首页 > 最新文献

Journal of Virology最新文献

英文 中文
Nectin1 is a pivotal host factor involved in attachment and entry of red-spotted grouper nervous necrosis virus in the early stages of the viral life cycle. 在病毒生命周期的早期阶段,Nectin1 是参与红点石斑鱼神经坏死病毒附着和进入的关键宿主因子。
IF 4 2区 医学 Q2 VIROLOGY Pub Date : 2024-08-28 DOI: 10.1128/jvi.00901-24
Zhiqi Zhang, Jing Xing, Xiaoqian Tang, Xiuzhen Sheng, Heng Chi, Wenbin Zhan

Nervous necrosis virus (NNV) is a highly neurotropic virus that poses a persistent threat to the survival of multiple fish species. However, its inimitable neuropathogenesis remains largely elusive. To rummage potential partners germane to the nervous system, we investigated the interaction between red-spotted grouper NNV (RGNNV) and grouper brain by immunoprecipitation coupled with mass spectrometry and discerned Nectin1 as a novel host factor subtly involved in viral early invasion events. Nectin1 was abundant in neural tissues and implicated in the inception of tunnel nanotubes triggered by RGNNV. Its overexpression not only dramatically potentiated the replication dynamics of RGNNV in susceptible cells, but also empowered non-sensitive cells to expeditiously capture free virions within 2 min. This potency was impervious to low temperatures but was dose-dependently suppressed by soluble protein or specific antibody of Nectin1 ectodomain, indicating Nectin1 as an attachment receptor for RGNNV. Mechanistically, efficient hijacking of virions by Nectin1 strictly depended on intricate linkages to different modules of viral capsid protein, especially the direct binding between the IgC1 loop and P-domain. More strikingly, despite abortive proliferation in Nectin1-reconstructed CHSE-214 cells, a non-sensitive cell, RGNNV could gain access to the intracellular compartment by capitalizing on Nectin1, thereby inducing canonical cytoplasmic vacuolation. Altogether, our findings delineate a candidate entrance for RGNNV infiltration into the nervous system, which may shed unprecedented insights into the exploration and elucidation of RGNNV pathogenesis.IMPORTANCENervous necrosis virus (NNV) is one of the most virulent pathogens in the aquaculture industry, which inflicts catastrophic damage to ecology, environment, and economy annually around the world. Nevertheless, its idiosyncratic invasion and latency mechanisms pose enormous hardships to epidemic prevention and control. In this study, deploying grouper brain as a natural screening library, a single-transmembrane glycoprotein, Nectin1, was first identified as an emergent functional receptor for red-spotted grouper NNV (RGNNV) that widely allocated in nervous tissues and directly interacted with viral capsid protein through distinct Ig-like loops to bridge virus-host crosstalk, apprehend free virions, and concomitantly propel viral entry. Our findings illuminate the critical role of Nectin1 in RGNNV attachment and entry and provide a potential target for future clinical intervention strategies in the therapeutic race against RGNNV.

神经坏死病毒(NNV)是一种高度神经性病毒,对多种鱼类的生存构成持续威胁。然而,其独特的神经发病机制在很大程度上仍然难以捉摸。为了寻找与神经系统相关的潜在伙伴,我们通过免疫沉淀与质谱联用技术研究了红点石斑鱼 NNV(RGNNV)与石斑鱼大脑之间的相互作用,发现 Nectin1 是一种新型宿主因子,微妙地参与了病毒的早期入侵事件。Nectin1在神经组织中含量丰富,与RGNNV引发的隧道纳米管的形成有关。过表达 Nectin1 不仅能显著增强 RGNNV 在易感细胞中的复制动态,还能使非易感细胞在 2 分钟内迅速捕获游离病毒。这种效力不受低温影响,但受可溶性蛋白或 Nectin1 外显子特异性抗体的剂量依赖性抑制,表明 Nectin1 是 RGNNV 的附着受体。从机理上讲,Nectin1对病毒的有效劫持严格依赖于与病毒帽蛋白不同模块的复杂联系,特别是IgC1环和P域之间的直接结合。更令人震惊的是,尽管在 Nectin1 重组的 CHSE-214 细胞(一种非敏感细胞)中增殖失败,RGNNV 仍能利用 Nectin1 进入细胞内,从而诱导典型的细胞质空泡化。重要意义神经坏死病毒(NNV)是水产养殖业中毒性最强的病原体之一,每年都会对世界各地的生态、环境和经济造成灾难性的破坏。然而,其特异的入侵和潜伏机制给疫病防控带来了巨大困难。本研究以石斑鱼脑为天然筛选库,首次发现了一种单跨膜糖蛋白--Nectin1,它是红点石斑鱼NNV(RGNNV)的新兴功能受体,广泛分布于神经组织中,并通过独特的Ig样环与病毒帽蛋白直接相互作用,以弥合病毒-宿主间的串扰,捕获游离病毒,并同时推动病毒进入。我们的研究结果阐明了 Nectin1 在 RGNNV 附着和进入过程中的关键作用,并为未来 RGNNV 治疗竞赛中的临床干预策略提供了潜在靶点。
{"title":"Nectin1 is a pivotal host factor involved in attachment and entry of red-spotted grouper nervous necrosis virus in the early stages of the viral life cycle.","authors":"Zhiqi Zhang, Jing Xing, Xiaoqian Tang, Xiuzhen Sheng, Heng Chi, Wenbin Zhan","doi":"10.1128/jvi.00901-24","DOIUrl":"https://doi.org/10.1128/jvi.00901-24","url":null,"abstract":"<p><p>Nervous necrosis virus (NNV) is a highly neurotropic virus that poses a persistent threat to the survival of multiple fish species. However, its inimitable neuropathogenesis remains largely elusive. To rummage potential partners germane to the nervous system, we investigated the interaction between red-spotted grouper NNV (RGNNV) and grouper brain by immunoprecipitation coupled with mass spectrometry and discerned Nectin1 as a novel host factor subtly involved in viral early invasion events. Nectin1 was abundant in neural tissues and implicated in the inception of tunnel nanotubes triggered by RGNNV. Its overexpression not only dramatically potentiated the replication dynamics of RGNNV in susceptible cells, but also empowered non-sensitive cells to expeditiously capture free virions within 2 min. This potency was impervious to low temperatures but was dose-dependently suppressed by soluble protein or specific antibody of Nectin1 ectodomain, indicating Nectin1 as an attachment receptor for RGNNV. Mechanistically, efficient hijacking of virions by Nectin1 strictly depended on intricate linkages to different modules of viral capsid protein, especially the direct binding between the IgC1 loop and P-domain. More strikingly, despite abortive proliferation in Nectin1-reconstructed CHSE-214 cells, a non-sensitive cell, RGNNV could gain access to the intracellular compartment by capitalizing on Nectin1, thereby inducing canonical cytoplasmic vacuolation. Altogether, our findings delineate a candidate entrance for RGNNV infiltration into the nervous system, which may shed unprecedented insights into the exploration and elucidation of RGNNV pathogenesis.IMPORTANCENervous necrosis virus (NNV) is one of the most virulent pathogens in the aquaculture industry, which inflicts catastrophic damage to ecology, environment, and economy annually around the world. Nevertheless, its idiosyncratic invasion and latency mechanisms pose enormous hardships to epidemic prevention and control. In this study, deploying grouper brain as a natural screening library, a single-transmembrane glycoprotein, Nectin1, was first identified as an emergent functional receptor for red-spotted grouper NNV (RGNNV) that widely allocated in nervous tissues and directly interacted with viral capsid protein through distinct Ig-like loops to bridge virus-host crosstalk, apprehend free virions, and concomitantly propel viral entry. Our findings illuminate the critical role of Nectin1 in RGNNV attachment and entry and provide a potential target for future clinical intervention strategies in the therapeutic race against RGNNV.</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":"142080756","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 cellular protein phosphatase 2A is a crucial host factor for Marburg virus transcription. 细胞蛋白磷酸酶 2A 是马尔堡病毒转录的关键宿主因子。
IF 4 2区 医学 Q2 VIROLOGY Pub Date : 2024-08-28 DOI: 10.1128/jvi.01047-24
Isabel von Creytz, Cornelius Rohde, Nadine Biedenkopf

Little is known regarding the molecular mechanisms that highly pathogenic Marburg virus (MARV) utilizes to transcribe and replicate its genome. Previous studies assumed that dephosphorylation of the filoviral transcription factor VP30 supports transcription, while phosphorylated VP30 reduces transcription. Here, we focused on the role of the host protein phosphatase 2A (PP2A) for VP30 dephosphorylation and promotion of viral transcription. We could show that MARV NP interacts with the subunit B56 of PP2A, as previously shown for the Ebola virus, and that this interaction is important for MARV transcription activity. Inhibition of the interaction between PP2A and NP either by mutating the B56 binding motif encoded on NP, or the use of a PP2A inhibitor, induced VP30 hyperphosphorylation, and as a consequence a decrease of MARV transcription as well as viral growth. These results suggest that NP plays a key role in the dephosphorylation of VP30 by recruiting PP2A. Generation of recombinant (rec) MARV lacking the PP2A-B56 interaction motif on NP was not possible suggesting an essential role of PP2A-mediated VP30 dephosphorylation for the MARV replication cycle. Likewise, we were not able to generate recMARV containing VP30 phosphomimetic mutants indicating that dynamic cycles of VP30 de- and rephosphorylation are a prerequisite for an efficient viral life cycle. As the specific binding motifs of PP2A-B56 and VP30 within NP are highly conserved among the filoviral family, our data suggest a conserved mechanism for filovirus VP30 dephosphorylation by PP2A, revealing the host factor PP2A as a promising target for pan-filoviral therapies.

