首页 > 最新文献

Journal of Virology最新文献

英文 中文
Subnanometer structure of medusavirus capsid during maturation using cryo-electron microscopy. 利用低温电子显微镜观察介体病毒囊膜成熟过程中的亚纳米结构。
IF 4 2区 医学 Q2 VIROLOGY Pub Date : 2024-08-28 DOI: 10.1128/jvi.00436-24
Ryoto Watanabe, Chihong Song, Masaharu Takemura, Kazuyoshi Murata

Medusavirus is a giant virus classified into an independent family of Mamonoviridae. Amoebae infected with medusavirus release immature particles in addition to virions. These particles were suggested to exhibit the maturation process of this virus, but the structure of these capsids during maturation remains unknown. Here, we apply a block-based reconstruction method in cryo-electron microscopy (cryo-EM) single particle analysis to these viral capsids, extending the resolution to 7-10 Å. The maps reveal a novel network composed of minor capsid proteins (mCPs) supporting major capsid proteins (MCPs). A predicted molecular model of the MCP fitted into the cryo-EM maps clarified the boundaries between the MCP and the underlining mCPs, as well as between the MCP and the outer spikes, and identified molecular interactions between the MCP and these components. Several structural changes of the mCPs under the fivefold vertices of the immature particles were observed, depending on the presence or absence of the underlying internal membrane. In addition, the lower part of the penton proteins on the fivefold vertices was also missing in mature virions. These dynamic conformational changes of mCPs indicate an important function in the maturation process of medusavirus.IMPORTANCEThe structural changes of giant virus capsids during maturation have not thus far been well clarified. Medusavirus is a unique giant virus in which infected amoebae release immature particles in addition to mature virus particles. In this study, we used cryo-electron microscopy to investigate immature and mature medusavirus particles and elucidate the structural changes of the viral capsid during the maturation process. In DNA-empty particles, the conformation of the minor capsid proteins changed dynamically depending on the presence or absence of the underlying internal membranes. In DNA-full particles, the lower part of the penton proteins was lost. This is the first report of structural changes of the viral capsid during the maturation process of giant viruses.

Medusavirus 是一种巨型病毒,属于独立的 Mamonoviridae 科。感染了 Medusavirus 的变形虫除了释放病毒外,还会释放未成熟的颗粒。这些颗粒被认为展示了这种病毒的成熟过程,但这些囊壳在成熟过程中的结构仍然未知。在这里,我们将冷冻电子显微镜(cryo-EM)单颗粒分析中基于块的重建方法应用于这些病毒的囊膜,将分辨率提高到 7-10 Å。与低温电子显微镜图谱相匹配的 MCP 预测分子模型明确了 MCP 与底层 mCP 之间以及 MCP 与外层尖峰之间的界限,并确定了 MCP 与这些成分之间的分子相互作用。根据底层内膜的存在与否,观察到未成熟颗粒五倍顶点下的 mCP 发生了几种结构变化。此外,在成熟的病毒中,五倍顶点上的 penton 蛋白的下半部分也不见了。mCPs 的这些动态构象变化表明了介壳病毒在成熟过程中的重要功能。介壳病毒是一种独特的巨型病毒,受感染的变形虫除了释放成熟的病毒颗粒外,还会释放未成熟的颗粒。在这项研究中,我们利用低温电子显微镜研究了未成熟和成熟的髓病毒颗粒,并阐明了成熟过程中病毒帽的结构变化。在 DNA 空的颗粒中,次要囊膜蛋白的构象随底层内膜的有无而发生动态变化。在DNA饱满的颗粒中,penton蛋白的下半部分消失了。这是首次报道巨型病毒成熟过程中病毒外壳结构的变化。
{"title":"Subnanometer structure of medusavirus capsid during maturation using cryo-electron microscopy.","authors":"Ryoto Watanabe, Chihong Song, Masaharu Takemura, Kazuyoshi Murata","doi":"10.1128/jvi.00436-24","DOIUrl":"https://doi.org/10.1128/jvi.00436-24","url":null,"abstract":"<p><p>Medusavirus is a giant virus classified into an independent family of <i>Mamonoviridae</i>. Amoebae infected with medusavirus release immature particles in addition to virions. These particles were suggested to exhibit the maturation process of this virus, but the structure of these capsids during maturation remains unknown. Here, we apply a block-based reconstruction method in cryo-electron microscopy (cryo-EM) single particle analysis to these viral capsids, extending the resolution to 7-10 Å. The maps reveal a novel network composed of minor capsid proteins (mCPs) supporting major capsid proteins (MCPs). A predicted molecular model of the MCP fitted into the cryo-EM maps clarified the boundaries between the MCP and the underlining mCPs, as well as between the MCP and the outer spikes, and identified molecular interactions between the MCP and these components. Several structural changes of the mCPs under the fivefold vertices of the immature particles were observed, depending on the presence or absence of the underlying internal membrane. In addition, the lower part of the penton proteins on the fivefold vertices was also missing in mature virions. These dynamic conformational changes of mCPs indicate an important function in the maturation process of medusavirus.IMPORTANCEThe structural changes of giant virus capsids during maturation have not thus far been well clarified. Medusavirus is a unique giant virus in which infected amoebae release immature particles in addition to mature virus particles. In this study, we used cryo-electron microscopy to investigate immature and mature medusavirus particles and elucidate the structural changes of the viral capsid during the maturation process. In DNA-empty particles, the conformation of the minor capsid proteins changed dynamically depending on the presence or absence of the underlying internal membranes. In DNA-full particles, the lower part of the penton proteins was lost. This is the first report of structural changes of the viral capsid during the maturation process of giant viruses.</p>","PeriodicalId":17583,"journal":{"name":"Journal of Virology","volume":null,"pages":null},"PeriodicalIF":4.0,"publicationDate":"2024-08-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142080760","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
A reporter Oropouche virus expressing ZsGreen from the M segment enables pathogenesis studies in mice. 一种表达来自 M 段的 ZsGreen 的报告型 Oropouche 病毒可用于小鼠的致病机理研究。
IF 4 2区 医学 Q2 VIROLOGY Pub Date : 2024-08-28 DOI: 10.1128/jvi.00893-24
Krista Gunter, Dorcus Omoga, James M Bowen, Lorimar Robledo Gonzalez, Sydney Severt, Mackenzie Davis, Megan Szymanski, George Sandusky, W Paul Duprex, Natasha L Tilston-Lunel

Oropouche fever caused by Oropouche virus (OROV) is a significant zoonosis in Central and South America. Despite its public health significance, we lack high-throughput diagnostics, therapeutics, and a comprehensive knowledge of OROV biology. Reporter viruses are valuable tools to rapidly study virus dynamics and develop neutralization and antiviral screening assays. OROV is a tri-segmented bunyavirus, which makes generating a reporter virus challenging, as introducing foreign elements into the viral genome typically affects fitness. We previously demonstrated that the non-structural gene NSm on the OROV medium (M) segment is non-essential for replication in vitro. Taking advantage of this, we have now generated a recombinant OROV expressing fluorescent protein ZsGreen in place of NSm. This reporter OROV is both stable and pathogenic in IFNAR-/- mice and provides a powerful tool for OROV pathogenesis studies and assay development.IMPORTANCEEmerging and reemerging infectious agents such as zoonotic bunyaviruses are of global health concern. Oropouche virus (OROV) causes recurring outbreaks of acute febrile illness in the Central and South American human populations. Biting midges are the primary transmission vectors, whereas sloths and non-human primates are their reservoir hosts. As global temperatures increase, we will likely see an expansion in arthropod-borne pathogens such as OROV. Therefore, developing reagents to study pathogen biology to aid in identifying druggable targets is essential. Here, we demonstrate the feasibility and use of a fluorescent OROV reporter in mice to study viral dynamics and pathogenesis. We show that this reporter OROV maintains characteristics such as growth and pathogenicity similar to the wild-type virus. Using this reporter virus, we can now develop methods to assist OROV studies and establish various high-throughput assays.

