首页 > 最新文献

Journal of Virology最新文献

英文 中文
Electron tomography visualization of HIV-1 virions trapped by fusion inhibitors to host cells in infected tissues. 通过电子断层扫描观察受感染组织中被宿主细胞融合抑制剂困住的 HIV-1 病毒。
IF 4 2区 医学 Q2 VIROLOGY Pub Date : 2024-11-19 Epub Date: 2024-10-30 DOI: 10.1128/jvi.01432-24
Mark S Ladinsky, Li Zhu, Irfan Ullah, Pradeep D Uchil, Priti Kumar, Michael S Kay, Pamela J Bjorkman

HIV-1 delivers its genetic material to infect a cell after fusion of the viral and host cell membranes, which takes place after the viral envelope (Env) binds host receptor and co-receptor proteins. Binding of host receptor CD4 to Env results in conformational changes that allow interaction with a host co-receptor (CCR5 or CXCR4). Further conformational rearrangements result in an elongated pre-hairpin intermediate structure in which Env is anchored to the viral membrane by its transmembrane region and to the host cell membrane by its fusion peptide. Although budding virions can be readily imaged by electron tomography (ET) of HIV-1-infected tissues and cultured cells, virions that are fusing (attached to host cells via pre-hairpin intermediates) are not normally visualized, perhaps because the process of membrane fusion is too fast to capture by ET. To image virions during fusion, we used fusion inhibitors to prevent downstream conformational changes in Env that lead to membrane fusion, thereby trapping HIV-1 virions linked to target cells by pre-hairpin intermediates. ET of HIV-1 pseudovirions bound to CD4+/CCR5+ TZM-bl cells revealed presumptive pre-hairpin intermediates as 2-4 narrow spokes linking a virion to the cell surface. To extend these results to a more physiological setting, we used ET to image tissues and organs derived from humanized bone marrow/liver/thymus mice infected with HIV-1 and then treated with CPT31, a high-affinity D-peptide fusion inhibitor linked to cholesterol. Trapped HIV-1 virions were found in all tissues studied (small intestine, mesenteric lymph nodes, spleen, and bone marrow), and spokes representing pre-hairpin intermediates linking trapped virions to cell surfaces were similar in structure and number to those seen in the previous pseudovirus and cultured cell ET study.IMPORTANCETrapped and untrapped HIV-1 virions, both mature and immature, were distinguished by localizing spokes via 3D tomographic reconstructions of HIV-1 infected and fusion-inhibitor-treated tissues of humanized mice. The findings of trapped HIV-1 virions in all tissues examined demonstrate a wide distribution of the CPT31 inhibitor, a desirable property for a potential therapeutic. In addition, the presence of virions trapped by spokes, particularly in vascular endothelial cells, demonstrates that the fusion inhibitors can be used as markers for potential HIV-1-target cells within tissues, facilitating the mapping of HIV-1 target cells within the complex cellular milieu of infected tissues.