Importance: Our study elucidates the crucial role of host protein phosphatase 2A (PP2A) in Marburg virus (MARV) transcription. The regulatory subunit B56 of PP2A facilitates VP30 dephosphorylation, and hence transcription activation, via binding to NP. Our results, together with previous data, reveal a conserved mechanism of filovirus VP30 dephosphorylation by host factor PP2A at the NP interface and provide novel insights into potential pan-filovirus therapies.

人们对高致病性马尔堡病毒(MARV)转录和复制其基因组的分子机制知之甚少。以前的研究假设丝状病毒转录因子 VP30 的去磷酸化支持转录,而磷酸化的 VP30 则减少转录。在这里,我们重点研究了宿主蛋白磷酸酶 2A(PP2A)在 VP30 去磷酸化和促进病毒转录中的作用。我们可以证明,MARV NP 与 PP2A 的 B56 亚基相互作用,正如之前埃博拉病毒所显示的那样,这种相互作用对 MARV 的转录活性非常重要。通过突变 NP 上编码的 B56 结合位点或使用 PP2A 抑制剂来抑制 PP2A 与 NP 之间的相互作用,可诱导 VP30 过度磷酸化,从而降低 MARV 的转录和病毒的生长。这些结果表明,NP 通过招募 PP2A 在 VP30 的去磷酸化过程中发挥了关键作用。无法生成缺乏 NP 上 PP2A-B56 相互作用基团的重组 MARV,这表明 PP2A 介导的 VP30 去磷酸化在 MARV 复制周期中起着至关重要的作用。同样,我们也无法产生含有 VP30 磷酸化拟态突变体的 recMARV,这表明 VP30 去磷酸化和再磷酸化的动态循环是高效病毒生命周期的先决条件。由于PP2A-B56和VP30在NP中的特异性结合基团在丝状病毒家族中高度保守,我们的数据表明丝状病毒VP30被PP2A去磷酸化的机制是保守的,揭示了宿主因子PP2A是泛丝状病毒疗法的一个有希望的靶点:我们的研究阐明了宿主蛋白磷酸酶2A(PP2A)在马尔堡病毒(MARV)转录中的关键作用。PP2A 的调节亚基 B56 通过与 NP 结合促进 VP30 去磷酸化,从而激活转录。我们的研究结果以及之前的数据揭示了丝状病毒 VP30 在 NP 界面被宿主因子 PP2A 去磷酸化的保守机制,并为潜在的泛丝状病毒疗法提供了新的见解。
{"title":"The cellular protein phosphatase 2A is a crucial host factor for Marburg virus transcription.","authors":"Isabel von Creytz, Cornelius Rohde, Nadine Biedenkopf","doi":"10.1128/jvi.01047-24","DOIUrl":"https://doi.org/10.1128/jvi.01047-24","url":null,"abstract":"<p><p>Little is known regarding the molecular mechanisms that highly pathogenic Marburg virus (MARV) utilizes to transcribe and replicate its genome. Previous studies assumed that dephosphorylation of the filoviral transcription factor VP30 supports transcription, while phosphorylated VP30 reduces transcription. Here, we focused on the role of the host protein phosphatase 2A (PP2A) for VP30 dephosphorylation and promotion of viral transcription. We could show that MARV NP interacts with the subunit B56 of PP2A, as previously shown for the Ebola virus, and that this interaction is important for MARV transcription activity. Inhibition of the interaction between PP2A and NP either by mutating the B56 binding motif encoded on NP, or the use of a PP2A inhibitor, induced VP30 hyperphosphorylation, and as a consequence a decrease of MARV transcription as well as viral growth. These results suggest that NP plays a key role in the dephosphorylation of VP30 by recruiting PP2A. Generation of recombinant (rec) MARV lacking the PP2A-B56 interaction motif on NP was not possible suggesting an essential role of PP2A-mediated VP30 dephosphorylation for the MARV replication cycle. Likewise, we were not able to generate recMARV containing VP30 phosphomimetic mutants indicating that dynamic cycles of VP30 de- and rephosphorylation are a prerequisite for an efficient viral life cycle. As the specific binding motifs of PP2A-B56 and VP30 within NP are highly conserved among the filoviral family, our data suggest a conserved mechanism for filovirus VP30 dephosphorylation by PP2A, revealing the host factor PP2A as a promising target for pan-filoviral therapies.</p><p><strong>Importance: </strong>Our study elucidates the crucial role of host protein phosphatase 2A (PP2A) in Marburg virus (MARV) transcription. The regulatory subunit B56 of PP2A facilitates VP30 dephosphorylation, and hence transcription activation, via binding to NP. Our results, together with previous data, reveal a conserved mechanism of filovirus VP30 dephosphorylation by host factor PP2A at the NP interface and provide novel insights into potential pan-filovirus therapies.</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":"142080762","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
Pan-beta-coronavirus subunit vaccine prevents SARS-CoV-2 Omicron, SARS-CoV, and MERS-CoV challenge. 泛β-冠状病毒亚单位疫苗可预防 SARS-CoV-2 Omicron、SARS-CoV 和 MERS-CoV 的挑战。
IF 4 2区 医学 Q2 VIROLOGY Pub Date : 2024-08-27 DOI: 10.1128/jvi.00376-24
Gang Wang, Abhishek K Verma, Xiaoqing Guan, Fan Bu, Abby E Odle, Fang Li, Bin Liu, Stanley Perlman, Lanying Du

Three highly pathogenic coronaviruses (CoVs), SARS-CoV-2, SARS-CoV, and MERS-CoV, belonging to the genus beta-CoV, have caused outbreaks or pandemics. SARS-CoV-2 has evolved into many variants with increased resistance to the current vaccines. Spike (S) protein and its receptor-binding domain (RBD) fragment of these CoVs are important vaccine targets; however, the RBD of the SARS-CoV-2 Omicron variant is highly mutated, rending neutralizing antibodies elicited by ancestral-based vaccines targeting this region ineffective, emphasizing the need for effective vaccines with broad-spectrum efficacy against SARS-CoV-2 variants and other CoVs with pandemic potential. This study describes a pan-beta-CoV subunit vaccine, Om-S-MERS-RBD, by fusing the conserved and highly potent RBD of MERS-CoV into an RBD-truncated SARS-CoV-2 Omicron S protein, and evaluates its neutralizing immunogenicity and protective efficacy in mouse models. Om-S-MERS-RBD formed a conformational structure, maintained effective functionality and antigenicity, and bind efficiently to MERS-CoV receptor, human dipeptidyl peptidase 4, and MERS-CoV RBD or SARS-CoV-2 S-specific antibodies. Immunization of mice with Om-S-MERS-RBD and adjuvants (Alum plus monophosphoryl lipid A) induced broadly neutralizing antibodies against pseudotyped MERS-CoV, SARS-CoV, and SARS-CoV-2 original strain, as well as T-cell responses specific to RBD-truncated Omicron S protein. Moreover, the neutralizing activity against SARS-CoV-2 Omicron subvariants was effectively improved after priming with an Omicron-S-RBD protein. Adjuvanted Om-S-MERS-RBD protein protected mice against challenge with SARS-CoV-2 Omicron variant, MERS-CoV, and SARS-CoV, significantly reducing viral titers in the lungs. Overall, these findings indicated that Om-S-MERS-RBD protein could develop as an effective universal subunit vaccine to prevent infections with MERS-CoV, SARS-CoV, SARS-CoV-2, and its variants.