由奥罗普切病毒(OROV)引起的奥罗普切热是中美洲和南美洲的一种重要人畜共患病。尽管它对公共卫生意义重大,但我们缺乏高通量诊断和治疗方法,也缺乏对奥罗波切病毒生物学的全面了解。报告病毒是快速研究病毒动态、开发中和与抗病毒筛选试验的宝贵工具。OROV 是一种三片段布尼亚病毒,这使得生成报告病毒具有挑战性,因为将外来元素引入病毒基因组通常会影响病毒的适应性。我们以前曾证明,OROV 中段(M)上的非结构基因 NSm 对于体外复制是非必需的。利用这一优势,我们现在生成了一种表达荧光蛋白 ZsGreen 的重组 OROV,以替代 NSm。这种报告型 OROV 在 IFNAR-/- 小鼠中既稳定又具有致病性,为 OROV 致病机理研究和检测方法开发提供了强有力的工具。 重要意义新出现和再次出现的传染性病原体(如人畜共患的布尼亚病毒)是全球健康关注的问题。奥罗普切病毒(OROV)会在中美洲和南美洲人群中反复爆发急性发热性疾病。叮咬蠓是主要的传播媒介,而树懒和非人类灵长类动物则是它们的蓄积宿主。随着全球气温的升高,节肢动物传播的病原体(如 OROV)很可能会增加。因此,开发研究病原体生物学的试剂以帮助确定药物靶点至关重要。在这里,我们展示了在小鼠体内使用荧光 OROV 报告物研究病毒动态和致病机理的可行性。我们发现,这种报告型 OROV 保持了与野生型病毒相似的生长和致病性等特征。利用这种报告病毒,我们现在可以开发辅助 OROV 研究的方法,并建立各种高通量检测方法。
{"title":"A reporter Oropouche virus expressing ZsGreen from the M segment enables pathogenesis studies in mice.","authors":"Krista Gunter, Dorcus Omoga, James M Bowen, Lorimar Robledo Gonzalez, Sydney Severt, Mackenzie Davis, Megan Szymanski, George Sandusky, W Paul Duprex, Natasha L Tilston-Lunel","doi":"10.1128/jvi.00893-24","DOIUrl":"https://doi.org/10.1128/jvi.00893-24","url":null,"abstract":"<p><p>Oropouche fever caused by Oropouche virus (OROV) is a significant zoonosis in Central and South America. Despite its public health significance, we lack high-throughput diagnostics, therapeutics, and a comprehensive knowledge of OROV biology. Reporter viruses are valuable tools to rapidly study virus dynamics and develop neutralization and antiviral screening assays. OROV is a tri-segmented bunyavirus, which makes generating a reporter virus challenging, as introducing foreign elements into the viral genome typically affects fitness. We previously demonstrated that the non-structural gene NSm on the OROV medium (M) segment is non-essential for replication <i>in vitro</i>. Taking advantage of this, we have now generated a recombinant OROV expressing fluorescent protein ZsGreen in place of NSm. This reporter OROV is both stable and pathogenic in IFNAR<sup>-/-</sup> mice and provides a powerful tool for OROV pathogenesis studies and assay development.IMPORTANCEEmerging and reemerging infectious agents such as zoonotic bunyaviruses are of global health concern. Oropouche virus (OROV) causes recurring outbreaks of acute febrile illness in the Central and South American human populations. Biting midges are the primary transmission vectors, whereas sloths and non-human primates are their reservoir hosts. As global temperatures increase, we will likely see an expansion in arthropod-borne pathogens such as OROV. Therefore, developing reagents to study pathogen biology to aid in identifying druggable targets is essential. Here, we demonstrate the feasibility and use of a fluorescent OROV reporter in mice to study viral dynamics and pathogenesis. We show that this reporter OROV maintains characteristics such as growth and pathogenicity similar to the wild-type virus. Using this reporter virus, we can now develop methods to assist OROV studies and establish various high-throughput assays.</p>","PeriodicalId":17583,"journal":{"name":"Journal of Virology","volume":null,"pages":null},"PeriodicalIF":4.0,"publicationDate":"2024-08-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142080661","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
miR-451-targeted PSMB8 promotes PRRSV infection by degrading IRF3. miR-451 靶向 PSMB8 通过降解 IRF3 促进 PRRSV 感染。
IF 4 2区 医学 Q2 VIROLOGY Pub Date : 2024-08-28 DOI: 10.1128/jvi.00784-24
Sihan Li, Shuyuan Guo, Fang Liu, Yao Yao, Yingqi Zhu, Wen-Hai Feng

Porcine respiratory and reproductive syndrome (PRRS) is one of the most devastating infectious diseases of pigs, causing reproductive failures in sows and severe respiratory symptoms in piglets and growing pigs. MicroRNAs (miRNAs) are reported to play an essential role in virus-host interactions. In this study, we demonstrated that miR-451 enhanced type I interferon (IFN-I) production through targeting proteasome subunit β8 (PSMB8), therefore restricting PRRS virus (PRRSV) replication. We showed that the expression of PSMB8 was upregulated by PRRSV infection, and knockdown of PSMB8 inhibited PRRSV replication by promoting IFN-I production. Moreover, we demonstrated that PSMB8 interacted with the regulatory domain of IRF3 to mediate K48-linked polyubiquitination and degradation of IRF3. Also, importantly, we showed that PSMB8, as a target gene of miR-451, negatively regulated IFN-I production by promoting IRF3 degradation, which is a previously unknown mechanism for PSMB8 to modulate innate immune responses.

Importance: Porcine respiratory and reproductive syndrome virus (PRRSV), as a huge threat to the swine industry, is a causative agent that urgently needs to be solved. The dissecting of PRRSV pathogenesis and understanding of the host-pathogen interaction will provide insights into developing effective anti-PRRSV strategies. In this study, we showed that miR-451 dramatically inhibited PRRSV replication by targeting proteasome subunit β8 (PSMB8), a subunit of the immunoproteasome. Mutation of PSMB8 is often related to autoinflammatory diseases due to the elevated IFN production. We revealed that PSMB8 downregulated IFN production by promoting IRF3 degradation. In addition, we showed that PRRSV infection upregulated PSMB8 expression. Taken together, our findings reveal that miR-451 is a negative regulator of PRRSV replication, and PSMB8, a target gene of miR-451, negatively regulates IFN-I production by promoting IRF3 degradation, which is a previously unknown mechanism for PSMB8 to regulate innate immune responses.

猪呼吸与繁殖综合征(PRRS)是猪最具破坏性的传染病之一,会导致母猪繁殖失败,仔猪和生长猪出现严重的呼吸道症状。据报道,微小核糖核酸(miRNA)在病毒与宿主的相互作用中起着至关重要的作用。在这项研究中,我们证明了 miR-451 通过靶向蛋白酶体亚基 β8 (PSMB8),增强了 I 型干扰素 (IFN-I) 的产生,从而限制了 PRRS 病毒 (PRRSV) 的复制。我们发现,PSMB8的表达在PRRSV感染时上调,而敲除PSMB8可通过促进IFN-I的产生来抑制PRRSV的复制。此外,我们还发现 PSMB8 与 IRF3 的调节域相互作用,介导 K48 链接的多泛素化和 IRF3 的降解。此外,重要的是,我们发现 PSMB8 作为 miR-451 的靶基因,通过促进 IRF3 降解来负向调节 IFN-I 的产生,这是 PSMB8 调节先天性免疫反应的一种未知机制:猪呼吸与繁殖综合征病毒(PRRSV)对养猪业构成巨大威胁,是亟待解决的致病因子。剖析 PRRSV 的致病机理和了解宿主与病原体之间的相互作用将为制定有效的抗 PRRSV 策略提供启示。在这项研究中,我们发现 miR-451 通过靶向免疫蛋白酶体的一个亚基蛋白酶体亚基 β8 (PSMB8),显著抑制了 PRRSV 的复制。PSMB8的突变通常与自身炎症性疾病有关,原因是IFN产生增加。我们发现,PSMB8 通过促进 IRF3 降解来下调 IFN 的产生。此外,我们还发现 PRRSV 感染会上调 PSMB8 的表达。综上所述,我们的研究结果表明,miR-451是PRRSV复制的负调控因子,而PSMB8作为miR-451的靶基因,通过促进IRF3降解来负向调控IFN-I的产生,这是PSMB8调控先天性免疫反应的一种未知机制。
{"title":"miR-451-targeted PSMB8 promotes PRRSV infection by degrading IRF3.","authors":"Sihan Li, Shuyuan Guo, Fang Liu, Yao Yao, Yingqi Zhu, Wen-Hai Feng","doi":"10.1128/jvi.00784-24","DOIUrl":"https://doi.org/10.1128/jvi.00784-24","url":null,"abstract":"<p><p>Porcine respiratory and reproductive syndrome (PRRS) is one of the most devastating infectious diseases of pigs, causing reproductive failures in sows and severe respiratory symptoms in piglets and growing pigs. MicroRNAs (miRNAs) are reported to play an essential role in virus-host interactions. In this study, we demonstrated that miR-451 enhanced type I interferon (IFN-I) production through targeting proteasome subunit β8 (PSMB8), therefore restricting PRRS virus (PRRSV) replication. We showed that the expression of PSMB8 was upregulated by PRRSV infection, and knockdown of PSMB8 inhibited PRRSV replication by promoting IFN-I production. Moreover, we demonstrated that PSMB8 interacted with the regulatory domain of IRF3 to mediate K48-linked polyubiquitination and degradation of IRF3. Also, importantly, we showed that PSMB8, as a target gene of miR-451, negatively regulated IFN-I production by promoting IRF3 degradation, which is a previously unknown mechanism for PSMB8 to modulate innate immune responses.</p><p><strong>Importance: </strong>Porcine respiratory and reproductive syndrome virus (PRRSV), as a huge threat to the swine industry, is a causative agent that urgently needs to be solved. The dissecting of PRRSV pathogenesis and understanding of the host-pathogen interaction will provide insights into developing effective anti-PRRSV strategies. In this study, we showed that miR-451 dramatically inhibited PRRSV replication by targeting proteasome subunit β8 (PSMB8), a subunit of the immunoproteasome. Mutation of PSMB8 is often related to autoinflammatory diseases due to the elevated IFN production. We revealed that PSMB8 downregulated IFN production by promoting IRF3 degradation. In addition, we showed that PRRSV infection upregulated PSMB8 expression. Taken together, our findings reveal that miR-451 is a negative regulator of PRRSV replication, and PSMB8, a target gene of miR-451, negatively regulates IFN-I production by promoting IRF3 degradation, which is a previously unknown mechanism for PSMB8 to regulate innate immune responses.</p>","PeriodicalId":17583,"journal":{"name":"Journal of Virology","volume":null,"pages":null},"PeriodicalIF":4.0,"publicationDate":"2024-08-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142080755","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
The 5'UTR of porcine reproductive and respiratory syndrome virus strain JXwn06 harbors a uORF that regulates cellular inflammation. 猪繁殖与呼吸综合征病毒 JXwn06 株的 5'UTR 含有一个调节细胞炎症的 uORF。
IF 4 2区 医学 Q2 VIROLOGY Pub Date : 2024-08-28 DOI: 10.1128/jvi.01132-24
Teng Liu, Qi Shao, Zhiying Cui, Pengkai Qing, Peng Gao, Yongning Zhang, Lei Zhou, Xinna Ge, Xin Guo, Jun Han, Hanchun Yang