病毒包膜(Env)与宿主受体蛋白和共受体蛋白结合后,病毒与宿主细胞膜融合,HIV-1 将其遗传物质输送到细胞中进行感染。宿主受体 CD4 与 Env 结合后会发生构象变化,从而与宿主共受体(CCR5 或 CXCR4)相互作用。进一步的构象重排会产生一种拉长的前发夹中间结构,在这种结构中,Env通过其跨膜区固定在病毒膜上,并通过其融合肽固定在宿主细胞膜上。虽然通过对感染 HIV-1 的组织和培养细胞进行电子断层扫描(ET)可以很容易地观察到出芽的病毒,但通常无法观察到正在融合(通过前发夹中间体连接到宿主细胞)的病毒,这可能是因为膜融合过程太快,ET 无法捕捉。为了对融合过程中的病毒进行成像,我们使用了融合抑制剂来阻止 Env 中导致膜融合的下游构象变化,从而捕获通过前发夹中间体连接到靶细胞的 HIV-1 病毒。与 CD4+/CCR5+ TZM-bl 细胞结合的 HIV-1 假病毒的 ET 显示,假定的前发夹中间体是连接病毒与细胞表面的 2-4 条狭窄辐条。为了将这些结果扩展到更生理学的环境中,我们使用 ET 对来自人源化骨髓/肝脏/胸腺小鼠的组织和器官进行成像,这些小鼠感染了 HIV-1,然后用 CPT31(一种与胆固醇相连的高亲和力 D 肽融合抑制剂)处理。在研究的所有组织(小肠、肠系膜淋巴结、脾脏和骨髓)中都发现了被捕获的 HIV-1 病毒,代表连接被捕获病毒与细胞表面的前发夹中间体的辐条在结构和数量上与之前的假病毒和培养细胞 ET 研究中看到的相似。重要意义通过对人源化小鼠感染 HIV-1 病毒的组织和融合抑制剂处理过的组织进行三维断层扫描重建,定位辐条,从而区分成熟和未成熟的被困和未被困 HIV-1 病毒。在所有受检组织中都发现了被困的 HIV-1 病毒,这表明 CPT31 抑制剂的分布范围很广,而这正是潜在疗法的理想特性。此外,辐条捕获的病毒的存在,尤其是在血管内皮细胞中的存在,表明融合抑制剂可用作组织内潜在 HIV-1 靶细胞的标记物,有助于绘制受感染组织复杂细胞环境中的 HIV-1 靶细胞图。
{"title":"Electron tomography visualization of HIV-1 virions trapped by fusion inhibitors to host cells in infected tissues.","authors":"Mark S Ladinsky, Li Zhu, Irfan Ullah, Pradeep D Uchil, Priti Kumar, Michael S Kay, Pamela J Bjorkman","doi":"10.1128/jvi.01432-24","DOIUrl":"10.1128/jvi.01432-24","url":null,"abstract":"<p><p>HIV-1 delivers its genetic material to infect a cell after fusion of the viral and host cell membranes, which takes place after the viral envelope (Env) binds host receptor and co-receptor proteins. Binding of host receptor CD4 to Env results in conformational changes that allow interaction with a host co-receptor (CCR5 or CXCR4). Further conformational rearrangements result in an elongated pre-hairpin intermediate structure in which Env is anchored to the viral membrane by its transmembrane region and to the host cell membrane by its fusion peptide. Although budding virions can be readily imaged by electron tomography (ET) of HIV-1-infected tissues and cultured cells, virions that are fusing (attached to host cells via pre-hairpin intermediates) are not normally visualized, perhaps because the process of membrane fusion is too fast to capture by ET. To image virions during fusion, we used fusion inhibitors to prevent downstream conformational changes in Env that lead to membrane fusion, thereby trapping HIV-1 virions linked to target cells by pre-hairpin intermediates. ET of HIV-1 pseudovirions bound to CD4<sup>+</sup>/CCR5<sup>+</sup> TZM-bl cells revealed presumptive pre-hairpin intermediates as 2-4 narrow spokes linking a virion to the cell surface. To extend these results to a more physiological setting, we used ET to image tissues and organs derived from humanized bone marrow/liver/thymus mice infected with HIV-1 and then treated with CPT31, a high-affinity D-peptide fusion inhibitor linked to cholesterol. Trapped HIV-1 virions were found in all tissues studied (small intestine, mesenteric lymph nodes, spleen, and bone marrow), and spokes representing pre-hairpin intermediates linking trapped virions to cell surfaces were similar in structure and number to those seen in the previous pseudovirus and cultured cell ET study.IMPORTANCETrapped and untrapped HIV-1 virions, both mature and immature, were distinguished by localizing spokes via 3D tomographic reconstructions of HIV-1 infected and fusion-inhibitor-treated tissues of humanized mice. The findings of trapped HIV-1 virions in all tissues examined demonstrate a wide distribution of the CPT31 inhibitor, a desirable property for a potential therapeutic. In addition, the presence of virions trapped by spokes, particularly in vascular endothelial cells, demonstrates that the fusion inhibitors can be used as markers for potential HIV-1-target cells within tissues, facilitating the mapping of HIV-1 target cells within the complex cellular milieu of infected tissues.</p>","PeriodicalId":17583,"journal":{"name":"Journal of Virology","volume":" ","pages":"e0143224"},"PeriodicalIF":4.0,"publicationDate":"2024-11-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11575291/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142546215","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
SARS-CoV-2 nsp13 suppresses hepatitis B virus replication by targeting cccDNA transcription. SARS-CoV-2 nsp13通过靶向cccDNA转录抑制乙型肝炎病毒复制。
IF 4 2区 医学 Q2 VIROLOGY Pub Date : 2024-11-19 Epub Date: 2024-10-07 DOI: 10.1128/jvi.01042-24
Aixin Li, Kaitao Zhao, Yurong Duan, Bei Zhang, Yingcheng Zheng, Chengliang Zhu, Qiongrong Chen, Wen-Bo Liu, Lixia Hui, Yuchen Xia, Xiaoming Cheng

SARS-CoV-2 nonstructural protein 13 (nsp13) has been shown to selectively suppress the transcription of episomal DNA while sparing chromosomal DNA. Hepatitis B Virus (HBV) harbors covalently closed circular DNA (cccDNA), a form of viral episomal DNA found within infected hepatocyte nuclei. The persistence of cccDNA is the major cause of chronic HBV infection. In this study, we investigated the impact of SARS-CoV-2 nsp13 on HBV replication, particularly in the context of cccDNA. Our findings demonstrate that nsp13 effectively hinders HBV replication by suppressing the transcription of HBV cccDNA, both in vitro and in vivo. Additionally, we observed that SARS-CoV-2 nsp13 binds to HBV cccDNA and its NTPase and helicase activities contribute significantly to inhibiting HBV replication. Furthermore, our screening identified the interaction between nsp13 and structural maintenance of chromosomes 4, opening new avenues for future mechanistic inquiries. This study presents the evidence suggesting the potential utilization of SARS-CoV-2 nsp13 as a strategy to impede HBV replication by specifically targeting cccDNA. These findings provide a proof of concept for exploring nsp13 as a prospective approach in combating HBV infection.

Importance: To effectively combat hepatitis B virus (HBV), it is imperative to develop potent antiviral medications targeting covalently closed circular DNA (cccDNA). Our investigation aimed to assess the impact of SARS-CoV-2 nsp13 on HBV replication across diverse HBV models, confirming its ability to significantly reduce several HBV replication markers. Additionally, our identification of the interaction between nsp13 and SMC4 opens the door for further mechanistic exploration. This marks a paradigm shift in our approach to HBV antiviral therapy, introducing an entirely novel perspective. Our findings propose a novel strategy for developing anti-HBV drugs that specifically target HBV cccDNA.