Importance: Coronaviruses (CoVs), SARS-CoV-2, SARS-CoV, and MERS-CoV, the respective causative agents of coronavirus disease 2019, SARS, and MERS, continually threaten human health. The spike (S) protein and its receptor-binding domain (RBD) fragment of these CoVs are critical vaccine targets. Nevertheless, the highly mutated RBD of SARS-CoV-2 variants, especially Omicron, significantly reduces the efficacy of current vaccines against SARS-CoV-2 variants. Here a protein-based pan-beta-CoV subunit vaccine is designed by fusing the potent and conserved RBD of MERS-CoV into an RBD-truncated Omicron S protein. The resulting vaccine maintained effective functionality and antigenicity, induced broadly neutralizing antibodies against all of these highly pathogenic human CoVs, and elicited Omicron S-specific cellular immune responses, protecting immunized mice from SARS-CoV-2 Omicron, SARS-CoV, and MERS-CoV infections. Taken together, this study rationally designed a pan-beta-CoV subunit vaccine with broad-spectrum

三种高致病性冠状病毒(CoVs),即 SARS-CoV-2、SARS-CoV 和 MERS-CoV,都属于 beta-CoV 属,曾引起过疾病爆发或流行。SARS-CoV-2 已经进化成许多变种,对现有疫苗的抗药性增强。这些CoV的尖峰(S)蛋白及其受体结合域(RBD)片段是重要的疫苗靶标;然而,SARS-CoV-2 Omicron变体的RBD发生了高度突变,使得以该区域为靶标的祖传疫苗所激发的中和抗体失效,这就强调了需要针对SARS-CoV-2变体和其他具有大流行潜力的CoV研制具有广谱疗效的有效疫苗。本研究介绍了一种泛β-CoV亚单位疫苗 Om-S-MERS-RBD,它将 MERS-CoV 保守的高效力 RBD 与 RBD 截短的 SARS-CoV-2 Omicron S 蛋白融合,并在小鼠模型中评估了其中和免疫原性和保护效力。Om-S-MERS-RBD形成了构象结构,保持了有效的功能性和抗原性,并能与MERS-CoV受体、人二肽基肽酶4、MERS-CoV RBD或SARS-CoV-2 S特异性抗体有效结合。用 Om-S-MERS-RBD 和佐剂(明矾加单磷脂 A)对小鼠进行免疫,可诱导针对伪型 MERS-CoV、SARS-CoV 和 SARS-CoV-2 原始株的广泛中和抗体,以及针对 RBD 截断的 Omicron S 蛋白的特异性 T 细胞反应。此外,以 Omicron-S-RBD 蛋白为引物后,针对 SARS-CoV-2 Omicron 亚变种的中和活性也得到了有效提高。Om-S-MERS-RBD蛋白能保护小鼠免受SARS-CoV-2 Omicron变体、MERS-CoV和SARS-CoV的挑战,显著降低肺部的病毒滴度。总之,这些研究结果表明,Om-S-MERS-RBD 蛋白可作为一种有效的通用亚单位疫苗,用于预防 MERS-CoV、SARS-CoV、SARS-CoV-2 及其变种的感染:冠状病毒(CoVs)、SARS-CoV-2、SARS-CoV 和 MERS-CoV 是 2019 年冠状病毒病、SARS 和 MERS 的病原体,它们不断威胁着人类健康。这些冠状病毒的尖峰(S)蛋白及其受体结合域(RBD)片段是关键的疫苗靶标。然而,SARS-CoV-2 变体(尤其是 Omicron)的 RBD 高度变异,大大降低了当前疫苗对 SARS-CoV-2 变体的效力。本文通过将 MERS-CoV 的强效保守 RBD 与 RBD 截断的 Omicron S 蛋白融合,设计了一种基于蛋白质的泛乙型 CoV 亚基疫苗。由此产生的疫苗保持了有效的功能性和抗原性,诱导了针对所有这些高致病性人类 CoV 的广泛中和抗体,并激发了 Omicron S 特异性细胞免疫反应,保护免疫小鼠免受 SARS-CoV-2 Omicron、SARS-CoV 和 MERS-CoV 感染。综上所述,本研究合理地设计了一种具有广谱效力的泛β-CoV亚单位疫苗,有望开发成一种有效的通用疫苗,用于预防SARS-CoV-2变种和其他具有大流行潜力的CoV。
{"title":"Pan-beta-coronavirus subunit vaccine prevents SARS-CoV-2 Omicron, SARS-CoV, and MERS-CoV challenge.","authors":"Gang Wang, Abhishek K Verma, Xiaoqing Guan, Fan Bu, Abby E Odle, Fang Li, Bin Liu, Stanley Perlman, Lanying Du","doi":"10.1128/jvi.00376-24","DOIUrl":"https://doi.org/10.1128/jvi.00376-24","url":null,"abstract":"<p><p>Three highly pathogenic coronaviruses (CoVs), SARS-CoV-2, SARS-CoV, and MERS-CoV, belonging to the genus beta-CoV, have caused outbreaks or pandemics. SARS-CoV-2 has evolved into many variants with increased resistance to the current vaccines. Spike (S) protein and its receptor-binding domain (RBD) fragment of these CoVs are important vaccine targets; however, the RBD of the SARS-CoV-2 Omicron variant is highly mutated, rending neutralizing antibodies elicited by ancestral-based vaccines targeting this region ineffective, emphasizing the need for effective vaccines with broad-spectrum efficacy against SARS-CoV-2 variants and other CoVs with pandemic potential. This study describes a pan-beta-CoV subunit vaccine, Om-S-MERS-RBD, by fusing the conserved and highly potent RBD of MERS-CoV into an RBD-truncated SARS-CoV-2 Omicron S protein, and evaluates its neutralizing immunogenicity and protective efficacy in mouse models. Om-S-MERS-RBD formed a conformational structure, maintained effective functionality and antigenicity, and bind efficiently to MERS-CoV receptor, human dipeptidyl peptidase 4, and MERS-CoV RBD or SARS-CoV-2 S-specific antibodies. Immunization of mice with Om-S-MERS-RBD and adjuvants (Alum plus monophosphoryl lipid A) induced broadly neutralizing antibodies against pseudotyped MERS-CoV, SARS-CoV, and SARS-CoV-2 original strain, as well as T-cell responses specific to RBD-truncated Omicron S protein. Moreover, the neutralizing activity against SARS-CoV-2 Omicron subvariants was effectively improved after priming with an Omicron-S-RBD protein. Adjuvanted Om-S-MERS-RBD protein protected mice against challenge with SARS-CoV-2 Omicron variant, MERS-CoV, and SARS-CoV, significantly reducing viral titers in the lungs. Overall, these findings indicated that Om-S-MERS-RBD protein could develop as an effective universal subunit vaccine to prevent infections with MERS-CoV, SARS-CoV, SARS-CoV-2, and its variants.</p><p><strong>Importance: </strong>Coronaviruses (CoVs), SARS-CoV-2, SARS-CoV, and MERS-CoV, the respective causative agents of coronavirus disease 2019, SARS, and MERS, continually threaten human health. The spike (S) protein and its receptor-binding domain (RBD) fragment of these CoVs are critical vaccine targets. Nevertheless, the highly mutated RBD of SARS-CoV-2 variants, especially Omicron, significantly reduces the efficacy of current vaccines against SARS-CoV-2 variants. Here a protein-based pan-beta-CoV subunit vaccine is designed by fusing the potent and conserved RBD of MERS-CoV into an RBD-truncated Omicron S protein. The resulting vaccine maintained effective functionality and antigenicity, induced broadly neutralizing antibodies against all of these highly pathogenic human CoVs, and elicited Omicron S-specific cellular immune responses, protecting immunized mice from SARS-CoV-2 Omicron, SARS-CoV, and MERS-CoV infections. Taken together, this study rationally designed a pan-beta-CoV subunit vaccine with broad-spectrum","PeriodicalId":17583,"journal":{"name":"Journal of Virology","volume":null,"pages":null},"PeriodicalIF":4.0,"publicationDate":"2024-08-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142073180","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
Small hydrophobic (SH) proteins of Pneumoviridae and Paramyxoviridae: small but mighty. 肺炎病毒科和副粘病毒科的小疏水(SH)蛋白:小而强大。
IF 4 2区 医学 Q2 VIROLOGY Pub Date : 2024-08-23 DOI: 10.1128/jvi.00809-24
Adam Brynes, John V Williams

Small hydrophobic (SH) proteins are a class of viral accessory proteins expressed by many members of the negative-stranded RNA viral families Paramyxoviridae and Pneumoviridae. Identified SH proteins are type I or II transmembrane (TM) proteins with a single-pass TM domain. Little is known about the functions of SH proteins; however, several possess viroporin activity, enhancing membrane permeability of infected cells or those expressing SH protein. Moreover, several SH proteins inhibit apoptosis and immune signaling pathways within infected cells, including TNF and interferon signaling, or activate inflammasomes. SH proteins are generally nonessential for viral replication in vitro, but loss of SH is often associated with reduced replication in vivo, suggesting a role in enhancing viral replication or evading host immunity. Analogous proteins are expressed by a variety of pathogens of public health importance; thus, understanding the functional importance and mechanisms of SH proteins provides insight into the pathogenesis and replication of negative-sense RNA viruses.