The 5' untranslated region (5'UTR) of many positive-stranded RNA viruses contain functional regulatory sequences. Here, we show that the porcine reproductive and respiratory syndrome virus (PRRSV), a member of arteriviruses, harbors small upstream open reading frames (uORFs) in its 5'UTR. Bioinformatics analysis shows that this feature is relatively well conserved among PRRSV strains and Arteriviridae. We also identified a uORF, namely uORF2, in the PRRSV strain JXwn06, that possesses translational activity and exerts a suppressive effect on the expression of the primary ORF evidenced by in vitro reporter assays. We tested its importance via reverse genetics by introducing a point mutation into the PRRSV infectious cDNA clone to inactivate the start codon of uORF2. The recovered mutant virus Mut2 surprisingly replicated to the same level as the wild-type virus (WT), but induced a higher level of inflammatory cytokines (e.g., TNF-α, IL-1β, and IL-6) both in vitro and in animal experiments, correlating well with more severe lung injury and higher death rate. In line with this, over-expression of uORF2 in transfected cells significantly inhibited poly(I:C)-induced expression of inflammatory cytokines. Together, our data support the idea that uORF2 encodes a novel, functional regulator of PRRSV virulence despite of its short size.

Importance: PRRSV has remained a major challenge to the world swine industry, but we still do not know much about its biology and pathogenesis. Here, we provide evidence to show that the 5'UTR of PRRSV strain JXwn06 harbors a functional uORF that has the coding capacity and regulates induction of inflammation as demonstrated by in vitro assays and animal experiment. The findings reveal a novel viral factor that regulates cellular inflammation and provide insight into the understanding of PRRSV pathogenesis.

许多正链 RNA 病毒的 5' 非翻译区 (5'UTR) 都含有功能性调控序列。在这里,我们发现猪繁殖与呼吸综合征病毒(PRRSV)作为动脉病毒的一员,在其 5'UTR 中含有小的上游开放阅读框(uORF)。生物信息学分析表明,这一特征在 PRRSV 株系和动脉病毒科之间相对保守。我们还在 PRRSV 株系 JXwn06 中发现了一个 uORF,即 uORF2,它具有翻译活性,体外报告实验证明它对主 ORF 的表达有抑制作用。我们在 PRRSV 感染性 cDNA 克隆中引入一个点突变,使 uORF2 的起始密码子失活,从而通过反向遗传学测试了它的重要性。恢复的突变病毒 Mut2 的复制水平竟然与野生型病毒(WT)相同,但在体外和动物实验中诱导的炎性细胞因子(如 TNF-α、IL-1β 和 IL-6)水平更高,与更严重的肺损伤和更高的死亡率密切相关。与此相一致,在转染细胞中过度表达 uORF2 能显著抑制 poly(I:C) 诱导的炎症细胞因子的表达。总之,我们的数据支持了一种观点,即尽管uORF2很短,但它编码了一种新型的 PRRSV 毒力功能调节因子:PRRSV一直是世界养猪业面临的重大挑战,但我们对其生物学和致病机理仍然知之甚少。本文提供的证据表明,PRRSV JXwn06 株的 5'UTR 含有一个功能性 uORF,该 uORF 具有编码能力,并通过体外实验和动物实验调节炎症诱导。这些发现揭示了一种新型的调控细胞炎症的病毒因子,并为人们了解 PRRSV 的致病机理提供了启示。
{"title":"The 5'UTR of porcine reproductive and respiratory syndrome virus strain JXwn06 harbors a uORF that regulates cellular inflammation.","authors":"Teng Liu, Qi Shao, Zhiying Cui, Pengkai Qing, Peng Gao, Yongning Zhang, Lei Zhou, Xinna Ge, Xin Guo, Jun Han, Hanchun Yang","doi":"10.1128/jvi.01132-24","DOIUrl":"https://doi.org/10.1128/jvi.01132-24","url":null,"abstract":"<p><p>The 5' untranslated region (5'UTR) of many positive-stranded RNA viruses contain functional regulatory sequences. Here, we show that the porcine reproductive and respiratory syndrome virus (PRRSV), a member of arteriviruses, harbors small upstream open reading frames (uORFs) in its 5'UTR. Bioinformatics analysis shows that this feature is relatively well conserved among PRRSV strains and <i>Arteriviridae</i>. We also identified a uORF, namely uORF2, in the PRRSV strain JXwn06, that possesses translational activity and exerts a suppressive effect on the expression of the primary ORF evidenced by <i>in vitro</i> reporter assays. We tested its importance via reverse genetics by introducing a point mutation into the PRRSV infectious cDNA clone to inactivate the start codon of uORF2. The recovered mutant virus Mut2 surprisingly replicated to the same level as the wild-type virus (WT), but induced a higher level of inflammatory cytokines (e.g., TNF-<i>α</i>, IL-1<i>β</i>, and IL-6) both <i>in vitro</i> and in animal experiments, correlating well with more severe lung injury and higher death rate. In line with this, over-expression of uORF2 in transfected cells significantly inhibited poly(I:C)-induced expression of inflammatory cytokines. Together, our data support the idea that uORF2 encodes a novel, functional regulator of PRRSV virulence despite of its short size.</p><p><strong>Importance: </strong>PRRSV has remained a major challenge to the world swine industry, but we still do not know much about its biology and pathogenesis. Here, we provide evidence to show that the 5'UTR of PRRSV strain JXwn06 harbors a functional uORF that has the coding capacity and regulates induction of inflammation as demonstrated by <i>in vitro</i> assays and animal experiment. The findings reveal a novel viral factor that regulates cellular inflammation and provide insight into the understanding of PRRSV pathogenesis.</p>","PeriodicalId":17583,"journal":{"name":"Journal of Virology","volume":null,"pages":null},"PeriodicalIF":4.0,"publicationDate":"2024-08-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142080761","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
LKB1 suppresses KSHV reactivation and promotes primary effusion lymphoma progression. LKB1 可抑制 KSHV 再激活并促进原发性渗出淋巴瘤的发展。
IF 4 2区 医学 Q2 VIROLOGY Pub Date : 2024-08-28 DOI: 10.1128/jvi.00604-24
Guanya Li, Yinan Li, Xinyu Tang, Lijie Wang, Shusheng Yue, Shanping He, Tingting Li