研究表明,SARS-CoV-2 非结构蛋白 13(nsp13)可选择性地抑制外显子 DNA 的转录,同时保护染色体 DNA。乙型肝炎病毒(HBV)携带共价闭合环状 DNA(cccDNA),这是一种病毒表观 DNA,存在于受感染的肝细胞核内。cccDNA的持续存在是HBV慢性感染的主要原因。在这项研究中,我们研究了 SARS-CoV-2 nsp13 对 HBV 复制的影响,特别是在 cccDNA 的情况下。我们的研究结果表明,无论在体外还是体内,nsp13 都能通过抑制 HBV cccDNA 的转录有效阻止 HBV 复制。此外,我们还观察到 SARS-CoV-2 nsp13 与 HBV cccDNA 结合,其 NTPase 和螺旋酶活性对抑制 HBV 复制有显著作用。此外,我们的筛选还发现了 nsp13 与 4 号染色体结构维护之间的相互作用,为今后的机理研究开辟了新的途径。本研究提供的证据表明,SARS-CoV-2 nsp13 可作为一种策略,通过特异性靶向 cccDNA 来抑制 HBV 复制。这些发现为探索 nsp13 作为抗击 HBV 感染的前瞻性方法提供了概念证明:为有效抗击乙型肝炎病毒(HBV),开发针对共价闭合环状 DNA(cccDNA)的强效抗病毒药物势在必行。我们的研究旨在评估SARS-CoV-2 nsp13在各种HBV模型中对HBV复制的影响,证实它能显著减少几种HBV复制标记物。此外,我们对 nsp13 和 SMC4 之间相互作用的鉴定为进一步的机理探索打开了大门。这标志着我们的 HBV 抗病毒治疗方法发生了范式转变,引入了一个全新的视角。我们的发现为开发特异性靶向 HBV cccDNA 的抗 HBV 药物提出了一种新策略。
{"title":"SARS-CoV-2 nsp13 suppresses hepatitis B virus replication by targeting cccDNA transcription.","authors":"Aixin Li, Kaitao Zhao, Yurong Duan, Bei Zhang, Yingcheng Zheng, Chengliang Zhu, Qiongrong Chen, Wen-Bo Liu, Lixia Hui, Yuchen Xia, Xiaoming Cheng","doi":"10.1128/jvi.01042-24","DOIUrl":"10.1128/jvi.01042-24","url":null,"abstract":"<p><p>SARS-CoV-2 nonstructural protein 13 (nsp13) has been shown to selectively suppress the transcription of episomal DNA while sparing chromosomal DNA. Hepatitis B Virus (HBV) harbors covalently closed circular DNA (cccDNA), a form of viral episomal DNA found within infected hepatocyte nuclei. The persistence of cccDNA is the major cause of chronic HBV infection. In this study, we investigated the impact of SARS-CoV-2 nsp13 on HBV replication, particularly in the context of cccDNA. Our findings demonstrate that nsp13 effectively hinders HBV replication by suppressing the transcription of HBV cccDNA, both <i>in vitro</i> and <i>in vivo</i>. Additionally, we observed that SARS-CoV-2 nsp13 binds to HBV cccDNA and its NTPase and helicase activities contribute significantly to inhibiting HBV replication. Furthermore, our screening identified the interaction between nsp13 and structural maintenance of chromosomes 4, opening new avenues for future mechanistic inquiries. This study presents the evidence suggesting the potential utilization of SARS-CoV-2 nsp13 as a strategy to impede HBV replication by specifically targeting cccDNA. These findings provide a proof of concept for exploring nsp13 as a prospective approach in combating HBV infection.</p><p><strong>Importance: </strong>To effectively combat hepatitis B virus (HBV), it is imperative to develop potent antiviral medications targeting covalently closed circular DNA (cccDNA). Our investigation aimed to assess the impact of SARS-CoV-2 nsp13 on HBV replication across diverse HBV models, confirming its ability to significantly reduce several HBV replication markers. Additionally, our identification of the interaction between nsp13 and SMC4 opens the door for further mechanistic exploration. This marks a paradigm shift in our approach to HBV antiviral therapy, introducing an entirely novel perspective. Our findings propose a novel strategy for developing anti-HBV drugs that specifically target HBV cccDNA.</p>","PeriodicalId":17583,"journal":{"name":"Journal of Virology","volume":" ","pages":"e0104224"},"PeriodicalIF":4.0,"publicationDate":"2024-11-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11575256/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142381184","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
DCLK1 mediated cooperative acceleration of EMT by avian leukosis virus subgroup J and Marek's disease virus via the Wnt/β-catenin pathway promotes tumor metastasis. DCLK1通过Wnt/β-catenin途径介导禽白血病病毒J亚群和马立克氏病病毒协同加速EMT,促进肿瘤转移。
IF 4 2区 医学 Q2 VIROLOGY Pub Date : 2024-11-19 Epub Date: 2024-10-24 DOI: 10.1128/jvi.01112-24
Jing Zhou, Defang Zhou, Qian Zhang, Xinyue Zhang, Xiaoyang Liu, Longying Ding, Jing Wen, Xiaoyu Xu, Ziqiang Cheng

Co-infection with oncogenic retrovirus and herpesvirus significantly facilitates tumor metastasis in human and animals. Co-infection with avian leukosis virus subgroup J (ALV-J) and Marek's disease virus (MDV), which are typical oncogenic retrovirus and herpesvirus, respectively, leads to enhanced oncogenicity and accelerated tumor formation, resulting in increased mortality of affected chickens. Previously, we found that ALV-J and MDV cooperatively promoted tumor metastasis. However, the molecular mechanism remains elusive. Here, we found that doublecortin-like kinase 1 (DCLK1) mediated cooperative acceleration of epithelial-mesenchymal transition (EMT) by ALV-J and MDV promoted tumor metastasis. Mechanistically, DCLK1 induced EMT via activating Wnt/β-catenin pathway by interacting with β-catenin, thereby cooperatively promoting tumor metastasis. Initially, we screened and found that DCLK1 was a potential mediator for the cooperative activation of EMT by ALV-J and MDV, and enhanced cell proliferation, migration, and invasion. Subsequently, we revealed that DCLK1 physically interacted with β-catenin to promote the formation of the β-catenin-TCF4 complex, inducing transcription of the Wnt target gene, c-Myc, promoting EMT by increasing the expression of N-cadherin, Vimentin, and Snail, and decreasing the expression of E-cadherin. Taken together, we discovered that jointly activated DCLK1 by ALV-J and MDV accelerated cell proliferation, migration and invasion, and ultimately activated EMT, paving the way for tumor metastasis. This study elucidated the molecular mechanism underlying cooperative metastasis induced by co-infection with retrovirus and herpesvirus.