小疏水(SH)蛋白是负链 RNA 病毒科 Paramyxoviridae 和 Pneumoviridae 中许多成员表达的一类病毒附属蛋白。已发现的 SH 蛋白是具有单通 TM 结构域的 I 型或 II 型跨膜(TM)蛋白。人们对 SH 蛋白的功能知之甚少;不过,有几种 SH 蛋白具有病毒蛋白活性,能增强受感染细胞或表达 SH 蛋白的细胞膜的通透性。此外,几种 SH 蛋白还能抑制受感染细胞内的凋亡和免疫信号通路,包括 TNF 和干扰素信号,或激活炎性体。SH 蛋白通常对病毒的体外复制并不重要,但 SH 蛋白的缺失往往与体内复制的减少有关,这表明它在增强病毒复制或逃避宿主免疫方面发挥了作用。对公共卫生具有重要意义的多种病原体都表达类似的蛋白;因此,了解 SH 蛋白的功能重要性和机制有助于深入了解负意义 RNA 病毒的致病机理和复制。
{"title":"Small hydrophobic (SH) proteins of <i>Pneumoviridae</i> and <i>Paramyxoviridae</i>: small but mighty.","authors":"Adam Brynes, John V Williams","doi":"10.1128/jvi.00809-24","DOIUrl":"https://doi.org/10.1128/jvi.00809-24","url":null,"abstract":"<p><p>Small hydrophobic (SH) proteins are a class of viral accessory proteins expressed by many members of the negative-stranded RNA viral families <i>Paramyxoviridae and Pneumoviridae</i>. Identified SH proteins are type I or II transmembrane (TM) proteins with a single-pass TM domain. Little is known about the functions of SH proteins; however, several possess viroporin activity, enhancing membrane permeability of infected cells or those expressing SH protein. Moreover, several SH proteins inhibit apoptosis and immune signaling pathways within infected cells, including TNF and interferon signaling, or activate inflammasomes. SH proteins are generally nonessential for viral replication <i>in vitro</i>, but loss of SH is often associated with reduced replication <i>in vivo</i>, suggesting a role in enhancing viral replication or evading host immunity. Analogous proteins are expressed by a variety of pathogens of public health importance; thus, understanding the functional importance and mechanisms of SH proteins provides insight into the pathogenesis and replication of negative-sense RNA viruses.</p>","PeriodicalId":17583,"journal":{"name":"Journal of Virology","volume":null,"pages":null},"PeriodicalIF":4.0,"publicationDate":"2024-08-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142036208","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
Deciphering the adsorption machinery of Deep-Blue and Vp4, two myophages targeting members of the Bacillus cereus group. 解密 Deep-Blue 和 Vp4(两种以蜡样芽孢杆菌为目标的噬菌体)的吸附机制。
IF 4 2区 医学 Q2 VIROLOGY Pub Date : 2024-08-23 DOI: 10.1128/jvi.00745-24
Manon Nuytten, Audrey Leprince, Adeline Goulet, Jacques Mahillon

In tailed phages, the baseplate is the macromolecular structure located at the tail distal part, which is directly implicated in host recognition and cell wall penetration. In myophages (i.e., with contractile tails), the baseplate is complex and comprises a central puncturing device and baseplate wedges connecting the hub to the receptor-binding proteins (RBPs). In this work, we investigated the structures and functions of adsorption-associated tail proteins of Deep-Blue and Vp4, two Herelleviridae phages infecting members of the Bacillus cereus group. Their interest resides in their different host spectrum despite a high degree of similarity. Analysis of their tail module revealed that the gene order is similar to that of the Listeria phage A511. Among their tail proteins, Gp185 (Deep-Blue) and Gp112 (Vp4) had no structural homolog, but the C-terminal variable parts of these proteins were able to bind B. cereus strains, confirming their implication in the phage adsorption. Interestingly, Vp4 and Deep-Blue adsorption to their hosts was also shown to require polysaccharides, which are likely to be bound by the arsenal of carbohydrate-binding modules (CBMs) of these phages' baseplates, suggesting that the adsorption does not rely solely on the RBPs. In particular, the BW Gp119 (Vp4), harboring a CBM fold, was shown to effectively bind to bacterial cells. Finally, we also showed that the putative baseplate hub proteins (i.e., Deep-Blue Gp189 and Vp4 Gp110) have a bacteriolytic activity against B. cereus strains, which supports their role as ectolysins locally degrading the peptidoglycan to facilitate genome injection.

Importance: The Bacillus cereus group comprises closely related species, including some with pathogenic potential (e.g., Bacillus anthracis and Bacillus cytotoxicus). Their toxins represent the most frequently reported cause of food poisoning outbreaks at the European level. Bacteriophage research is undergoing a remarkable renaissance for its potential in the biocontrol and detection of such pathogens. As the primary site of phage-bacteria interactions and a prerequisite for successful phage infection, adsorption is a crucial process that needs further investigation. The current knowledge about B. cereus phage adsorption is currently limited to siphoviruses and tectiviruses. Here, we present the first insights into the adsorption process of Herelleviridae Vp4 and Deep-Blue myophages preying on B. cereus hosts, highlighting the importance of polysaccharide moieties in this process and confirming the binding to the host surface of Deep-Blue Gp185 and Vp4 Gp112 receptor-binding proteins and Gp119 baseplate wedge.

在有尾噬菌体中,基盘是位于尾部远端的大分子结构,直接参与宿主识别和细胞壁穿透。在噬菌体(即具有收缩尾部的噬菌体)中,基板结构复杂,由中央穿刺装置和连接枢纽与受体结合蛋白(RBPs)的基板楔组成。在这项工作中,我们研究了 Deep-Blue 和 Vp4 的吸附相关尾蛋白的结构和功能,这是两种感染蜡样芽孢杆菌的 Herelleviridae 噬菌体。这两种噬菌体尽管具有高度相似性,但它们的宿主谱却各不相同。对它们尾部模块的分析表明,基因顺序与李斯特菌噬菌体 A511 相似。在它们的尾部蛋白中,Gp185(Deep-Blue)和 Gp112(Vp4)没有结构上的同源物,但这些蛋白的 C 端可变部分能够与蜡样芽孢杆菌菌株结合,证实了它们在噬菌体吸附中的作用。有趣的是,Vp4 和 Deep-Blue 对宿主的吸附也需要多糖,这些噬菌体基板上的碳水化合物结合模块(CBMs)很可能与多糖结合,这表明吸附并不完全依赖于 RBPs。尤其是 BW Gp119(Vp4),它含有一个 CBM 折叠,能有效地与细菌细胞结合。最后,我们还发现,假定的基板枢纽蛋白(即深蓝 Gp189 和 Vp4 Gp110)对蜡样芽孢杆菌菌株具有杀菌活性,这支持了它们作为外溶酶在局部降解肽聚糖以促进基因组注入的作用:重要意义:蜡样芽孢杆菌属由密切相关的物种组成,包括一些具有致病潜力的物种(如炭疽芽孢杆菌和细胞毒性芽孢杆菌)。在欧洲,它们的毒素是导致食物中毒爆发的最常见原因。由于噬菌体在生物控制和检测此类病原体方面的潜力,噬菌体研究正在经历一次引人注目的复兴。作为噬菌体与细菌相互作用的主要场所和噬菌体成功感染的先决条件,吸附是一个需要进一步研究的关键过程。目前,有关蜡样芽孢杆菌噬菌体吸附的知识仅限于虹吸病毒和ectiviruses。在这里,我们首次揭示了噬菌体Vp4和深蓝噬菌体捕食蜡样芽孢杆菌宿主的吸附过程,强调了多糖分子在这一过程中的重要性,并证实了深蓝噬菌体Gp185和Vp4 Gp112受体结合蛋白以及Gp119基板楔与宿主表面的结合。
{"title":"Deciphering the adsorption machinery of Deep-Blue and Vp4, two myophages targeting members of the <i>Bacillus cereus</i> group.","authors":"Manon Nuytten, Audrey Leprince, Adeline Goulet, Jacques Mahillon","doi":"10.1128/jvi.00745-24","DOIUrl":"https://doi.org/10.1128/jvi.00745-24","url":null,"abstract":"<p><p>In tailed phages, the baseplate is the macromolecular structure located at the tail distal part, which is directly implicated in host recognition and cell wall penetration. In myophages (i.e., with contractile tails), the baseplate is complex and comprises a central puncturing device and baseplate wedges connecting the hub to the receptor-binding proteins (RBPs). In this work, we investigated the structures and functions of adsorption-associated tail proteins of Deep-Blue and Vp4, two <i>Herelleviridae</i> phages infecting members of the <i>Bacillus cereus</i> group. Their interest resides in their different host spectrum despite a high degree of similarity. Analysis of their tail module revealed that the gene order is similar to that of the <i>Listeria</i> phage A511. Among their tail proteins, Gp185 (Deep-Blue) and Gp112 (Vp4) had no structural homolog, but the C-terminal variable parts of these proteins were able to bind <i>B. cereus</i> strains, confirming their implication in the phage adsorption. Interestingly, Vp4 and Deep-Blue adsorption to their hosts was also shown to require polysaccharides, which are likely to be bound by the arsenal of carbohydrate-binding modules (CBMs) of these phages' baseplates, suggesting that the adsorption does not rely solely on the RBPs. In particular, the BW Gp119 (Vp4), harboring a CBM fold, was shown to effectively bind to bacterial cells. Finally, we also showed that the putative baseplate hub proteins (i.e., Deep-Blue Gp189 and Vp4 Gp110) have a bacteriolytic activity against <i>B. cereus</i> strains, which supports their role as ectolysins locally degrading the peptidoglycan to facilitate genome injection.</p><p><strong>Importance: </strong>The <i>Bacillus cereus</i> group comprises closely related species, including some with pathogenic potential (e.g., <i>Bacillus anthracis</i> and <i>Bacillus cytotoxicus</i>). Their toxins represent the most frequently reported cause of food poisoning outbreaks at the European level. Bacteriophage research is undergoing a remarkable renaissance for its potential in the biocontrol and detection of such pathogens. As the primary site of phage-bacteria interactions and a prerequisite for successful phage infection, adsorption is a crucial process that needs further investigation. The current knowledge about <i>B. cereus</i> phage adsorption is currently limited to siphoviruses and tectiviruses. Here, we present the first insights into the adsorption process of <i>Herelleviridae</i> Vp4 and Deep-Blue myophages preying on <i>B. cereus</i> hosts, highlighting the importance of polysaccharide moieties in this process and confirming the binding to the host surface of Deep-Blue Gp185 and Vp4 Gp112 receptor-binding proteins and Gp119 baseplate wedge.</p>","PeriodicalId":17583,"journal":{"name":"Journal of Virology","volume":null,"pages":null},"PeriodicalIF":4.0,"publicationDate":"2024-08-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142036207","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
Intranasal administration of octavalent next-generation influenza vaccine elicits protective immune responses against seasonal and pre-pandemic viruses. 鼻内注射八价新一代流感疫苗可引起针对季节性和大流行前病毒的保护性免疫反应。
IF 4 2区 医学 Q2 VIROLOGY Pub Date : 2024-08-22 DOI: 10.1128/jvi.00354-24
Naoko Uno, Thomas Ebensen, Carlos A Guzman, Ted M Ross