Viruses normally reprogram the host cell metabolic pathways as well as metabolic sensors to facilitate their persistence. The serine-threonine liver kinase B1 (LKB1) is a master upstream kinase of 5'-AMP-activated protein kinase (AMPK) that senses the energy status and therefore regulates the intracellular metabolic homeostasis. Previous studies showed that AMPK restricts Kaposi's sarcoma-associated herpesvirus (KSHV) lytic replication in endothelial cells during primary infection and promotes primary effusion lymphoma (PEL) cell survival. However, the role of LKB1 in KSHV lytic reactivation and KSHV-associated malignancies is unclear. In this study, we found that LKB1 is phosphorylated or activated in KSHV-positive PEL cells. Mechanistically, KSHV-encoded vCyclin mediated LKB1 activation in PEL cells, as vCyclin knockout ablated, while vCyclin overexpression enhanced LKB1 activation. Furthermore, knockdown of LKB1 inactivated AMPK and induced KSHV reactivation, as indicated by the increased expression of viral lytic genes and the increased virions in supernatants. Accordingly, AMPK inhibition by functional knockdown or a pharmacologic inhibitor, Compound C, promoted KSHV reactivation in PEL cells. Furthermore, inhibition of either LKB1 or AMPKα1 efficiently induced cell death by apoptosis of PEL cells both in vitro and in vivo. Together, these results identify LKB1 as a vulnerable target for PEL, which could be potentially exploited for treating other virus-associated diseases.IMPORTANCEKaposi's sarcoma-associated herpesvirus (KSHV) is an oncogenic virus associated with several human cancers, such as primary effusion lymphoma (PEL). Here, we showed that serine-threonine liver kinase B1 (LKB1), upstream of 5' AMP-activated protein kinase (AMPK), is activated by KSHV-encoded vCyclin and maintains KSHV latency in PEL cells. Inhibition of either LKB1 or AMPK enhances KSHV lytic replication from latency, which at least partially accounts for PEL cell death by apoptosis. Compound C, a potent AMPK inhibitor, induced KSHV reactivation and efficiently inhibited PEL progression in vivo. Thus, our work revealed that LKB1 is a potential therapeutic target for KSHV-associated cancers.

病毒通常会对宿主细胞的新陈代谢途径以及新陈代谢传感器进行重编程,以促进其持续存在。丝氨酸-苏氨酸肝激酶 B1(LKB1)是 5'-AMP 激活蛋白激酶(AMPK)的上游主激酶,可感知能量状态,从而调节细胞内的代谢平衡。先前的研究表明,AMPK 可在原发性感染期间限制卡波西肉瘤相关疱疹病毒(KSHV)在内皮细胞中的淋巴复制,并促进原发性渗出淋巴瘤(PEL)细胞的存活。然而,LKB1 在 KSHV 溶解性再活化和 KSHV 相关恶性肿瘤中的作用尚不清楚。在这项研究中,我们发现 LKB1 在 KSHV 阳性的 PEL 细胞中被磷酸化或激活。从机理上讲,KSHV编码的vCyclin介导了PEL细胞中LKB1的活化,因为vCyclin敲除会消融,而vCyclin过表达会增强LKB1的活化。此外,敲除 LKB1 会使 AMPK 失活并诱导 KSHV 再激活,病毒裂解基因的表达增加和上清液中病毒数量的增加都表明了这一点。因此,通过功能性敲除或药物抑制剂化合物 C 抑制 AMPK 可促进 PEL 细胞中的 KSHV 再激活。此外,在体外和体内抑制 LKB1 或 AMPKα1 都能有效诱导 PEL 细胞凋亡。重要意义卡波西肉瘤相关疱疹病毒(KSHV)是一种致癌病毒,与原发性渗出性淋巴瘤(PEL)等多种人类癌症有关。在这里,我们发现丝氨酸-苏氨酸肝激酶 B1(LKB1)是 5' AMP 激活蛋白激酶(AMPK)的上游,它被 KSHV 编码的 vCyclin 激活,并维持 KSHV 在 PEL 细胞中的潜伏期。抑制 LKB1 或 AMPK 会增强 KSHV 从潜伏期开始的溶解复制,这至少是 PEL 细胞凋亡的部分原因。化合物 C 是一种强效的 AMPK 抑制剂,它能诱导 KSHV 重新激活并有效抑制 PEL 在体内的发展。因此,我们的研究揭示了 LKB1 是 KSHV 相关癌症的潜在治疗靶点。
{"title":"LKB1 suppresses KSHV reactivation and promotes primary effusion lymphoma progression.","authors":"Guanya Li, Yinan Li, Xinyu Tang, Lijie Wang, Shusheng Yue, Shanping He, Tingting Li","doi":"10.1128/jvi.00604-24","DOIUrl":"https://doi.org/10.1128/jvi.00604-24","url":null,"abstract":"<p><p>Viruses normally reprogram the host cell metabolic pathways as well as metabolic sensors to facilitate their persistence. The serine-threonine liver kinase B1 (LKB1) is a master upstream kinase of 5'-AMP-activated protein kinase (AMPK) that senses the energy status and therefore regulates the intracellular metabolic homeostasis. Previous studies showed that AMPK restricts Kaposi's sarcoma-associated herpesvirus (KSHV) lytic replication in endothelial cells during primary infection and promotes primary effusion lymphoma (PEL) cell survival. However, the role of LKB1 in KSHV lytic reactivation and KSHV-associated malignancies is unclear. In this study, we found that LKB1 is phosphorylated or activated in KSHV-positive PEL cells. Mechanistically, KSHV-encoded vCyclin mediated LKB1 activation in PEL cells, as vCyclin knockout ablated, while vCyclin overexpression enhanced LKB1 activation. Furthermore, knockdown of LKB1 inactivated AMPK and induced KSHV reactivation, as indicated by the increased expression of viral lytic genes and the increased virions in supernatants. Accordingly, AMPK inhibition by functional knockdown or a pharmacologic inhibitor, Compound C, promoted KSHV reactivation in PEL cells. Furthermore, inhibition of either LKB1 or AMPKα1 efficiently induced cell death by apoptosis of PEL cells both <i>in vitro</i> and <i>in vivo</i>. Together, these results identify LKB1 as a vulnerable target for PEL, which could be potentially exploited for treating other virus-associated diseases.IMPORTANCEKaposi's sarcoma-associated herpesvirus (KSHV) is an oncogenic virus associated with several human cancers, such as primary effusion lymphoma (PEL). Here, we showed that serine-threonine liver kinase B1 (LKB1), upstream of 5<i>'</i> AMP-activated protein kinase (AMPK), is activated by KSHV-encoded vCyclin and maintains KSHV latency in PEL cells. Inhibition of either LKB1 or AMPK enhances KSHV lytic replication from latency, which at least partially accounts for PEL cell death by apoptosis. Compound C, a potent AMPK inhibitor, induced KSHV reactivation and efficiently inhibited PEL progression <i>in vivo</i>. Thus, our work revealed that LKB1 is a potential therapeutic target for KSHV-associated cancers.</p>","PeriodicalId":17583,"journal":{"name":"Journal of Virology","volume":null,"pages":null},"PeriodicalIF":4.0,"publicationDate":"2024-08-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142080754","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Characterization of viroplasm-like structures by co-expression of NSP5 and NSP2 across rotavirus species A to J. 通过共同表达轮状病毒 A 至 J 型的 NSP5 和 NSP2,确定类病毒质结构的特征。
IF 4 2区 医学 Q2 VIROLOGY Pub Date : 2024-08-28 DOI: 10.1128/jvi.00975-24
Melissa Lee, Ariana Cosic, Kurt Tobler, Claudio Aguilar, Cornel Fraefel, Catherine Eichwald

Rotaviruses (RVs) are classified into nine species, A-D and F-J, with species A being the most studied. In rotavirus of species A (RVA), replication occurs in viroplasms, which are cytosolic globular inclusions composed of main building block proteins NSP5, NSP2, and VP2. The co-expression of NSP5 with either NSP2 or VP2 in uninfected cells leads to the formation of viroplasm-like structures (VLSs). Although morphologically identical to viroplasms, VLSs do not produce viral progeny but serve as excellent tools for studying complex viroplasms. A knowledge gap exists regarding non-RVA viroplasms due to the lack of specific antibodies and suitable cell culture systems. In this study, we explored the ability of NSP5 and NSP2 from non-RVA species to form VLSs. The co-expression of these two proteins led to globular VLSs in RV species A, B, D, F, G, and I, while RVC formed filamentous VLSs. The co-expression of NSP5 and NSP2 of RV species H and J did not result in VLS formation. Interestingly, NSP5 of all RV species self-oligomerizes, with the ordered C-terminal region, termed the tail, being necessary for self-oligomerization of RV species A-C and G-J. Except for NSP5 from RVJ, all NSP5 interacted with their cognate NSP2. We also found that interspecies VLS are formed between closely related RV species B with G and D with F. Additionally, VLS from RVH and RVJ formed when the tail of NSP5 RVH and RVJ was replaced by the tail of NSP5 from RVA and co-expressed with their respective NSP2.