Importance: Tumor metastasis, a complex phenomenon in which tumor cells spread to new organs, is one of the greatest challenges in cancer research and is the leading cause of cancer-induced death. Numerous studies have shown that oncoviruses and their encoded proteins significantly affect metastasis, especially the EMT process. ALV-J and MDV are classic tumorigenic retrovirus and herpesvirus, respectively. We found that ALV-J and MDV synergistically promoted EMT. Further, we identified the tumor stem cell marker DCLK1 in ALV-J and MDV co-infected cells. DCLK1 directly interacted with β-catenin, promoting the formation of the β-catenin-TCF4 complex. This interaction activated the Wnt/β-catenin pathway, thereby inducing EMT and paving the way for synergistic tumor metastasis. Exploring the molecular mechanisms by which ALV-J and MDV cooperate during EMT will contribute to our understanding of tumor progression and metastasis. This study provides new insights into the cooperative induced tumor metastasis by retroviruses and herpesviruses.

致癌逆转录病毒和疱疹病毒共同感染会显著促进人类和动物的肿瘤转移。禽白血病病毒 J 亚群(ALV-J)和马立克氏病病毒(MDV)分别是典型的致癌逆转录病毒和疱疹病毒,它们共同感染会导致致癌能力增强和肿瘤加速形成,从而增加患鸡的死亡率。此前,我们发现 ALV-J 和 MDV 能协同促进肿瘤转移。然而,其分子机制仍不明确。在这里,我们发现双皮质素样激酶 1(DCLK1)介导 ALV-J 和 MDV 合作加速上皮-间质转化(EMT),促进肿瘤转移。从机理上讲,DCLK1通过与β-catenin相互作用激活Wnt/β-catenin通路诱导EMT,从而协同促进肿瘤转移。最初,我们筛选发现DCLK1是ALV-J和MDV协同激活EMT的潜在介质,并能增强细胞增殖、迁移和侵袭。随后,我们发现DCLK1与β-catenin发生物理作用,促进β-catenin-TCF4复合物的形成,诱导Wnt靶基因c-Myc的转录,通过增加N-cadherin、Vimentin和Snail的表达以及降低E-cadherin的表达来促进EMT。综上所述,我们发现ALV-J和MDV共同激活的DCLK1可加速细胞增殖、迁移和侵袭,并最终激活EMT,为肿瘤转移铺平道路。该研究阐明了逆转录病毒和疱疹病毒联合感染诱导协同转移的分子机制:肿瘤转移是肿瘤细胞扩散到新器官的一种复杂现象,是癌症研究中最大的挑战之一,也是癌症导致死亡的主要原因。大量研究表明,肿瘤病毒及其编码的蛋白对转移,尤其是EMT过程有显著影响。ALV-J 和 MDV 分别是典型的致瘤逆转录病毒和疱疹病毒。我们发现,ALV-J 和 MDV 能协同促进 EMT。此外,我们还在 ALV-J 和 MDV 共同感染的细胞中发现了肿瘤干细胞标记物 DCLK1。DCLK1直接与β-catenin相互作用,促进了β-catenin-TCF4复合物的形成。这种相互作用激活了 Wnt/β-catenin 通路,从而诱导了 EMT,为肿瘤的协同转移铺平了道路。探索ALV-J和MDV在EMT过程中合作的分子机制将有助于我们了解肿瘤的进展和转移。本研究为逆转录病毒和疱疹病毒协同诱导肿瘤转移提供了新的见解。
{"title":"DCLK1 mediated cooperative acceleration of EMT by avian leukosis virus subgroup J and Marek's disease virus via the Wnt/β-catenin pathway promotes tumor metastasis.","authors":"Jing Zhou, Defang Zhou, Qian Zhang, Xinyue Zhang, Xiaoyang Liu, Longying Ding, Jing Wen, Xiaoyu Xu, Ziqiang Cheng","doi":"10.1128/jvi.01112-24","DOIUrl":"10.1128/jvi.01112-24","url":null,"abstract":"<p><p>Co-infection with oncogenic retrovirus and herpesvirus significantly facilitates tumor metastasis in human and animals. Co-infection with avian leukosis virus subgroup J (ALV-J) and Marek's disease virus (MDV), which are typical oncogenic retrovirus and herpesvirus, respectively, leads to enhanced oncogenicity and accelerated tumor formation, resulting in increased mortality of affected chickens. Previously, we found that ALV-J and MDV cooperatively promoted tumor metastasis. However, the molecular mechanism remains elusive. Here, we found that doublecortin-like kinase 1 (DCLK1) mediated cooperative acceleration of epithelial-mesenchymal transition (EMT) by ALV-J and MDV promoted tumor metastasis. Mechanistically, DCLK1 induced EMT via activating Wnt/β-catenin pathway by interacting with β-catenin, thereby cooperatively promoting tumor metastasis. Initially, we screened and found that DCLK1 was a potential mediator for the cooperative activation of EMT by ALV-J and MDV, and enhanced cell proliferation, migration, and invasion. Subsequently, we revealed that DCLK1 physically interacted with β-catenin to promote the formation of the β-catenin-TCF4 complex, inducing transcription of the Wnt target gene, c-Myc, promoting EMT by increasing the expression of N-cadherin, Vimentin, and Snail, and decreasing the expression of E-cadherin. Taken together, we discovered that jointly activated DCLK1 by ALV-J and MDV accelerated cell proliferation, migration and invasion, and ultimately activated EMT, paving the way for tumor metastasis. This study elucidated the molecular mechanism underlying cooperative metastasis induced by co-infection with retrovirus and herpesvirus.</p><p><strong>Importance: </strong>Tumor metastasis, a complex phenomenon in which tumor cells spread to new organs, is one of the greatest challenges in cancer research and is the leading cause of cancer-induced death. Numerous studies have shown that oncoviruses and their encoded proteins significantly affect metastasis, especially the EMT process. ALV-J and MDV are classic tumorigenic retrovirus and herpesvirus, respectively. We found that ALV-J and MDV synergistically promoted EMT. Further, we identified the tumor stem cell marker DCLK1 in ALV-J and MDV co-infected cells. DCLK1 directly interacted with β-catenin, promoting the formation of the β-catenin-TCF4 complex. This interaction activated the Wnt/β-catenin pathway, thereby inducing EMT and paving the way for synergistic tumor metastasis. Exploring the molecular mechanisms by which ALV-J and MDV cooperate during EMT will contribute to our understanding of tumor progression and metastasis. This study provides new insights into the cooperative induced tumor metastasis by retroviruses and herpesviruses.</p>","PeriodicalId":17583,"journal":{"name":"Journal of Virology","volume":" ","pages":"e0111224"},"PeriodicalIF":4.0,"publicationDate":"2024-11-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11575233/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142503001","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
Conserved residues of the immunosuppressive domain of MLV are essential for regulating the fusion-critical SU-TM disulfide bond. MLV 免疫抑制结构域的保守残基对于调节融合关键的 SU-TM 二硫键至关重要。
IF 4 2区 医学 Q2 VIROLOGY Pub Date : 2024-11-19 Epub Date: 2024-10-29 DOI: 10.1128/jvi.00989-24
Victoria A Hogan, Julia Harmon, Miguel Cid-Rosas, Laura R Hall, Welkin E Johnson

