatory monocytes (iMOs) to the dLN. Additionally, the CD8 T-cell responses in Mlkl-/- and WT mice were similar, even though in the dLNs of Mlkl-/- mice, professional antigen-presenting cells were more heavily infected. Finally, the histopathology in the livers of Mlkl-/- and WT mice at 7 dpi did not differ. Thus, the mechanism of the increased virus control by Mlkl-/- mice remains to be defined. IMPORTANCE The molecules RIPK3 and MLKL are required for necroptotic cell death, which is widely thought of as an antiviral mechanism. Here we show that C57BL/6 (B6) mice deficient in RIPK3 or MLKL are as susceptible as WT B6 mice to ECTV lethality after a low-dose intraperitoneal infection and are as resistant as WT B6 mice after ECTV infection through the natural footpad route. Mice deficient in MLKL are more efficient than WT mice at controlling virus loads in various organs. This improved viral control is not due to enhanced interferon, natural killer cell, or CD8 T-cell responses. Overall, the data indicate that deficiencies in the molecules that are critical to necroptosis do not necessarily result in worse outcomes following viral infection and may improve virus 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
    炎性单核细胞(iMO)和B细胞是小鼠淋巴结中痘病毒异位病毒(ECTV)的主要靶标,并在感染中发挥不同的作用。受感染和旁观者的IMO控制ECTV的系统传播,防止过早死亡,而B细胞产生抗体消除ECTV。我们的工作表明,在ECTV感染中幸存下来的受感染动物中,iMO和B细胞的内在和旁观者感染差异控制对免疫细胞功能重要的基因的转录,也许,细胞身份。旁观者细胞上调新陈代谢,抗原呈递,和干扰素刺激的基因。受感染的细胞下调许多细胞类型特异性基因,并上调非免疫细胞典型的转录本。旁观者(Bys)和感染的(Inf)iMO非冗余地有助于细胞因子环境和干扰素反应。此外,我们揭示了I型干扰素(IFN-I)或IFN-γ信号传导如何差异调节Inf和BysiMO的免疫途径,在稳定状态下,IFN-I启动iMO用于快速IFN-I产生和抗原呈递。
    Inflammatory monocytes (iMOs) and B cells are the main targets of the poxvirus ectromelia virus (ECTV) in the lymph nodes of mice and play distinct roles in surviving the infection. Infected and bystander iMOs control ECTV\'s systemic spread, preventing early death, while B cells make antibodies that eliminate ECTV. Our work demonstrates that within an infected animal that survives ECTV infection, intrinsic and bystander infection of iMOs and B cells differentially control the transcription of genes important for immune cell function and, perhaps, cell identity. Bystander cells upregulate metabolism, antigen presentation, and interferon-stimulated genes. Infected cells downregulate many cell-type-specific genes and upregulate transcripts typical of non-immune cells. Bystander (Bys) and infected (Inf) iMOs non-redundantly contribute to the cytokine milieu and the interferon response. Furthermore, we uncover how type I interferon (IFN-I) or IFN-γ signaling differentially regulates immune pathways in Inf and Bys iMOs and that, at steady state, IFN-I primes iMOs for rapid IFN-I production and antigen presentation.
    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
    Cytotoxic CD4 T lymphocytes (CD4-CTL) are important in antiviral immunity. For example, we have previously shown that in mice, CD4-CTL are important to control ectromelia virus (ECTV) infection. How viral infections induce CD4-CTL responses remains incompletely understood. We demonstrate here that not only ECTV but also vaccinia virus and lymphocytic choriomeningitis virus induce CD4-CTL, though the response to ECTV is stronger. Using ECTV, we also demonstrate that in contrast to CD8-CTL, CD4-CTL differentiation requires constant virus replication and ceases once the virus is controlled. We also show that major histocompatibility complex class II molecules on CD11c+ cells are required for CD4-CTL differentiation and for mousepox resistance. Transcriptional analysis indicated that antiviral CD4-CTL and noncytolytic T helper 1 (Th1) CD4 T cells have similar transcriptional profiles, suggesting that CD4-CTL are terminally differentiated classical Th1 cells. Interestingly, CD4-CTL and classical Th1 cells expressed similar mRNA levels of the transcription factors ThPOK and GATA-3, necessary for CD4 T cell linage commitment, and Runx3, required for CD8 T cell development and effector function. However, at the protein level, CD4-CTL had higher levels of the three transcription factors, suggesting that further posttranscriptional regulation is required for CD4-CTL differentiation. Finally, CRISPR/Cas9-mediated deletion of Runx3 in CD4 T cells inhibited CD4-CTL but not classical Th1 cell differentiation in response to ECTV infection. These results further our understanding of the mechanisms of CD4-CTL differentiation during viral infection and the role of posttranscriptionally regulated Runx3 in this process. IMPORTANCE While it is well established that cytotoxic CD4 T cells (CD4-CTLs) directly contribute to viral clearance, it remains unclear how CD4-CTL are induced. We now show that CD4-CTLs require sustained antigen presentation and are induced by CD11c-expressing antigen-presenting cells. Moreover, we show that CD4-CTLs are derived from the terminal differentiation of classical T helper 1 (Th1) subset of CD4 cells. Compared to Th1 cells, CD4-CTLs upregulate protein levels of the transcription factors ThPOK, Runx3, and GATA-3 posttranscriptionally. Deletion of Runx3 in differentiated CD4 T cells prevents induction of CD4-CTLs but not classical Th1 cells. These results advance our knowledge of how CD4-CTLs are induced during viral infection.