Development of next-generation influenza virus vaccines is crucial to improve protection against circulating and emerging viruses. Current vaccine formulations have to be updated annually due to mutations in seasonal strains and do not offer protection against strains with pandemic potential. Computationally optimized broadly reactive antigen (COBRA) methodology has been utilized by our group to generate broadly reactive immunogens for individual influenza subtypes, which elicit protective immune responses against a broad range of strains over numerous seasons. Octavalent mixtures of COBRA hemagglutinin (HA) (H1, H2, H3, H5, H7, and influenza B virus) plus neuraminidase (NA) (N1 and N2) recombinant proteins mixed with c-di-AMP adjuvant were administered intranasally to naive or pre-immune ferrets in prime-boost fashion. Four weeks after final vaccination, collected sera were analyzed for breadth of antibody response, and the animals were challenged with seasonal or pre-pandemic strains. The octavalent COBRA vaccine elicited antibodies that recognized a broad panel of strains representing different subtypes, and these vaccinated animals were protected against influenza virus challenges. Overall, this study demonstrated that the mixture of eight COBRA HA/NA proteins mixed with an intranasal adjuvant is a promising candidate for a universal influenza vaccine.

Importance: Influenza is a respiratory virus which infects around a billion people globally every year, with millions experiencing severe illness. Commercial vaccine efficacy varies year to year and can be low due to mismatch of circulating virus strains. Thus, the formulation of current vaccines has to be adapted accordingly every year. The development of a broadly reactive influenza vaccine would lessen the global economic and public health burden caused by the different types of influenza viruses. The significance of our research is producing a promising universal vaccine candidate which provides protection against a wider range of virus strains over a wider range of time.

开发新一代流感病毒疫苗对于提高对流行病毒和新出现病毒的保护至关重要。由于季节性毒株的变异,目前的疫苗配方每年都需要更新,而且无法提供对潜在大流行毒株的保护。我们的研究小组利用计算优化宽反应性抗原(COBRA)方法生成了针对个别流感亚型的宽反应性免疫原,这些免疫原可在多个季节针对多种毒株引起保护性免疫反应。将 COBRA 血凝素(HA)(H1、H2、H3、H5、H7 和 B 型流感病毒)和神经氨酸苷酶(NA)(N1 和 N2)重组蛋白的八价混合物与 c-di-AMP 佐剂混合,以原初强化方式经鼻注射给天真或预免疫雪貂。最终接种四周后,对收集的血清进行抗体反应广度分析,并用季节性或流行前毒株对动物进行挑战。八价 COBRA 疫苗激发的抗体可识别代表不同亚型的多种毒株,这些接种过疫苗的动物在面对流感病毒挑战时受到了保护。总之,这项研究表明,八种 COBRA HA/NA 蛋白与鼻内佐剂的混合物是一种很有希望的通用流感疫苗候选物:流感是一种呼吸道病毒,每年全球约有十亿人感染,数百万人重病。由于流行病毒株的不匹配,商业疫苗的效力每年都不同,而且可能很低。因此,目前疫苗的配方每年都要进行相应的调整。研发一种反应广泛的流感疫苗将减轻不同类型流感病毒对全球经济和公共卫生造成的负担。我们研究的意义在于生产出一种有希望的通用候选疫苗,它能在更长的时间内对更多的病毒株提供保护。
{"title":"Intranasal administration of octavalent next-generation influenza vaccine elicits protective immune responses against seasonal and pre-pandemic viruses.","authors":"Naoko Uno, Thomas Ebensen, Carlos A Guzman, Ted M Ross","doi":"10.1128/jvi.00354-24","DOIUrl":"https://doi.org/10.1128/jvi.00354-24","url":null,"abstract":"<p><p>Development of next-generation influenza virus vaccines is crucial to improve protection against circulating and emerging viruses. Current vaccine formulations have to be updated annually due to mutations in seasonal strains and do not offer protection against strains with pandemic potential. Computationally optimized broadly reactive antigen (COBRA) methodology has been utilized by our group to generate broadly reactive immunogens for individual influenza subtypes, which elicit protective immune responses against a broad range of strains over numerous seasons. Octavalent mixtures of COBRA hemagglutinin (HA) (H1, H2, H3, H5, H7, and influenza B virus) plus neuraminidase (NA) (N1 and N2) recombinant proteins mixed with c-di-AMP adjuvant were administered intranasally to naive or pre-immune ferrets in prime-boost fashion. Four weeks after final vaccination, collected sera were analyzed for breadth of antibody response, and the animals were challenged with seasonal or pre-pandemic strains. The octavalent COBRA vaccine elicited antibodies that recognized a broad panel of strains representing different subtypes, and these vaccinated animals were protected against influenza virus challenges. Overall, this study demonstrated that the mixture of eight COBRA HA/NA proteins mixed with an intranasal adjuvant is a promising candidate for a universal influenza vaccine.</p><p><strong>Importance: </strong>Influenza is a respiratory virus which infects around a billion people globally every year, with millions experiencing severe illness. Commercial vaccine efficacy varies year to year and can be low due to mismatch of circulating virus strains. Thus, the formulation of current vaccines has to be adapted accordingly every year. The development of a broadly reactive influenza vaccine would lessen the global economic and public health burden caused by the different types of influenza viruses. The significance of our research is producing a promising universal vaccine candidate which provides protection against a wider range of virus strains over a wider range of time.</p>","PeriodicalId":17583,"journal":{"name":"Journal of Virology","volume":null,"pages":null},"PeriodicalIF":4.0,"publicationDate":"2024-08-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142017907","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
AhR ligands from LGG metabolites promote piglet intestinal ILC3 activation and IL-22 secretion to inhibit PEDV infection. LGG 代谢物中的 AhR 配体可促进仔猪肠道 ILC3 的活化和 IL-22 的分泌,从而抑制 PEDV 感染。
IF 4 2区 医学 Q2 VIROLOGY Pub Date : 2024-08-20 Epub Date: 2024-07-16 DOI: 10.1128/jvi.01039-24
Junhong Wang, Yibo Zhao, Tong Cui, Hongyu Bao, Ming Gao, Mingyang Cheng, Yu Sun, Yiyuan Lu, Jiayao Guan, Di Zhang, Yanlong Jiang, Haibin Huang, Chunwei Shi, Jianzhong Wang, Nan Wang, Jingtao Hu, Wentao Yang, Hongxi Qian, Qingrong Jiang, Guilian Yang, Yan Zeng, Chunfeng Wang, Xin Cao

In maintaining organismal homeostasis, gut immunity plays a crucial role. The coordination between the microbiota and the immune system through bidirectional interactions regulates the impact of microorganisms on the host. Our research focused on understanding the relationships between substantial changes in jejunal intestinal flora and metabolites and intestinal immunity during porcine epidemic diarrhea virus (PEDV) infection in piglets. We discovered that Lactobacillus rhamnosus GG (LGG) could effectively prevent PEDV infection in piglets. Further investigation revealed that LGG metabolites interact with type 3 innate lymphoid cells (ILC3s) in the jejunum of piglets through the aryl hydrocarbon receptor (AhR). This interaction promotes the activation of ILC3s and the production of interleukin-22 (IL-22). Subsequently, IL-22 facilitates the proliferation of IPEC-J2 cells and activates the STAT3 signaling pathway, thereby preventing PEDV infection. Moreover, the AhR receptor influences various cell types within organoids, including intestinal stem cells (ISCs), Paneth cells, and enterocytes, to promote their growth and development, suggesting that AhR has a broad impact on intestinal health. In conclusion, our study demonstrated the ability of LGG to modulate intestinal immunity and effectively prevent PEDV infection in piglets. These findings highlight the potential application of LGG as a preventive measure against viral infections in livestock.IMPORTANCEWe observed high expression of the AhR receptor on pig and human ILC3s, although its expression was negligible in mouse ILC3s. ILC3s are closely related to the gut microbiota, particularly the secretion of IL-22 stimulated by microbial signals, which plays a crucial regulatory role in intestinal immunity. In our study, we found that metabolites produced by beneficial gut bacteria interact with ILC3s through AhR, thereby maintaining intestinal immune homeostasis in pigs. Moreover, LGG feeding can enhance the activation of ILC3s and promote IL-22 secretion in the intestines of piglets, ultimately preventing PEDV infection.