The Env protein of murine leukemia virus (MLV) is the prototype of a large clade of retroviral fusogens, collectively known as gamma-type Envs. Gamma-type Envs are found in retroviruses and endogenous retroviruses (ERVs) representing a broad range of vertebrate hosts. All gamma-type Envs contain a highly conserved stretch of 26-residues in the transmembrane subunit (TM) comprising two motifs, a putative immunosuppressive domain (ISD) and a CX6CC motif. Extraordinary conservation of the ISD and its invariant association with the CX6CC suggests a fundamental contribution to Env function. To investigate ISD function, we characterized several mutants with single amino acid substitutions at conserved positions in the MLV ISD. A majority abolished infectivity, although we did not observe a corresponding loss in intrinsic ability to mediate membrane fusion. Ratios of the surface subunit (SU) to capsid protein (CA) in virions were diminished for a majority of the ISD mutants, while TM:CA ratios were similar to wild type. Specific loss of SU reflected premature isomerization of the labile disulfide bond that links SU and TM prior to fusion. Indeed, all non-infectious mutants displayed significantly lower disulfide stability than wild-type Env. These results reveal a role for ISD positions 2, 3, 4, 7, and 10 in regulating a late step in entry after fusion peptide insertion but prior to creation of the fusion pore. This implies that the ISD is part of a larger domain, comprising the ISD and CX6CC motifs, that is critical for the formation and regulation of the metastable, intersubunit disulfide bond.IMPORTANCEThe gamma-type Env is a prevalent viral fusogen, found within retroviruses and endogenous retroviruses across vertebrate species and in filoviruses such as Ebolavirus. The fusion mechanism of gamma-type Envs is unique from other Class I fusogens such as those of influenza A virus and HIV-1. Gamma-type Envs contain a hallmark feature known as the immunosuppressive domain (ISD) that has been the subject of some controversy in the literature surrounding its putative immunosuppressive effects. Despite the distinctive conservation of the ISD, little has been done to investigate the role of this region for the function of this widespread fusogen. Our work demonstrates the importance of the ISD for the function of gamma-type Envs in infection, particularly in regulating the intermediate steps of membrane fusion. Understanding the fusion mechanism of gamma-type Envs has broad implications for understanding the entry of extant viruses and aspects of host biology connected to co-opted endogenous gamma-type Envs.