Importance: Rotaviruses (RVs) are classified into nine species, A-D and F-J, infecting mammals and birds. Due to the lack of research tools, all cumulative knowledge on RV replication is based on RV species A (RVA). The RV replication compartments are globular cytosolic structures named viroplasms, which have only been identified in RV species A. In this study, we examined the formation of viroplasm-like structures (VLSs) by the co-expression of NSP5 with NSP2 across RV species A to J. Globular VLSs formed for RV species A, B, D, F, G, and I, while RV species C formed filamentous structures. The RV species H and J did not form VLS with their cognates NSP5 and NSP2. Similar to RVA, NSP5 self-oligomerizes in all RV species, which is required for VLS formation. This study provides basic knowledge of the non-RVA replication mechanisms, which could help develop strategies to halt virus infection across RV species.

轮状病毒(RV)分为 A-D 和 F-J 九个种,其中研究最多的是 A 种。在 A 种轮状病毒(RVA)中,复制发生在病毒浆液中,病毒浆液是由主要结构单元蛋白 NSP5、NSP2 和 VP2 组成的细胞膜球状包涵体。在未感染的细胞中,NSP5 与 NSP2 或 VP2 共同表达会形成类病毒浆状结构(VLS)。虽然在形态上与病毒浆膜相同,但 VLSs 不会产生病毒后代,而是研究复杂病毒浆膜的绝佳工具。由于缺乏特异性抗体和合适的细胞培养系统,关于非 RVA 病毒原体的知识存在空白。在本研究中,我们探讨了非 RVA 物种的 NSP5 和 NSP2 形成 VLS 的能力。在 RV 物种 A、B、D、F、G 和 I 中,共表达这两种蛋白可形成球状 VLS,而 RVC 则形成丝状 VLS。在 RV H 和 J 中,NSP5 和 NSP2 的共表达不会导致 VLS 的形成。有趣的是,所有 RV 物种的 NSP5 都能自聚,有序的 C 端区域(称为尾部)是 RV 物种 A-C 和 G-J 自聚的必要条件。除来自 RVJ 的 NSP5 外,所有 NSP5 都与其同源的 NSP2 相互作用。此外,当 RVH 和 RVJ 的 NSP5 尾部被 RVA 的 NSP5 尾部取代并与各自的 NSP2 共同表达时,RVH 和 RVJ 的 VLS 也会形成:轮状病毒(RV)分为 A-D 和 F-J 九种,感染哺乳动物和鸟类。由于缺乏研究工具,所有关于轮状病毒复制的累积知识都是基于轮状病毒 A 种(RVA)。RV 复制区是球状胞浆结构,被命名为 "病毒浆"(viroplasms),目前只在 RV A 种中发现了这种结构。在本研究中,我们考察了 NSP5 与 NSP2 共同表达在 RV A 至 J 种中病毒浆样结构(VLS)的形成情况。RV 物种 H 和 J 没有与其同源物 NSP5 和 NSP2 形成 VLS。与 RVA 相似,NSP5 在所有 RV 物种中都会自聚,而这是形成 VLS 所必需的。这项研究提供了有关非 RVA 复制机制的基本知识,有助于开发出阻止病毒跨 RV 物种感染的策略。
{"title":"Characterization of viroplasm-like structures by co-expression of NSP5 and NSP2 across rotavirus species A to J.","authors":"Melissa Lee, Ariana Cosic, Kurt Tobler, Claudio Aguilar, Cornel Fraefel, Catherine Eichwald","doi":"10.1128/jvi.00975-24","DOIUrl":"https://doi.org/10.1128/jvi.00975-24","url":null,"abstract":"<p><p>Rotaviruses (RVs) are classified into nine species, A-D and F-J, with species A being the most studied. In rotavirus of species A (RVA), replication occurs in viroplasms, which are cytosolic globular inclusions composed of main building block proteins NSP5, NSP2, and VP2. The co-expression of NSP5 with either NSP2 or VP2 in uninfected cells leads to the formation of viroplasm-like structures (VLSs). Although morphologically identical to viroplasms, VLSs do not produce viral progeny but serve as excellent tools for studying complex viroplasms. A knowledge gap exists regarding non-RVA viroplasms due to the lack of specific antibodies and suitable cell culture systems. In this study, we explored the ability of NSP5 and NSP2 from non-RVA species to form VLSs. The co-expression of these two proteins led to globular VLSs in RV species A, B, D, F, G, and I, while RVC formed filamentous VLSs. The co-expression of NSP5 and NSP2 of RV species H and J did not result in VLS formation. Interestingly, NSP5 of all RV species self-oligomerizes, with the ordered C-terminal region, termed the tail, being necessary for self-oligomerization of RV species A-C and G-J. Except for NSP5 from RVJ, all NSP5 interacted with their cognate NSP2. We also found that interspecies VLS are formed between closely related RV species B with G and D with F. Additionally, VLS from RVH and RVJ formed when the tail of NSP5 RVH and RVJ was replaced by the tail of NSP5 from RVA and co-expressed with their respective NSP2.</p><p><strong>Importance: </strong>Rotaviruses (RVs) are classified into nine species, A-D and F-J, infecting mammals and birds. Due to the lack of research tools, all cumulative knowledge on RV replication is based on RV species A (RVA). The RV replication compartments are globular cytosolic structures named viroplasms, which have only been identified in RV species A. In this study, we examined the formation of viroplasm-like structures (VLSs) by the co-expression of NSP5 with NSP2 across RV species A to J. Globular VLSs formed for RV species A, B, D, F, G, and I, while RV species C formed filamentous structures. The RV species H and J did not form VLS with their cognates NSP5 and NSP2. Similar to RVA, NSP5 self-oligomerizes in all RV species, which is required for VLS formation. This study provides basic knowledge of the non-RVA replication mechanisms, which could help develop strategies to halt virus infection across RV species.</p>","PeriodicalId":17583,"journal":{"name":"Journal of Virology","volume":null,"pages":null},"PeriodicalIF":4.0,"publicationDate":"2024-08-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142080663","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Ipsilateral or contralateral boosting of mice with mRNA vaccines confers equivalent immunity and protection against a SARS-CoV-2 Omicron strain. 用 mRNA 疫苗对小鼠进行同侧或对侧增殖,可获得同等的免疫力和对 SARS-CoV-2 Omicron 株的保护。
IF 4 2区 医学 Q2 VIROLOGY Pub Date : 2024-08-28 DOI: 10.1128/jvi.00574-24
Baoling Ying, Chieh-Yu Liang, Pritesh Desai, Suzanne M Scheaffer, Sayda M Elbashir, Darin K Edwards, Larissa B Thackray, Michael S Diamond

Boosting with mRNA vaccines encoding variant-matched spike proteins has been implemented to mitigate their reduced efficacy against emerging SARS-CoV-2 variants. Nonetheless, in humans, it remains unclear whether boosting in the ipsilateral or contralateral arm with respect to the priming doses impacts immunity and protection. Here, we boosted K18-hACE2 mice with either monovalent mRNA-1273 (Wuhan-1 spike) or bivalent mRNA-1273.214 (Wuhan-1 + BA.1 spike) vaccine in the ipsilateral or contralateral leg after a two-dose priming series with mRNA-1273. Boosting in the ipsilateral or contralateral leg elicited equivalent levels of serum IgG and neutralizing antibody responses against Wuhan-1 and BA.1. While contralateral boosting with mRNA vaccines resulted in the expansion of spike-specific B and T cells beyond the ipsilateral draining lymph node (DLN) to the contralateral DLN, administration of a third mRNA vaccine dose at either site resulted in similar levels of antigen-specific germinal center B cells, plasmablasts/plasma cells, T follicular helper cells, and CD8+ T cells in the DLNs and the spleen. Furthermore, ipsilateral and contralateral boosting with mRNA-1273 or mRNA-1273.214 vaccines conferred similar homologous or heterologous immune protection against SARS-CoV-2 BA.1 virus challenge with equivalent reductions in viral RNA and infectious virus in the nasal turbinates and lungs. Collectively, our data show limited differences in B and T cell immune responses after ipsilateral and contralateral site boosting by mRNA vaccines that do not substantively impact protection against an Omicron strain.IMPORTANCESequential boosting with mRNA vaccines has been an effective strategy to overcome waning immunity and neutralization escape by emerging SARS-CoV-2 variants. However, it remains unclear how the site of boosting relative to the primary vaccination series shapes optimal immune responses or breadth of protection against variants. In K18-hACE2 transgenic mice, we observed that intramuscular boosting with historical monovalent or variant-matched bivalent vaccines in the ipsilateral or contralateral limb elicited comparable levels of serum spike-specific antibody and antigen-specific B and T cell responses. Moreover, boosting on either side conferred equivalent protection against a SARS-CoV-2 Omicron challenge strain. Our data in mice suggest that the site of intramuscular boosting with an mRNA vaccine does not substantially impact immunity or protection against SARS-CoV-2 infection.