    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
    I型干扰素(IFN-I)是通过普遍存在的IFN-I受体(IFNAR)发出信号的抗病毒细胞因子。足垫感染外胚层病毒(ECTV)后,一种小鼠特异性病原体,C57BL/6(B6)小鼠无病存活,而广泛缺乏IFNAR的B6小鼠迅速死亡。我们现在向ECTV展示为了生存,只有造血细胞需要IFNAR表达。ECTV的存活特别需要在自然杀伤(NK)细胞和单核细胞中的IFNAR。然而,固有的IFNAR信号传导对于适应性免疫细胞应答或直接保护非造血细胞如肝细胞不是必需的,这是ECTV的主要目标。机械上,缺乏IFNAR的NK细胞具有降低的细胞溶解功能,而单核细胞中缺乏IFNAR会抑制IFN-I的产生并加速病毒的传播。因此,在致病性病毒感染期间,IFN-I通过在正反馈回路中刺激单核细胞和通过诱导NK细胞溶细胞功能来协调先天免疫。
    Type I interferons (IFN-I) are antiviral cytokines that signal through the ubiquitous IFN-I receptor (IFNAR). Following footpad infection with ectromelia virus (ECTV), a mouse-specific pathogen, C57BL/6 (B6) mice survive without disease, while B6 mice broadly deficient in IFNAR succumb rapidly. We now show that for survival to ECTV, only hematopoietic cells require IFNAR expression. Survival to ECTV specifically requires IFNAR in both natural killer (NK) cells and monocytes. However, intrinsic IFNAR signaling is not essential for adaptive immune cell responses or to directly protect non-hematopoietic cells such as hepatocytes, which are principal ECTV targets. Mechanistically, IFNAR-deficient NK cells have reduced cytolytic function, while lack of IFNAR in monocytes dampens IFN-I production and hastens virus dissemination. Thus, during a pathogenic viral infection, IFN-I coordinates innate immunity by stimulating monocytes in a positive feedback loop and by inducing NK cell cytolytic function.
    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
    Mitochondria are multitasking organelles that play a central role in energy production, survival and primary host defense against viral infections. Therefore, viruses target mitochondria dynamics and functions to benefit their replication and morphogenetic processes. We endeavor to understand the role of mitochondria during infection of ectromelia virus (ECTV), hence our investigations on mitochondria-related genes in non-immune (L929 fibroblasts) and immune (RAW 264.7 macrophages) cells. Our results show that during later stages of infection, ECTV significantly decreases the expression of mitochondria-related genes regulating many aspects of mitochondrial physiology and functions, including mitochondrial transport, small molecule transport, membrane polarization and potential, targeting proteins to mitochondria, inner membrane translocation, and apoptosis. Such down-regulation is cell-specific, since macrophages exhibited a more profound down-regulation of mitochondria-related genes compared to infected L929 fibroblasts. Only L929 cells exhibited up-regulation of two important genes responsible for oxidative phosphorylation and subsequent ATP production: Slc25a23 and Slc25a31. Changes in the expression of mitochondria-related genes are accompanied by altered mitochondria morphology and distribution in both types of cells. In depth Ingenuity Pathway Analysis (IPA) identified the \"Sirtuin Signaling Pathway\" as the most significant top canonical pathway associated with ECTV infection in both analyzed cell types. Taken together, down-regulation of mitochondria-related genes observed especially in macrophages indicates dysfunctional mitochondria, possibly contributing to energy collapse and induction of intrinsic pathway of apoptosis. Meanwhile, alteration of the expression of several mitochondria-related genes in fibroblasts without apoptosis induction may represent poxviral strategy to control cellular energy metabolism for efficient replication. Keywords: ectromelia virus; mitochondria; fibroblasts; macrophages.
    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
    Vaccination with vaccinia virus (VACV) elicits heterotypic immunity to smallpox, monkeypox, and mousepox, the mechanistic basis for which is poorly understood. It is generally assumed that heterotypic immunity arises from the presentation of a wide array of VACV-derived, CD8+ T cell epitopes that share homology with other poxviruses. Herein this assumption was tested using a large panel of VACV-derived peptides presented by HLA-B*07:02 (B7.2) molecules in a mousepox/ectromelia virus (ECTV)-infection, B7.2 transgenic mouse model. Most dominant epitopes recognized by ECTV- and VACV-reactive CD8+ T cells overlapped significantly without altering immunodominance hierarchy. Further, several epitopes recognized by ECTV-reactive CD8+ T cells were not recognized by VACV-reactive CD8+ T cells, and vice versa. In one instance, the lack of recognition owed to a N72K variation in the ECTV C4R70-78 variant of the dominant VACV B8R70-78 epitope. C4R70-78 does not bind to B7.2 and, hence, it was neither immunogenic nor antigenic. These findings provide a mechanistic basis for VACV vaccination-induced heterotypic immunity which can protect against Variola and Monkeypox disease. The understanding of how cross-reactive responses develop is essential for the rational design of a subunit-based vaccine that would be safe, and effectively protect against heterologous infection.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

公众号