小鼠白血病病毒(MLV)的 Env 蛋白是一大类逆转录病毒融合体的原型,统称为伽马型 Envs。γ型 Envs 存在于代表多种脊椎动物宿主的逆转录病毒和内源性逆转录病毒(ERVs)中。所有伽马型 Envs 的跨膜亚基(TM)中都有一段高度保守的 26 个残基的结构,其中包括两个基团,一个是假定的免疫抑制结构域(ISD),另一个是 CX6CC 基团。ISD 的超常保留及其与 CX6CC 的不变关联表明,它对 Env 的功能有根本性的贡献。为了研究 ISD 的功能,我们鉴定了在 MLV ISD 的保守位置进行单氨基酸置换的几个突变体。虽然我们没有观察到介导膜融合的内在能力有相应的丧失,但大多数突变体都丧失了感染性。大多数 ISD 突变体的病毒表面亚基(SU)与囊膜蛋白(CA)的比例降低,而 TM:CA 的比例与野生型相似。SU的特异性损失反映了连接SU和TM的易变二硫键在融合前的过早异构化。事实上,所有非感染突变体的二硫键稳定性都明显低于野生型 Env。这些结果揭示了 ISD 的 2、3、4、7 和 10 号位置在融合肽插入后、融合孔形成前的晚期进入过程中的调控作用。这意味着 ISD 是由 ISD 和 CX6CC 基序组成的更大结构域的一部分,对于形成和调节可转移的亚基间二硫键至关重要。 重要意义γ-型 Env 是一种常见的病毒融合原,存在于脊椎动物中的逆转录病毒和内源性逆转录病毒以及埃博拉病毒等丝状病毒中。伽马型 Envs 的融合机制与其他 I 类融合原(如甲型流感病毒和 HIV-1 病毒)不同。γ-型 Envs 含有一个标志性特征,即免疫抑制结构域(ISD)。尽管 ISD 具有独特的保护性,但人们对这一区域在这种广泛存在的致病菌功能中的作用却鲜有研究。我们的工作证明了 ISD 对γ型 Envs 在感染中的功能的重要性,尤其是在调节膜融合的中间步骤方面。了解γ型Envs的融合机制对于了解现存病毒的进入以及与共生内源性γ型Envs有关的宿主生物学方面具有广泛的意义。
{"title":"Conserved residues of the immunosuppressive domain of MLV are essential for regulating the fusion-critical SU-TM disulfide bond.","authors":"Victoria A Hogan, Julia Harmon, Miguel Cid-Rosas, Laura R Hall, Welkin E Johnson","doi":"10.1128/jvi.00989-24","DOIUrl":"10.1128/jvi.00989-24","url":null,"abstract":"<p><p>The Env protein of murine leukemia virus (MLV) is the prototype of a large clade of retroviral fusogens, collectively known as gamma-type Envs. Gamma-type Envs are found in retroviruses and endogenous retroviruses (ERVs) representing a broad range of vertebrate hosts. All gamma-type Envs contain a highly conserved stretch of 26-residues in the transmembrane subunit (TM) comprising two motifs, a putative immunosuppressive domain (ISD) and a CX<sub>6</sub>CC motif. Extraordinary conservation of the ISD and its invariant association with the CX<sub>6</sub>CC suggests a fundamental contribution to Env function. To investigate ISD function, we characterized several mutants with single amino acid substitutions at conserved positions in the MLV ISD. A majority abolished infectivity, although we did not observe a corresponding loss in intrinsic ability to mediate membrane fusion. Ratios of the surface subunit (SU) to capsid protein (CA) in virions were diminished for a majority of the ISD mutants, while TM:CA ratios were similar to wild type. Specific loss of SU reflected premature isomerization of the labile disulfide bond that links SU and TM prior to fusion. Indeed, all non-infectious mutants displayed significantly lower disulfide stability than wild-type Env. These results reveal a role for ISD positions 2, 3, 4, 7, and 10 in regulating a late step in entry after fusion peptide insertion but prior to creation of the fusion pore. This implies that the ISD is part of a larger domain, comprising the ISD and CX<sub>6</sub>CC motifs, that is critical for the formation and regulation of the metastable, intersubunit disulfide bond.IMPORTANCEThe gamma-type Env is a prevalent viral fusogen, found within retroviruses and endogenous retroviruses across vertebrate species and in filoviruses such as Ebolavirus. The fusion mechanism of gamma-type Envs is unique from other Class I fusogens such as those of influenza A virus and HIV-1. Gamma-type Envs contain a hallmark feature known as the immunosuppressive domain (ISD) that has been the subject of some controversy in the literature surrounding its putative immunosuppressive effects. Despite the distinctive conservation of the ISD, little has been done to investigate the role of this region for the function of this widespread fusogen. Our work demonstrates the importance of the ISD for the function of gamma-type Envs in infection, particularly in regulating the intermediate steps of membrane fusion. Understanding the fusion mechanism of gamma-type Envs has broad implications for understanding the entry of extant viruses and aspects of host biology connected to co-opted endogenous gamma-type Envs.</p>","PeriodicalId":17583,"journal":{"name":"Journal of Virology","volume":" ","pages":"e0098924"},"PeriodicalIF":4.0,"publicationDate":"2024-11-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11575397/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142522155","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
Single-cell transcriptional analysis of murine norovirus infection in a human intestinal cell line. 人肠道细胞系感染鼠诺如病毒的单细胞转录分析。
IF 4 2区 医学 Q2 VIROLOGY Pub Date : 2024-11-19 Epub Date: 2024-10-30 DOI: 10.1128/jvi.01617-24
Yuki Matsushima, Eric A Levenson, Natthawan Chaimongkol, Loyall Harris, Yongmei Zhao, Sevilay Turan, Francisco Otaizo-Carrasquero, Sundar Ganesan, Katherine M Hornick, Craig Martens, Stanislav V Sosnovtsev, Kim Y Green

Noroviruses are a major agent of acute gastroenteritis in humans, but host cell requirements for efficient replication in vitro have not been established. We engineered a human intestinal cell line (designated mCD300lf-hCaco2) expressing the murine norovirus (MNV) receptor, mouse CD300lf to become fully permissive for MNV replication. To explore the replicative machinery and host response of these cells, we performed a single-cell RNA sequencing (scRNA-seq) transcriptomics analysis of an MNV infection over time. Marked similarities were observed between certain global features of MNV infection in human cells compared to those previously reported in mouse cells by whole population transcriptomics such as downregulation of ribosome biogenesis, mitochondrial dysfunction, and cell cycle preference for G1. Our scRNA-seq analysis allowed further resolution of an infected cell population into distinct clusters with varying levels of viral RNA and interferon-stimulated gene ISG15 transcripts. Cells with high viral replication displayed downregulated ribosomal protein small (RPS) and large (RPL) genes and mitochondrial complexes I, III, IV, and V genes during exponential viral propagation. Ferritin subunit genes FTL and FTH1 were also downregulated during active MNV replication, suggesting that inhibition of iron metabolism may increase replication efficiency. Consistent with this, transcriptional activation of these genes with ferric ammonium citrate and overexpression of FTL lowered virus yields. Comparative studies of cells that support varying levels of norovirus replication efficiency, as determined by scRNA-seq may lead to improved human cell-based culture systems and effective viral interventions.IMPORTANCEHuman noroviruses cause acute gastroenteritis in all age groups. Vaccines and antiviral drugs are not yet available, in part, because it is difficult to propagate the viruses causing human disease in standard laboratory cell culture systems. In contrast, a norovirus found in mice [murine norovirus (MNV)] replicates efficiently in murine-based cell culture and has served as a model system. In this study, we established a new human intestinal cell line that was genetically modified to express the murine norovirus receptor so that the human cells became permissive to murine norovirus infection. We then defined the host response to MNV infection in the engineered human cell line at a single-cell resolution and identified cellular genes associated with the highest levels of MNV replication. This study may lead to the improvement of the current human norovirus cell culture systems and help to identify norovirus-host interactions that could be targeted for antiviral drugs.