为了减轻 mRNA 疫苗对新出现的 SARS-CoV-2 变异株的疗效降低,人们使用了编码变异株匹配尖峰蛋白的 mRNA 疫苗进行增强。然而,在人类中,同侧或对侧手臂上的启动剂量是否会影响免疫力和保护作用仍不清楚。在此,我们用单价mRNA-1273(武汉-1钉螺)或双价mRNA-1273.214(武汉-1 + BA.1钉螺)疫苗对K18-hACE2小鼠进行同侧或对侧腿部增强,然后再用mRNA-1273进行两剂系列引种。同侧或对侧腿部接种可引起同等水平的血清 IgG 和针对武汉-1 和 BA.1 的中和抗体反应。虽然用 mRNA 疫苗进行对侧增强会导致尖峰特异性 B 细胞和 T 细胞从同侧引流淋巴结 (DLN) 扩展到对侧 DLN,但在任一部位接种第三剂 mRNA 疫苗都会在 DLN 和脾脏中产生相似水平的抗原特异性生殖中心 B 细胞、浆细胞/浆细胞、T 滤泡辅助细胞和 CD8+ T 细胞。此外,同侧和对侧接种 mRNA-1273 或 mRNA-1273.214 疫苗可在 SARS-CoV-2 BA.1 病毒挑战下获得相似的同源或异源免疫保护,鼻甲和肺部的病毒 RNA 和感染性病毒减少量相当。总之,我们的数据表明,mRNA 疫苗在同侧和对侧部位增强后,B 细胞和 T 细胞免疫反应的差异有限,不会对抵御 Omicron 株的保护产生实质性影响。 重要意义mRNA 疫苗的等效增强一直是克服免疫力下降和新出现的 SARS-CoV-2 变异株中和逃逸的有效策略。然而,目前仍不清楚相对于主要疫苗接种系列的增强接种部位如何形成最佳免疫反应或对变异株的保护广度。在 K18-hACE2 转基因小鼠中,我们观察到在同侧或对侧肢体肌肉注射历史单价疫苗或与变异株匹配的二价疫苗可引起相当水平的血清尖峰特异性抗体以及抗原特异性 B 细胞和 T 细胞反应。此外,在两侧肢体上接种二价疫苗都能对 SARS-CoV-2 Omicron 挑战株产生同等的保护作用。我们在小鼠身上获得的数据表明,肌肉注射 mRNA 疫苗的部位不会对免疫力或对 SARS-CoV-2 感染的保护产生重大影响。
{"title":"Ipsilateral or contralateral boosting of mice with mRNA vaccines confers equivalent immunity and protection against a SARS-CoV-2 Omicron strain.","authors":"Baoling Ying, Chieh-Yu Liang, Pritesh Desai, Suzanne M Scheaffer, Sayda M Elbashir, Darin K Edwards, Larissa B Thackray, Michael S Diamond","doi":"10.1128/jvi.00574-24","DOIUrl":"https://doi.org/10.1128/jvi.00574-24","url":null,"abstract":"<p><p>Boosting with mRNA vaccines encoding variant-matched spike proteins has been implemented to mitigate their reduced efficacy against emerging SARS-CoV-2 variants. Nonetheless, in humans, it remains unclear whether boosting in the ipsilateral or contralateral arm with respect to the priming doses impacts immunity and protection. Here, we boosted K18-hACE2 mice with either monovalent mRNA-1273 (Wuhan-1 spike) or bivalent mRNA-1273.214 (Wuhan-1 + BA.1 spike) vaccine in the ipsilateral or contralateral leg after a two-dose priming series with mRNA-1273. Boosting in the ipsilateral or contralateral leg elicited equivalent levels of serum IgG and neutralizing antibody responses against Wuhan-1 and BA.1. While contralateral boosting with mRNA vaccines resulted in the expansion of spike-specific B and T cells beyond the ipsilateral draining lymph node (DLN) to the contralateral DLN, administration of a third mRNA vaccine dose at either site resulted in similar levels of antigen-specific germinal center B cells, plasmablasts/plasma cells, T follicular helper cells, and CD8<sup>+</sup> T cells in the DLNs and the spleen. Furthermore, ipsilateral and contralateral boosting with mRNA-1273 or mRNA-1273.214 vaccines conferred similar homologous or heterologous immune protection against SARS-CoV-2 BA.1 virus challenge with equivalent reductions in viral RNA and infectious virus in the nasal turbinates and lungs. Collectively, our data show limited differences in B and T cell immune responses after ipsilateral and contralateral site boosting by mRNA vaccines that do not substantively impact protection against an Omicron strain.IMPORTANCESequential boosting with mRNA vaccines has been an effective strategy to overcome waning immunity and neutralization escape by emerging SARS-CoV-2 variants. However, it remains unclear how the site of boosting relative to the primary vaccination series shapes optimal immune responses or breadth of protection against variants. In K18-hACE2 transgenic mice, we observed that intramuscular boosting with historical monovalent or variant-matched bivalent vaccines in the ipsilateral or contralateral limb elicited comparable levels of serum spike-specific antibody and antigen-specific B and T cell responses. Moreover, boosting on either side conferred equivalent protection against a SARS-CoV-2 Omicron challenge strain. Our data in mice suggest that the site of intramuscular boosting with an mRNA vaccine does not substantially impact immunity or protection against SARS-CoV-2 infection.</p>","PeriodicalId":17583,"journal":{"name":"Journal of Virology","volume":null,"pages":null},"PeriodicalIF":4.0,"publicationDate":"2024-08-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142080686","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Protective role of cytosolic prion protein against virus infection in prion-infected cells. 朊病毒感染细胞中细胞膜朊病毒蛋白对病毒感染的保护作用。
IF 4 2区 医学 Q2 VIROLOGY Pub Date : 2024-08-28 DOI: 10.1128/jvi.01262-24
Hideyuki Hara, Junji Chida, Batzaya Batchuluun, Etsuhisa Takahashi, Hiroshi Kido, Suehiro Sakaguchi

Production of the amyloidogenic prion protein, PrPSc, which forms infectious protein aggregates, or prions, is a key pathogenic event in prion diseases. Functional prion-like protein aggregations, such as the mitochondrial adaptor protein MAVS and the inflammasome component protein ASC, have been identified to play a protective role in viral infections in mammalian cells. In this study, to investigate if PrPSc could play a functional role against external stimuli, we infected prion-infected cells with a neurotropic influenza A virus strain, IAV/WSN. We found that prion-infected cells were highly resistant to IAV/WSN infection. In these cells, NF-κB nuclear translocation was disturbed; therefore, mitochondrial superoxide dismutase (mtSOD) expression was suppressed, and mitochondrial reactive oxygen species (mtROS) was increased. The elevated mtROS subsequently activated NLRP3 inflammasomes, leading to the suppression of IAV/WSN-induced necroptosis. We also found that prion-infected cells accumulated a portion of PrP molecules in the cytosol, and that the N-terminal potential nuclear translocation signal of PrP impeded NF-κB nuclear translocation. These results suggest that PrPSc might play a functional role in protection against viral infections by stimulating the NLRP3 inflammasome-dependent antivirus mechanism through the cytosolic PrP-mediated disturbance of NF-κB nuclear translocation, which leads to suppression of mtSOD expression and consequently upregulation of the NLRP3 inflammasome activator mtROS.

Importance: Cytosolic PrP has been detected in prion-infected cells and suggested to be involved in the neurotoxicity of prions. Here, we also detected cytosolic PrP in prion-infected cells. We further found that the nuclear translocation of NF-κB was disturbed in prion-infected cells and that the N-terminal potential nuclear translocation signal of PrP expressed in the cytosol disturbed the nuclear translocation of NF-κB. Thus, the N-terminal nuclear translocation signal of cytosolic PrP might play a role in prion neurotoxicity. Prion-like protein aggregates in other protein misfolding disorders, including Alzheimer's disease were reported to play a protective role against various environmental stimuli. We here showed that prion-infected cells were partially resistant to IAV/WSN infection due to the cytosolic PrP-mediated disturbance of the nuclear translocation of NF-κB, which consequently activated NLRP3 inflammasomes after IAV/WSN infection. It is thus possible that prions could also play a protective role in viral infections.