在维持机体平衡的过程中,肠道免疫发挥着至关重要的作用。微生物群和免疫系统之间通过双向互动进行协调,调节微生物对宿主的影响。我们的研究重点是了解仔猪感染猪流行性腹泻病毒(PEDV)期间空肠肠道菌群和代谢物的实质性变化与肠道免疫之间的关系。我们发现鼠李糖乳杆菌 GG(LGG)能有效预防仔猪感染 PEDV。进一步研究发现,LGG 的代谢产物通过芳基烃受体(AhR)与仔猪空肠中的 3 型先天性淋巴细胞(ILC3s)相互作用。这种相互作用促进了 ILC3 的活化和白细胞介素-22(IL-22)的产生。随后,IL-22 会促进 IPEC-J2 细胞的增殖并激活 STAT3 信号通路,从而防止 PEDV 感染。此外,AhR受体还能影响器官组织内的各种细胞类型,包括肠干细胞(ISC)、Paneth细胞和肠细胞,促进它们的生长和发育,这表明AhR对肠道健康有着广泛的影响。总之,我们的研究证明了 LGG 调节肠道免疫和有效预防仔猪 PEDV 感染的能力。重要意义我们在猪和人的 ILC3s 上观察到 AhR 受体的高表达,而在小鼠的 ILC3s 上其表达可忽略不计。ILC3 与肠道微生物群密切相关,尤其是微生物信号刺激 IL-22 的分泌,在肠道免疫中发挥着重要的调节作用。在我们的研究中,我们发现有益肠道细菌产生的代谢产物通过 AhR 与 ILC3 相互作用,从而维持猪的肠道免疫平衡。此外,饲喂 LGG 能增强 ILC3 的活化,促进仔猪肠道中 IL-22 的分泌,最终预防 PEDV 感染。
{"title":"AhR ligands from <i>LGG</i> metabolites promote piglet intestinal ILC3 activation and IL-22 secretion to inhibit PEDV infection.","authors":"Junhong Wang, Yibo Zhao, Tong Cui, Hongyu Bao, Ming Gao, Mingyang Cheng, Yu Sun, Yiyuan Lu, Jiayao Guan, Di Zhang, Yanlong Jiang, Haibin Huang, Chunwei Shi, Jianzhong Wang, Nan Wang, Jingtao Hu, Wentao Yang, Hongxi Qian, Qingrong Jiang, Guilian Yang, Yan Zeng, Chunfeng Wang, Xin Cao","doi":"10.1128/jvi.01039-24","DOIUrl":"10.1128/jvi.01039-24","url":null,"abstract":"<p><p>In maintaining organismal homeostasis, gut immunity plays a crucial role. The coordination between the microbiota and the immune system through bidirectional interactions regulates the impact of microorganisms on the host. Our research focused on understanding the relationships between substantial changes in jejunal intestinal flora and metabolites and intestinal immunity during porcine epidemic diarrhea virus (PEDV) infection in piglets. We discovered that <i>Lactobacillus rhamnosus GG</i> (<i>LGG</i>) could effectively prevent PEDV infection in piglets. Further investigation revealed that <i>LGG</i> metabolites interact with type 3 innate lymphoid cells (ILC3s) in the jejunum of piglets through the aryl hydrocarbon receptor (AhR). This interaction promotes the activation of ILC3s and the production of interleukin-22 (IL-22). Subsequently, IL-22 facilitates the proliferation of IPEC-J2 cells and activates the STAT3 signaling pathway, thereby preventing PEDV infection. Moreover, the AhR receptor influences various cell types within organoids, including intestinal stem cells (ISCs), Paneth cells, and enterocytes, to promote their growth and development, suggesting that AhR has a broad impact on intestinal health. In conclusion, our study demonstrated the ability of <i>LGG</i> to modulate intestinal immunity and effectively prevent PEDV infection in piglets. These findings highlight the potential application of <i>LGG</i> as a preventive measure against viral infections in livestock.IMPORTANCEWe observed high expression of the AhR receptor on pig and human ILC3s, although its expression was negligible in mouse ILC3s. ILC3s are closely related to the gut microbiota, particularly the secretion of IL-22 stimulated by microbial signals, which plays a crucial regulatory role in intestinal immunity. In our study, we found that metabolites produced by beneficial gut bacteria interact with ILC3s through AhR, thereby maintaining intestinal immune homeostasis in pigs. Moreover, <i>LGG</i> feeding can enhance the activation of ILC3s and promote IL-22 secretion in the intestines of piglets, ultimately preventing PEDV infection.</p>","PeriodicalId":17583,"journal":{"name":"Journal of Virology","volume":null,"pages":null},"PeriodicalIF":4.0,"publicationDate":"2024-08-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11334530/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141620280","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Cell-surface d-glucuronyl C5-epimerase binds to porcine deltacoronavirus spike protein facilitating viral entry. 细胞表面 d-葡萄糖醛酸 C5-表聚酶与猪 deltacoronavirus 穗状病毒蛋白结合,促进病毒进入细胞。
IF 4 2区 医学 Q2 VIROLOGY Pub Date : 2024-08-20 Epub Date: 2024-07-30 DOI: 10.1128/jvi.00880-24
Wenwen Xiao, Chaoqun Chen, Sijin Xia, Zhuang Li, Tong Ding, Junwei Zhou, Liurong Fang, Puxian Fang, Shaobo Xiao

Porcine deltacoronavirus (PDCoV) is an emerging swine enteric coronavirus with zoonotic potential. The coronavirus spike (S) glycoprotein, especially the S1 subunit, mediates viral entry by binding to cellular receptors. However, the functional receptor of PDCoV remains poorly understood. In this study, we used the soluble PDCoV S1 protein as bait to capture the S1-binding cellular transmembrane proteins in combined immunoprecipitation and mass spectrometry analyses. A single guide RNA screen identified d-glucuronyl C5-epimerase (GLCE), a heparan sulfate-modifying enzyme, as a proviral host factor for PDCoV infection. GLCE knockout significantly inhibited the attachment and internalization stages of PDCoV infection. We also demonstrated the interaction between GLCE and PDCoV S with coimmunoprecipitation in both an overexpression system and PDCoV-infected cells. GLCE could be localized to the cell membrane, and an anti-GLCE antibody suppressed PDCoV infection. Although GLCE expression alone did not render nonpermissive cells susceptible to PDCoV infection, GLCE promoted the binding of PDCoV S to porcine amino peptidase N (pAPN), acting synergistically with pAPN to enhance PDCoV infection. In conclusion, our results demonstrate that GLCE is a novel cell-surface factor facilitating PDCoV entry and provide new insights into PDCoV infection.

Importance: The identification of viral receptors is of great significance, potentially extending our understanding of viral infection and pathogenesis. Porcine deltacoronavirus (PDCoV) is an emerging enteropathogenic coronavirus with the potential for cross-species transmission. However, the receptors or coreceptors of PDCoV are still poorly understood. The present study confirms that d-glucuronyl C5-epimerase (GLCE) is a positive regulator of PDCoV infection, promoting viral attachment and internalization. The anti-GLCE antibody suppressed PDCoV infection. Mechanically, GLCE interacts with PDCoV S and promotes the binding of PDCoV S to porcine amino peptidase N (pAPN), acting synergistically with pAPN to enhance PDCoV infection. This work identifies GLCE as a novel cell-surface factor facilitating PDCoV entry and paves the way for further insights into the mechanisms of PDCoV infection.