诺如病毒是人类急性肠胃炎的主要病原体,但宿主细胞在体外有效复制的要求尚未确定。我们设计了一种表达小鼠诺如病毒(MNV)受体--小鼠 CD300lf 的人肠道细胞系(命名为 mCD300lf-hCaco2),使其完全允许 MNV 复制。为了探索这些细胞的复制机制和宿主反应,我们对MNV感染的时间进行了单细胞RNA测序(scRNA-seq)转录组学分析。与之前在小鼠细胞中通过全群转录组学观察到的MNV感染的某些全局特征(如核糖体生物发生的下调、线粒体功能障碍和细胞周期对G1的偏好)相比,在人类细胞中观察到了明显的相似性。我们的 scRNA-seq 分析进一步将受感染的细胞群划分为不同的群组,这些群组的病毒 RNA 和干扰素刺激基因 ISG15 转录本水平各不相同。在病毒的指数传播过程中,病毒复制量大的细胞显示出核糖体蛋白小(RPS)和大(RPL)基因以及线粒体复合物 I、III、IV 和 V 基因的下调。铁蛋白亚基基因 FTL 和 FTH1 也在 MNV 复制活跃期下调,这表明抑制铁代谢可能会提高复制效率。与此相一致的是,柠檬酸铁铵对这些基因的转录激活以及 FTL 的过表达降低了病毒产量。通过 scRNA-seq 对支持不同水平诺如病毒复制效率的细胞进行比较研究,可能会改进基于人体细胞的培养系统和有效的病毒干预措施。目前还没有疫苗和抗病毒药物,部分原因是很难在标准的实验室细胞培养系统中繁殖导致人类疾病的病毒。与此相反,一种在小鼠体内发现的诺如病毒(鼠诺如病毒(MNV))却能在小鼠细胞培养中高效复制,并被用作模型系统。在本研究中,我们建立了一种新的人类肠道细胞系,并对其进行了基因改造,使其表达鼠诺如病毒受体,从而使人类细胞对鼠诺如病毒感染具有容许性。然后,我们以单细胞分辨率定义了宿主对工程人细胞系中 MNV 感染的反应,并确定了与最高水平 MNV 复制相关的细胞基因。这项研究可能会改进目前的人类诺如病毒细胞培养系统,并有助于确定诺如病毒与宿主之间的相互作用,从而成为抗病毒药物的靶标。
{"title":"Single-cell transcriptional analysis of murine norovirus infection in a human intestinal cell line.","authors":"Yuki Matsushima, Eric A Levenson, Natthawan Chaimongkol, Loyall Harris, Yongmei Zhao, Sevilay Turan, Francisco Otaizo-Carrasquero, Sundar Ganesan, Katherine M Hornick, Craig Martens, Stanislav V Sosnovtsev, Kim Y Green","doi":"10.1128/jvi.01617-24","DOIUrl":"10.1128/jvi.01617-24","url":null,"abstract":"<p><p>Noroviruses are a major agent of acute gastroenteritis in humans, but host cell requirements for efficient replication <i>in vitro</i> have not been established. We engineered a human intestinal cell line (designated mCD300lf-hCaco2) expressing the murine norovirus (MNV) receptor, mouse CD300lf to become fully permissive for MNV replication. To explore the replicative machinery and host response of these cells, we performed a single-cell RNA sequencing (scRNA-seq) transcriptomics analysis of an MNV infection over time. Marked similarities were observed between certain global features of MNV infection in human cells compared to those previously reported in mouse cells by whole population transcriptomics such as downregulation of ribosome biogenesis, mitochondrial dysfunction, and cell cycle preference for G1. Our scRNA-seq analysis allowed further resolution of an infected cell population into distinct clusters with varying levels of viral RNA and interferon-stimulated gene ISG15 transcripts. Cells with high viral replication displayed downregulated ribosomal protein small (RPS) and large (RPL) genes and mitochondrial complexes I, III, IV, and V genes during exponential viral propagation. Ferritin subunit genes FTL and FTH1 were also downregulated during active MNV replication, suggesting that inhibition of iron metabolism may increase replication efficiency. Consistent with this, transcriptional activation of these genes with ferric ammonium citrate and overexpression of FTL lowered virus yields. Comparative studies of cells that support varying levels of norovirus replication efficiency, as determined by scRNA-seq may lead to improved human cell-based culture systems and effective viral interventions.IMPORTANCEHuman noroviruses cause acute gastroenteritis in all age groups. Vaccines and antiviral drugs are not yet available, in part, because it is difficult to propagate the viruses causing human disease in standard laboratory cell culture systems. In contrast, a norovirus found in mice [murine norovirus (MNV)] replicates efficiently in murine-based cell culture and has served as a model system. In this study, we established a new human intestinal cell line that was genetically modified to express the murine norovirus receptor so that the human cells became permissive to murine norovirus infection. We then defined the host response to MNV infection in the engineered human cell line at a single-cell resolution and identified cellular genes associated with the highest levels of MNV replication. This study may lead to the improvement of the current human norovirus cell culture systems and help to identify norovirus-host interactions that could be targeted for antiviral drugs.</p>","PeriodicalId":17583,"journal":{"name":"Journal of Virology","volume":" ","pages":"e0161724"},"PeriodicalIF":4.0,"publicationDate":"2024-11-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11575399/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142546226","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
Exosome-mediated viral nucleic acid presentation in a crustacean expounds innate immunity from a novel perspective. 外泌体介导的甲壳动物病毒核酸呈递从一个新的角度揭示了先天性免疫。
IF 4 2区 医学 Q2 VIROLOGY Pub Date : 2024-11-15 DOI: 10.1128/jvi.01519-24
Yi Gong, Hang Hu, Xinshan Zhao, Weiqian Wei, Ming Zhang, Ngoc Tuan Tran, Hongyu Ma, Yueling Zhang, Kok-Gan Chan, Shengkang Li