产生淀粉样蛋白朊病毒蛋白 PrPSc,形成感染性蛋白聚集体或朊病毒,是朊病毒疾病的关键致病事件。已发现线粒体适配蛋白 MAVS 和炎性体组成蛋白 ASC 等功能性朊病毒样蛋白聚集体在哺乳动物细胞的病毒感染中发挥保护作用。在本研究中,为了研究PrPSc是否能在抵御外部刺激时发挥功能性作用,我们用一种神经性甲型流感病毒株IAV/WSN感染了朊病毒感染细胞。我们发现,朊病毒感染细胞对 IAV/WSN 感染具有很强的抵抗力。在这些细胞中,NF-κB 的核转位受到干扰,因此线粒体超氧化物歧化酶(mtSOD)的表达受到抑制,线粒体活性氧(mtROS)增加。升高的 mtROS 随后激活了 NLRP3 炎症小体,从而抑制了 IAV/WSN 诱导的坏死。我们还发现,受朊病毒感染的细胞在细胞质中积累了一部分 PrP 分子,而 PrP 的 N 端潜在核转位信号阻碍了 NF-κB 的核转位。这些结果表明,PrPSc可能通过细胞膜PrP介导的NF-κB核转位干扰,刺激NLRP3炎性体依赖的抗病毒机制,从而抑制mtSOD的表达,进而上调NLRP3炎性体激活剂mtROS,在抵御病毒感染方面发挥功能性作用:在朊病毒感染的细胞中检测到了细胞膜 PrP,并认为它与朊病毒的神经毒性有关。在这里,我们也在朊病毒感染的细胞中检测到了细胞质 PrP。我们进一步发现,在朊病毒感染的细胞中,NF-κB 的核转位受到干扰,在细胞质中表达的 PrP 的 N 端潜在核转位信号干扰了 NF-κB 的核转位。因此,胞浆 PrP 的 N 端核转位信号可能在朊病毒神经毒性中发挥作用。据报道,在包括阿尔茨海默病在内的其他蛋白质错误折叠疾病中,朊病毒样蛋白聚集体对各种环境刺激起到保护作用。我们在这里发现,朊病毒感染细胞对 IAV/WSN 感染具有部分抵抗力,这是由于细胞膜 PrP 介导的 NF-κB 核转位紊乱,从而在 IAV/WSN 感染后激活了 NLRP3 炎症体。因此,朊病毒也可能在病毒感染中发挥保护作用。
{"title":"Protective role of cytosolic prion protein against virus infection in prion-infected cells.","authors":"Hideyuki Hara, Junji Chida, Batzaya Batchuluun, Etsuhisa Takahashi, Hiroshi Kido, Suehiro Sakaguchi","doi":"10.1128/jvi.01262-24","DOIUrl":"https://doi.org/10.1128/jvi.01262-24","url":null,"abstract":"<p><p>Production of the amyloidogenic prion protein, PrP<sup>Sc</sup>, which forms infectious protein aggregates, or prions, is a key pathogenic event in prion diseases. Functional prion-like protein aggregations, such as the mitochondrial adaptor protein MAVS and the inflammasome component protein ASC, have been identified to play a protective role in viral infections in mammalian cells. In this study, to investigate if PrP<sup>Sc</sup> could play a functional role against external stimuli, we infected prion-infected cells with a neurotropic influenza A virus strain, IAV/WSN. We found that prion-infected cells were highly resistant to IAV/WSN infection. In these cells, NF-κB nuclear translocation was disturbed; therefore, mitochondrial superoxide dismutase (mtSOD) expression was suppressed, and mitochondrial reactive oxygen species (mtROS) was increased. The elevated mtROS subsequently activated NLRP3 inflammasomes, leading to the suppression of IAV/WSN-induced necroptosis. We also found that prion-infected cells accumulated a portion of PrP molecules in the cytosol, and that the N-terminal potential nuclear translocation signal of PrP impeded NF-κB nuclear translocation. These results suggest that PrP<sup>Sc</sup> might play a functional role in protection against viral infections by stimulating the NLRP3 inflammasome-dependent antivirus mechanism through the cytosolic PrP-mediated disturbance of NF-κB nuclear translocation, which leads to suppression of mtSOD expression and consequently upregulation of the NLRP3 inflammasome activator mtROS.</p><p><strong>Importance: </strong>Cytosolic PrP has been detected in prion-infected cells and suggested to be involved in the neurotoxicity of prions. Here, we also detected cytosolic PrP in prion-infected cells. We further found that the nuclear translocation of NF-κB was disturbed in prion-infected cells and that the N-terminal potential nuclear translocation signal of PrP expressed in the cytosol disturbed the nuclear translocation of NF-κB. Thus, the N-terminal nuclear translocation signal of cytosolic PrP might play a role in prion neurotoxicity. Prion-like protein aggregates in other protein misfolding disorders, including Alzheimer's disease were reported to play a protective role against various environmental stimuli. We here showed that prion-infected cells were partially resistant to IAV/WSN infection due to the cytosolic PrP-mediated disturbance of the nuclear translocation of NF-κB, which consequently activated NLRP3 inflammasomes after IAV/WSN infection. It is thus possible that prions could also play a protective role in viral infections.</p>","PeriodicalId":17583,"journal":{"name":"Journal of Virology","volume":null,"pages":null},"PeriodicalIF":4.0,"publicationDate":"2024-08-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142080757","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Coxsackievirus group B3 regulates ASS1-mediated metabolic reprogramming and promotes macrophage inflammatory polarization in viral myocarditis. 柯萨奇病毒 B3 组调控 ASS1 介导的代谢重编程,并促进病毒性心肌炎中巨噬细胞炎症极化。
IF 4 2区 医学 Q2 VIROLOGY Pub Date : 2024-08-28 DOI: 10.1128/jvi.00805-24
Qiong Liu, Yinpan Shang, Ziwei Tao, Xuan Li, Lu Shen, Hanchi Zhang, Zhili Liu, Zhirong Rao, Xiaomin Yu, Yanli Cao, Lingbing Zeng, Xiaotian Huang

Coxsackievirus group B3 (CVB3) belongs to the genus Enteroviruses of the family Picornaviridae and is the main pathogen underlying viral myocarditis (VMC). No specific therapeutic is available for this condition. Argininosuccinate synthase 1 (ASS1) is a key enzyme in the urea cycle that converts citrulline and aspartic acid to argininosuccinate. Here, we found that CVB3 and its capsid protein VP2 inhibit the autophagic degradation of ASS1 and that CVB3 consumes citrulline to upregulate ASS1, triggers urea cycle metabolic reprogramming, and then activates macrophages to develop pro-inflammatory polarization, thereby promoting the occurrence and development of VMC. Conversely, citrulline supplementation to prevent depletion can downregulate ASS1, rescue macrophage polarization, and alleviate the pathogenicity of VMC. These findings provide a new perspective on the occurrence and development of VMC, revealing ASS1 as a potential new target for treating this disease.

Importance: Viral myocarditis (VMC) is a common and potentially life-threatening myocardial inflammatory disease, most commonly caused by CVB3 infection. So far, the pathogenesis of VMC caused by CVB3 is mainly focused on two aspects: one is the direct myocardial injury caused by a large number of viral replication in the early stage of infection, and the other is the local immune cell infiltration and inflammatory damage of the myocardium in the adaptive immune response stage. There are few studies on the early innate immunity of CVB3 infection in myocardial tissue, but the appearance of macrophages in the early stage of CVB3 infection suggests that they can play a regulatory role as early innate immune response cells in myocardial tissue. Here, we discovered a possible new mechanism of VMC caused by CVB3, revealed new drug targets for anti-CVB3, and discovered the therapeutic potential of citrulline for VMC.