猪三角冠状病毒(PDCoV)是一种新出现的猪肠道冠状病毒,具有人畜共患病的潜能。冠状病毒尖峰(S)糖蛋白,尤其是 S1 亚基,通过与细胞受体结合介导病毒进入。然而,人们对 PDCoV 的功能受体仍然知之甚少。在这项研究中,我们以可溶性 PDCoV S1 蛋白为诱饵,在免疫沉淀和质谱分析中捕获与 S1 结合的细胞跨膜蛋白。单导 RNA 筛选确定了 d-葡萄糖醛酸 C5-酰亚胺酶(GLCE),这是一种硫酸肝素修饰酶,是 PDCoV 感染的病毒宿主因子。GLCE 基因敲除能明显抑制 PDCoV 感染的附着和内化阶段。我们还在过表达系统和 PDCoV 感染细胞中通过免疫共沉淀证明了 GLCE 与 PDCoV S 之间的相互作用。GLCE 可定位到细胞膜上,抗 GLCE 抗体可抑制 PDCoV 感染。虽然 GLCE 的单独表达并不会使非顺应性细胞易受 PDCoV 感染,但 GLCE 能促进 PDCoV S 与猪氨基肽酶 N(pAPN)的结合,与 pAPN 协同作用增强 PDCoV 感染。总之,我们的研究结果表明,GLCE 是一种新型的细胞表面因子,可促进 PDCoV 的进入,并为 PDCoV 感染提供了新的见解:重要意义:病毒受体的鉴定意义重大,有可能扩展我们对病毒感染和发病机制的认识。猪三角冠状病毒(PDCoV)是一种新出现的肠道致病性冠状病毒,具有跨物种传播的潜力。然而,人们对 PDCoV 的受体或核心受体仍然知之甚少。本研究证实,d-葡萄糖醛酸 C5-表聚酶(GLCE)是 PDCoV 感染的正调控因子,可促进病毒的附着和内化。抗 GLCE 抗体抑制了 PDCoV 感染。在机制上,GLCE 与 PDCoV S 相互作用,促进 PDCoV S 与猪氨基肽酶 N(pAPN)结合,与 pAPN 协同作用,增强 PDCoV 感染。这项研究发现 GLCE 是一种新型细胞表面因子,可促进 PDCoV 进入细胞,为进一步了解 PDCoV 感染机制铺平了道路。
{"title":"Cell-surface d-glucuronyl C5-epimerase binds to porcine deltacoronavirus spike protein facilitating viral entry.","authors":"Wenwen Xiao, Chaoqun Chen, Sijin Xia, Zhuang Li, Tong Ding, Junwei Zhou, Liurong Fang, Puxian Fang, Shaobo Xiao","doi":"10.1128/jvi.00880-24","DOIUrl":"10.1128/jvi.00880-24","url":null,"abstract":"<p><p>Porcine deltacoronavirus (PDCoV) is an emerging swine enteric coronavirus with zoonotic potential. The coronavirus spike (S) glycoprotein, especially the S1 subunit, mediates viral entry by binding to cellular receptors. However, the functional receptor of PDCoV remains poorly understood. In this study, we used the soluble PDCoV S1 protein as bait to capture the S1-binding cellular transmembrane proteins in combined immunoprecipitation and mass spectrometry analyses. A single guide RNA screen identified d-glucuronyl C5-epimerase (GLCE), a heparan sulfate-modifying enzyme, as a proviral host factor for PDCoV infection. GLCE knockout significantly inhibited the attachment and internalization stages of PDCoV infection. We also demonstrated the interaction between GLCE and PDCoV S with coimmunoprecipitation in both an overexpression system and PDCoV-infected cells. GLCE could be localized to the cell membrane, and an anti-GLCE antibody suppressed PDCoV infection. Although GLCE expression alone did not render nonpermissive cells susceptible to PDCoV infection, GLCE promoted the binding of PDCoV S to porcine amino peptidase N (pAPN), acting synergistically with pAPN to enhance PDCoV infection. In conclusion, our results demonstrate that GLCE is a novel cell-surface factor facilitating PDCoV entry and provide new insights into PDCoV infection.</p><p><strong>Importance: </strong>The identification of viral receptors is of great significance, potentially extending our understanding of viral infection and pathogenesis. Porcine deltacoronavirus (PDCoV) is an emerging enteropathogenic coronavirus with the potential for cross-species transmission. However, the receptors or coreceptors of PDCoV are still poorly understood. The present study confirms that d-glucuronyl C5-epimerase (GLCE) is a positive regulator of PDCoV infection, promoting viral attachment and internalization. The anti-GLCE antibody suppressed PDCoV infection. Mechanically, GLCE interacts with PDCoV S and promotes the binding of PDCoV S to porcine amino peptidase N (pAPN), acting synergistically with pAPN to enhance PDCoV infection. This work identifies GLCE as a novel cell-surface factor facilitating PDCoV entry and paves the way for further insights into the mechanisms of PDCoV infection.</p>","PeriodicalId":17583,"journal":{"name":"Journal of Virology","volume":null,"pages":null},"PeriodicalIF":4.0,"publicationDate":"2024-08-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11334431/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141792788","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
E3 ligase RNF2 inhibits porcine circovirus type 3 replication by targeting its capsid protein for ubiquitination-dependent degradation. E3连接酶RNF2通过靶向猪圆环病毒3型的囊膜蛋白进行泛素依赖性降解来抑制其复制。
IF 4 2区 医学 Q2 VIROLOGY Pub Date : 2024-08-20 Epub Date: 2024-07-24 DOI: 10.1128/jvi.00223-24
Dedong Wang, Jie Zhao, Xiaoyu Yang, Ying Ji, Ju Yu, Zhaoyang Li, Yongyan Shi, Jinshuo Guo, Jianwei Zhou, Lei Hou, Jue Liu

Porcine circovirus type 3 (PCV3) is closely associated with various diseases, such as the porcine dermatitis, nephropathy syndrome, and multisystemic clinicopathological diseases. PCV3-associated diseases are increasingly recognized as severe diseases in the global swine industry. Ring finger protein 2 (RNF2), an E3 ubiquitin ligase exclusively located in the nucleus, contributes to various biological processes. This ligase interacts with the PCV3 Cap. However, its role in PCV3 replication remains unclear. This study confirmed that the nuclear localization signal domain of the Cap and the RNF2 N-terminal RING domain facilitate the interaction between the Cap and RNF2. Furthermore, RNF2 promoted the binding of K48-linked polyubiquitination chains to lysine at positions 139 and 140 (K139 and K140) of the PCV3 Cap, thereby degrading the Cap. RNF2 knockdown and overexpression increased or decreased PCV3 replication, respectively. Moreover, the RING domain-deleted RNF2 mutant eliminated the RNF2-induced degradation of the PCV3 Cap and RNF2-mediated inhibition of viral replication. This indicates that both processes were associated with its E3 ligase activity. Our findings demonstrate that RNF2 can interact with and degrade the PCV3 Cap via its N-terminal RING domain in a ubiquitination-dependent manner, thereby inhibiting PCV3 replication.IMPORTANCEPorcine circovirus type 3 is a recently described pathogen that is prevalent worldwide, causing substantial economic losses to the swine industry. However, the mechanisms through which host proteins regulate its replication remain unclear. Here, we demonstrate that ring finger protein 2 inhibits porcine circovirus type 3 replication by interacting with and degrading the Cap of this pathogen in a ubiquitination-dependent manner, requiring its N-terminal RING domain. Ring finger protein 2-mediated degradation of the Cap relies on its E3 ligase activity and the simultaneous existence of K139 and K140 within the Cap. These findings reveal the mechanism by which this protein interacts with and degrades the Cap to inhibit porcine circovirus type 3 replication. This consequently provides novel insights into porcine circovirus type 3 pathogenesis and facilitates the development of preventative measures against this pathogen.

猪圆环病毒 3 型(PCV3)与多种疾病密切相关,如猪皮炎、肾病综合征和多系统临床病理疾病。在全球养猪业中,越来越多的人认识到 PCV3 相关疾病是一种严重的疾病。环指蛋白 2(RNF2)是一种专门位于细胞核内的 E3 泛素连接酶,对各种生物过程都有贡献。这种连接酶与 PCV3 Cap 相互作用。然而,它在 PCV3 复制中的作用仍不清楚。本研究证实,Cap 的核定位信号结构域和 RNF2 N 端 RING 结构域促进了 Cap 和 RNF2 之间的相互作用。此外,RNF2 促进了 K48 链接的多泛素化链与 PCV3 Cap 的 139 和 140 位赖氨酸(K139 和 K140)的结合,从而降解了 Cap。RNF2 的敲除和过表达分别增加或减少了 PCV3 的复制。此外,RING结构域缺失的RNF2突变体消除了RNF2诱导的PCV3 Cap降解和RNF2介导的病毒复制抑制。这表明这两个过程都与其 E3 连接酶活性有关。我们的研究结果表明,RNF2 可以通过其 N 端 RING 结构域以泛素依赖的方式与 PCV3 Cap 相互作用并降解 PCV3 Cap,从而抑制 PCV3 的复制。然而,宿主蛋白调控其复制的机制仍不清楚。在这里,我们证明环指蛋白 2 能抑制猪圆环病毒 3 型的复制,它以泛素依赖的方式与该病原体的 Cap 相互作用并降解 Cap,需要其 N 端 RING 结构域。环指蛋白 2 介导的 Cap 降解依赖于其 E3 连接酶活性以及 Cap 中同时存在的 K139 和 K140。这些发现揭示了环指蛋白 2 与 Cap 相互作用并降解 Cap 以抑制猪圆环病毒 3 型复制的机制。这为了解猪圆环病毒 3 型的致病机理提供了新的视角,有助于开发针对该病原体的预防措施。
{"title":"E3 ligase RNF2 inhibits porcine circovirus type 3 replication by targeting its capsid protein for ubiquitination-dependent degradation.","authors":"Dedong Wang, Jie Zhao, Xiaoyu Yang, Ying Ji, Ju Yu, Zhaoyang Li, Yongyan Shi, Jinshuo Guo, Jianwei Zhou, Lei Hou, Jue Liu","doi":"10.1128/jvi.00223-24","DOIUrl":"10.1128/jvi.00223-24","url":null,"abstract":"<p><p>Porcine circovirus type 3 (PCV3) is closely associated with various diseases, such as the porcine dermatitis, nephropathy syndrome, and multisystemic clinicopathological diseases. PCV3-associated diseases are increasingly recognized as severe diseases in the global swine industry. Ring finger protein 2 (RNF2), an E3 ubiquitin ligase exclusively located in the nucleus, contributes to various biological processes. This ligase interacts with the PCV3 Cap. However, its role in PCV3 replication remains unclear. This study confirmed that the nuclear localization signal domain of the Cap and the RNF2 N-terminal RING domain facilitate the interaction between the Cap and RNF2. Furthermore, RNF2 promoted the binding of K48-linked polyubiquitination chains to lysine at positions 139 and 140 (K139 and K140) of the PCV3 Cap, thereby degrading the Cap. RNF2 knockdown and overexpression increased or decreased PCV3 replication, respectively. Moreover, the RING domain-deleted RNF2 mutant eliminated the RNF2-induced degradation of the PCV3 Cap and RNF2-mediated inhibition of viral replication. This indicates that both processes were associated with its E3 ligase activity. Our findings demonstrate that RNF2 can interact with and degrade the PCV3 Cap via its N-terminal RING domain in a ubiquitination-dependent manner, thereby inhibiting PCV3 replication.IMPORTANCEPorcine circovirus type 3 is a recently described pathogen that is prevalent worldwide, causing substantial economic losses to the swine industry. However, the mechanisms through which host proteins regulate its replication remain unclear. Here, we demonstrate that ring finger protein 2 inhibits porcine circovirus type 3 replication by interacting with and degrading the Cap of this pathogen in a ubiquitination-dependent manner, requiring its N-terminal RING domain. Ring finger protein 2-mediated degradation of the Cap relies on its E3 ligase activity and the simultaneous existence of K139 and K140 within the Cap. These findings reveal the mechanism by which this protein interacts with and degrades the Cap to inhibit porcine circovirus type 3 replication. This consequently provides novel insights into porcine circovirus type 3 pathogenesis and facilitates the development of preventative measures against this pathogen.</p>","PeriodicalId":17583,"journal":{"name":"Journal of Virology","volume":null,"pages":null},"PeriodicalIF":4.0,"publicationDate":"2024-08-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11334428/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141751984","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Molecular insight into the neutralization mechanism of human-origin monoclonal antibody AH100 against Hantaan virus. 人源单克隆抗体 AH100 对抗汉坦病毒中和机制的分子研究。
IF 4 2区 医学 Q2 VIROLOGY Pub Date : 2024-08-20 Epub Date: 2024-07-30 DOI: 10.1128/jvi.00883-24
Feiran Wang, Tiezhu Liu, Liying Liao, Yan Chai, Jianxun Qi, Feng Gao, Mifang Liang, George Fu Gao, Yan Wu