柯萨奇病毒 B3 组(CVB3)属于微小病毒科肠道病毒属,是病毒性心肌炎(VMC)的主要病原体。目前还没有针对这种疾病的特效疗法。精氨酸琥珀酸合成酶 1(ASS1)是尿素循环中的一种关键酶,可将瓜氨酸和天冬氨酸转化为精氨酸琥珀酸。在这里,我们发现CVB3及其囊膜蛋白VP2能抑制ASS1的自噬降解,CVB3消耗瓜氨酸上调ASS1,引发尿素循环代谢重编程,进而激活巨噬细胞产生促炎极化,从而促进VMC的发生和发展。相反,补充瓜氨酸以防止耗竭,可以下调 ASS1,挽救巨噬细胞极化,减轻 VMC 的致病性。这些发现为 VMC 的发生和发展提供了新的视角,揭示了 ASS1 是治疗这种疾病的潜在新靶点:病毒性心肌炎(VMC)是一种常见且可能危及生命的心肌炎性疾病,最常见的病因是 CVB3 感染。迄今为止,由 CVB3 引起的病毒性心肌炎的发病机制主要集中在两个方面:一是感染早期大量病毒复制引起的直接心肌损伤,二是适应性免疫反应阶段局部免疫细胞浸润和心肌的炎性损伤。关于 CVB3 感染心肌组织早期先天性免疫的研究很少,但 CVB3 感染早期巨噬细胞的出现表明,巨噬细胞可作为心肌组织早期先天性免疫反应细胞发挥调节作用。在此,我们发现了 CVB3 导致 VMC 的可能新机制,揭示了抗 CVB3 的新药物靶点,并发现了瓜氨酸治疗 VMC 的潜力。
{"title":"Coxsackievirus group B3 regulates ASS1-mediated metabolic reprogramming and promotes macrophage inflammatory polarization in viral myocarditis.","authors":"Qiong Liu, Yinpan Shang, Ziwei Tao, Xuan Li, Lu Shen, Hanchi Zhang, Zhili Liu, Zhirong Rao, Xiaomin Yu, Yanli Cao, Lingbing Zeng, Xiaotian Huang","doi":"10.1128/jvi.00805-24","DOIUrl":"https://doi.org/10.1128/jvi.00805-24","url":null,"abstract":"<p><p>Coxsackievirus group B3 (CVB3) belongs to the genus <i>Enteroviruses</i> of the family <i>Picornaviridae</i> and is the main pathogen underlying viral myocarditis (VMC). No specific therapeutic is available for this condition. Argininosuccinate synthase 1 (ASS1) is a key enzyme in the urea cycle that converts citrulline and aspartic acid to argininosuccinate. Here, we found that CVB3 and its capsid protein VP2 inhibit the autophagic degradation of ASS1 and that CVB3 consumes citrulline to upregulate ASS1, triggers urea cycle metabolic reprogramming, and then activates macrophages to develop pro-inflammatory polarization, thereby promoting the occurrence and development of VMC. Conversely, citrulline supplementation to prevent depletion can downregulate ASS1, rescue macrophage polarization, and alleviate the pathogenicity of VMC. These findings provide a new perspective on the occurrence and development of VMC, revealing ASS1 as a potential new target for treating this disease.</p><p><strong>Importance: </strong>Viral myocarditis (VMC) is a common and potentially life-threatening myocardial inflammatory disease, most commonly caused by CVB3 infection. So far, the pathogenesis of VMC caused by CVB3 is mainly focused on two aspects: one is the direct myocardial injury caused by a large number of viral replication in the early stage of infection, and the other is the local immune cell infiltration and inflammatory damage of the myocardium in the adaptive immune response stage. There are few studies on the early innate immunity of CVB3 infection in myocardial tissue, but the appearance of macrophages in the early stage of CVB3 infection suggests that they can play a regulatory role as early innate immune response cells in myocardial tissue. Here, we discovered a possible new mechanism of VMC caused by CVB3, revealed new drug targets for anti-CVB3, and discovered the therapeutic potential of citrulline for VMC.</p>","PeriodicalId":17583,"journal":{"name":"Journal of Virology","volume":null,"pages":null},"PeriodicalIF":4.0,"publicationDate":"2024-08-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142080664","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Respiratory syncytial virus infection provides protection against severe acute respiratory syndrome coronavirus challenge. 呼吸道合胞病毒感染可防止严重急性呼吸系统综合征冠状病毒的挑战。
IF 4 2区 医学 Q2 VIROLOGY Pub Date : 2024-08-28 DOI: 10.1128/jvi.00669-24
Stacey M Hartwig, Abby Odle, Lok-Yin Roy Wong, David K Meyerholz, Stanley Perlman, Steven M Varga

Respiratory infections are a major health burden worldwide. Respiratory syncytial virus (RSV) is among the leading causes of hospitalization in both young children and older adults. The onset of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic and the public health response had a profound impact on the normal seasonal outbreaks of other respiratory viruses. However, little is known about how a prior respiratory virus infection impacts SARS-CoV-2 disease outcomes. In this study, we examine the impact of a previous RSV infection on the disease severity of a subsequent SARS-CoV-2 challenge in BALB/c mice. Mice infected with RSV, followed by a SARS-CoV-2 challenge, 30 days later, exhibited decreased weight loss and increased survival as compared to control groups. Our results suggest a prior RSV infection can provide protection against a subsequent SARS-CoV-2 infection.

Importance: Severe acute respiratory syndrome coronavirus 2 and respiratory syncytial virus are respiratory viruses that are a major health burden worldwide. Severe acute respiratory syndrome coronavirus 2 and respiratory syncytial virus frequently have peak seasonal outbreaks during the winter months, and are capable of causing severe respiratory disease, often leading to hospitalization. The 2019 pandemic brought attention to the importance of understanding how co-circulating viruses can impact the disease severity of other respiratory viruses. It is known that many hospitalized patients are undergoing multiple viral infections at once, yet not much has been studied to understand the impact this has on other respiratory viruses or patients. How co-circulating viruses impact one another can provide critical knowledge for future interventions of hospitalized patients and potential vaccination strategies.

呼吸道感染是全世界的主要健康负担。呼吸道合胞病毒(RSV)是导致幼儿和老年人住院治疗的主要原因之一。严重急性呼吸系统综合征冠状病毒 2(SARS-CoV-2)大流行的爆发和公共卫生应对措施对其他呼吸道病毒的正常季节性爆发产生了深远影响。然而,人们对之前的呼吸道病毒感染如何影响 SARS-CoV-2 的疾病结果知之甚少。在本研究中,我们研究了之前的 RSV 感染对 BALB/c 小鼠随后的 SARS-CoV-2 挑战的疾病严重程度的影响。与对照组相比,感染了 RSV 并在 30 天后接受了 SARS-CoV-2 病毒挑战的小鼠体重减轻,存活率提高。我们的研究结果表明,先感染 RSV 可为随后感染 SARS-CoV-2 提供保护:重要意义:严重急性呼吸系统综合征冠状病毒 2 和呼吸道合胞病毒是呼吸道病毒,是全球主要的健康负担。严重急性呼吸道综合征冠状病毒 2 和呼吸道合胞病毒经常在冬季出现季节性暴发高峰,可引起严重的呼吸道疾病,往往导致住院治疗。2019 年的流感大流行使人们开始关注了解共循环病毒如何影响其他呼吸道病毒疾病严重程度的重要性。众所周知,许多住院病人都会同时受到多种病毒感染,但人们对这种情况对其他呼吸道病毒或病人的影响了解不多。共同传播的病毒如何相互影响,可为未来对住院患者的干预和潜在的疫苗接种策略提供重要的知识。
{"title":"Respiratory syncytial virus infection provides protection against severe acute respiratory syndrome coronavirus challenge.","authors":"Stacey M Hartwig, Abby Odle, Lok-Yin Roy Wong, David K Meyerholz, Stanley Perlman, Steven M Varga","doi":"10.1128/jvi.00669-24","DOIUrl":"https://doi.org/10.1128/jvi.00669-24","url":null,"abstract":"<p><p>Respiratory infections are a major health burden worldwide. Respiratory syncytial virus (RSV) is among the leading causes of hospitalization in both young children and older adults. The onset of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic and the public health response had a profound impact on the normal seasonal outbreaks of other respiratory viruses. However, little is known about how a prior respiratory virus infection impacts SARS-CoV-2 disease outcomes. In this study, we examine the impact of a previous RSV infection on the disease severity of a subsequent SARS-CoV-2 challenge in BALB/c mice. Mice infected with RSV, followed by a SARS-CoV-2 challenge, 30 days later, exhibited decreased weight loss and increased survival as compared to control groups. Our results suggest a prior RSV infection can provide protection against a subsequent SARS-CoV-2 infection.</p><p><strong>Importance: </strong>Severe acute respiratory syndrome coronavirus 2 and respiratory syncytial virus are respiratory viruses that are a major health burden worldwide. Severe acute respiratory syndrome coronavirus 2 and respiratory syncytial virus frequently have peak seasonal outbreaks during the winter months, and are capable of causing severe respiratory disease, often leading to hospitalization. The 2019 pandemic brought attention to the importance of understanding how co-circulating viruses can impact the disease severity of other respiratory viruses. It is known that many hospitalized patients are undergoing multiple viral infections at once, yet not much has been studied to understand the impact this has on other respiratory viruses or patients. How co-circulating viruses impact one another can provide critical knowledge for future interventions of hospitalized patients and potential vaccination strategies.</p>","PeriodicalId":17583,"journal":{"name":"Journal of Virology","volume":null,"pages":null},"PeriodicalIF":4.0,"publicationDate":"2024-08-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142080759","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Journal of Virology
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1