Both Old World and New World hantaviruses are transmitted through rodents and can lead to hemorrhagic fever with renal syndrome or hantavirus cardiopulmonary syndrome in humans without the availability of specific therapeutics. The square-shaped surface spikes of hantaviruses consist of four Gn-Gc heterodimers that are pivotal for viral entry into host cells and serve as targets for the immune system. Previously, a human-derived neutralizing monoclonal antibody, AH100, demonstrated specific neutralization against the Old World hantavirus, Hantaan virus. However, the precise mode binding of this neutralizing monoclonal antibody remains unclear. In the present study, we determined the structure of the Hantaan virus Gn-AH100 antigen-binding fragment complex and identified its epitope. Crystallography revealed that AH100 targeted the epitopes on domain A and b-ribbon and E3-like domain. Epitope mapping onto a model of the higher order (Gn-Gc)4 spike revealed its localization between neighboring Gn protomers, distinguishing this epitope as a unique site compared to the previously reported monoclonal antibodies. This study provides crucial insights into the structural basis of hantavirus neutralizing antibody epitopes, thereby facilitating the development of therapeutic antibodies.IMPORTANCEHantaan virus (HTNV) poses a significant threat to humans by causing hemorrhagic fever with renal syndrome with high mortality rates. In the absence of FDA-approved drugs or vaccines, it is urgent to develop specific therapeutics. Here, we elucidated the epitope of a human-derived neutralizing antibody, AH100, by determining the HTNV glycoprotein Gn-AH100 antigen-binding fragment (Fab) complex structure. Our findings revealed that the epitopes situated on the domain A and b-ribbon and E3-like domain of the HTNV Gn head. By modeling the complex structure in the viral lattice, we propose that AH100 neutralizes the virus by impeding conformational changes of Gn protomer, which is crucial for viral entry. Additionally, sequence analysis of all reported natural isolates indicated the absence of mutations in epitope residues, suggesting the potential neutralization ability of AH100 in diverse isolates. Therefore, our results provide novel insights into the epitope and the molecular basis of AH100 neutralization.

旧世界汉坦病毒和新世界汉坦病毒都通过啮齿动物传播,在没有特效疗法的情况下,可导致人类出血热并发肾综合征或汉坦病毒心肺综合征。汉坦病毒的方形表面尖峰由四个 Gn-Gc 异二聚体组成,它们是病毒进入宿主细胞的关键,也是免疫系统的靶标。此前,一种人源中和单克隆抗体 AH100 对旧世界汉坦病毒有特异性中和作用。然而,这种中和单克隆抗体的精确结合模式仍不清楚。在本研究中,我们测定了汉坦病毒 Gn-AH100 抗原结合片段复合物的结构,并确定了其表位。结晶学显示,AH100靶向的表位位于结构域A和b-带状结构域以及E3-样结构域。表位在高阶(Gn-Gc)4尖峰模型上的映射显示,其定位在相邻的Gn原体之间,与之前报道的单克隆抗体相比,该表位是一个独特的位点。这项研究为了解汉坦病毒中和抗体表位的结构基础提供了重要依据,从而促进了治疗性抗体的开发。 重要意义汉坦病毒(HTNV)会引起高死亡率的出血热并伴有肾综合征,对人类构成严重威胁。由于缺乏美国食品及药物管理局批准的药物或疫苗,开发特异性疗法迫在眉睫。在此,我们通过确定 HTNV 糖蛋白 Gn-AH100 抗原结合片段(Fab)复合物结构,阐明了人源中和抗体 AH100 的表位。我们的研究结果表明,表位位于 HTNV Gn 头部的 A、b-带状结构域和 E3 样结构域上。通过模拟病毒晶格中的复合物结构,我们提出 AH100 是通过阻碍 Gn 原体的构象变化来中和病毒的,而构象变化是病毒进入的关键。此外,对所有报道的天然分离物进行的序列分析表明,表位残基没有发生突变,这表明 AH100 在不同分离物中具有潜在的中和能力。因此,我们的研究结果为表位和 AH100 中和的分子基础提供了新的见解。
{"title":"Molecular insight into the neutralization mechanism of human-origin monoclonal antibody AH100 against Hantaan virus.","authors":"Feiran Wang, Tiezhu Liu, Liying Liao, Yan Chai, Jianxun Qi, Feng Gao, Mifang Liang, George Fu Gao, Yan Wu","doi":"10.1128/jvi.00883-24","DOIUrl":"10.1128/jvi.00883-24","url":null,"abstract":"<p><p>Both Old World and New World hantaviruses are transmitted through rodents and can lead to hemorrhagic fever with renal syndrome or hantavirus cardiopulmonary syndrome in humans without the availability of specific therapeutics. The square-shaped surface spikes of hantaviruses consist of four Gn-Gc heterodimers that are pivotal for viral entry into host cells and serve as targets for the immune system. Previously, a human-derived neutralizing monoclonal antibody, AH100, demonstrated specific neutralization against the Old World hantavirus, Hantaan virus. However, the precise mode binding of this neutralizing monoclonal antibody remains unclear. In the present study, we determined the structure of the Hantaan virus Gn-AH100 antigen-binding fragment complex and identified its epitope. Crystallography revealed that AH100 targeted the epitopes on domain A and b-ribbon and E3-like domain. Epitope mapping onto a model of the higher order (Gn-Gc)<sub>4</sub> spike revealed its localization between neighboring Gn protomers, distinguishing this epitope as a unique site compared to the previously reported monoclonal antibodies. This study provides crucial insights into the structural basis of hantavirus neutralizing antibody epitopes, thereby facilitating the development of therapeutic antibodies.IMPORTANCEHantaan virus (HTNV) poses a significant threat to humans by causing hemorrhagic fever with renal syndrome with high mortality rates. In the absence of FDA-approved drugs or vaccines, it is urgent to develop specific therapeutics. Here, we elucidated the epitope of a human-derived neutralizing antibody, AH100, by determining the HTNV glycoprotein Gn-AH100 antigen-binding fragment (Fab) complex structure. Our findings revealed that the epitopes situated on the domain A and b-ribbon and E3-like domain of the HTNV Gn head. By modeling the complex structure in the viral lattice, we propose that AH100 neutralizes the virus by impeding conformational changes of Gn protomer, which is crucial for viral entry. Additionally, sequence analysis of all reported natural isolates indicated the absence of mutations in epitope residues, suggesting the potential neutralization ability of AH100 in diverse isolates. Therefore, our results provide novel insights into the epitope and the molecular basis of AH100 neutralization.</p>","PeriodicalId":17583,"journal":{"name":"Journal of Virology","volume":null,"pages":null},"PeriodicalIF":4.0,"publicationDate":"2024-08-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11334459/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141792831","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","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