As an enduring hot topic in the field of innate immunity, apoptosis is widely considered an effective approach to eliminate pathogenic microbes and plays a crucial role during host-pathogen interactions. Recently, researchers have found that the virus-containing host cells could transmit apoptotic signals to the surrounding uninfected cells during infection, but the mechanism remains unclear. Here, we found that exosomes secreted by WSSV-infected mud crab hemocytes contain viral nucleic acid wsv277, which could be transported to the recipient cells and further expressed viral protein with phosphokinase activity. Besides, by using transcriptome, proteome, ChIP-seq, and coIP techniques, the results revealed that wsv277 could activate the transcription and translation of apoptotic genes via interacting with CBF and EF-1α so as to suppress the spread of virus infection by inducing apoptosis of the surrounding cells. Therefore, for the first time, our study proved that the components of DNA virus could be encapsulated into exosomes and elucidated the mechanism of apoptotic signal transduction between cells from the perspective of exosomes.

Importance: Our study revealed that the components of DNA virus could be packaged and transmitted through the exosomes of lower invertebrates, which strongly demonstrated the diversity of exosome-mediated viral immunity and its universality in animals. Furthermore, we elucidated the mechanism of apoptotic signal transduction between cells from the perspective of exosomes and revealed a novel strategy for the host to cope with viral infection.

作为先天性免疫领域一个持久的热门话题,细胞凋亡被广泛认为是消灭病原微生物的有效方法,并在宿主与病原体的相互作用中发挥着至关重要的作用。最近,研究人员发现,在感染过程中,含有病毒的宿主细胞可向周围未感染细胞传递凋亡信号,但其机制仍不清楚。在这里,我们发现受 WSSV 感染的泥蟹血细胞分泌的外泌体含有病毒核酸 wsv277,它可以被转运到受体细胞,并进一步表达具有磷酸激酶活性的病毒蛋白。此外,通过转录组、蛋白质组、ChIP-seq和coIP技术,结果发现wsv277可通过与CBF和EF-1α相互作用激活凋亡基因的转录和翻译,从而通过诱导周围细胞凋亡抑制病毒感染的扩散。因此,我们的研究首次证明了DNA病毒成分可被包裹到外泌体中,并从外泌体的角度阐明了细胞间凋亡信号转导的机制:我们的研究发现,DNA病毒的成分可以通过低等无脊椎动物的外泌体进行包装和传播,这有力地证明了外泌体介导的病毒免疫的多样性及其在动物中的普遍性。此外,我们还从外泌体的角度阐明了细胞间凋亡信号转导的机制,并揭示了宿主应对病毒感染的一种新策略。
{"title":"Exosome-mediated viral nucleic acid presentation in a crustacean expounds innate immunity from a novel perspective.","authors":"Yi Gong, Hang Hu, Xinshan Zhao, Weiqian Wei, Ming Zhang, Ngoc Tuan Tran, Hongyu Ma, Yueling Zhang, Kok-Gan Chan, Shengkang Li","doi":"10.1128/jvi.01519-24","DOIUrl":"https://doi.org/10.1128/jvi.01519-24","url":null,"abstract":"<p><p>As an enduring hot topic in the field of innate immunity, apoptosis is widely considered an effective approach to eliminate pathogenic microbes and plays a crucial role during host-pathogen interactions. Recently, researchers have found that the virus-containing host cells could transmit apoptotic signals to the surrounding uninfected cells during infection, but the mechanism remains unclear. Here, we found that exosomes secreted by WSSV-infected mud crab hemocytes contain viral nucleic acid wsv277, which could be transported to the recipient cells and further expressed viral protein with phosphokinase activity. Besides, by using transcriptome, proteome, ChIP-seq, and coIP techniques, the results revealed that wsv277 could activate the transcription and translation of apoptotic genes via interacting with CBF and EF-1α so as to suppress the spread of virus infection by inducing apoptosis of the surrounding cells. Therefore, for the first time, our study proved that the components of DNA virus could be encapsulated into exosomes and elucidated the mechanism of apoptotic signal transduction between cells from the perspective of exosomes.</p><p><strong>Importance: </strong>Our study revealed that the components of DNA virus could be packaged and transmitted through the exosomes of lower invertebrates, which strongly demonstrated the diversity of exosome-mediated viral immunity and its universality in animals. Furthermore, we elucidated the mechanism of apoptotic signal transduction between cells from the perspective of exosomes and revealed a novel strategy for the host to cope with viral infection.</p>","PeriodicalId":17583,"journal":{"name":"Journal of Virology","volume":" ","pages":"e0151924"},"PeriodicalIF":4.0,"publicationDate":"2024-11-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142639227","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 threshold of a potent neutralizing Zika virus monoclonal antibody in rhesus macaques. 猕猴体内强效中和寨卡病毒单克隆抗体的保护阈值。
IF 4 2区 医学 Q2 VIROLOGY Pub Date : 2024-11-15 DOI: 10.1128/jvi.01429-24
Joseph P Nkolola, David Hope, Ruoran Guan, Alessandro Colarusso, Malika Aid, Deborah Weiss, John Misamore, Hanne Andersen, Mark G Lewis, Lauren Williamson, Robert H Carnahan, James E Crowe, Dan H Barouch

Zika virus (ZIKV) is a mosquito-borne flavivirus that caused a global pandemic in 2016-2017 with continued ongoing transmission at low levels in several countries. In the absence of an approved ZIKV vaccine, neutralizing monoclonal antibodies (mAbs) provide an option for the prevention and treatment of ZIKV infection. Previous studies identified a potent neutralizing human mAb ZIKV-117 that reduced fetal infection and death in mice following ZIKV challenge. In this study, we report exquisite potency of ZIKV-117-LALA-YTE, which has been engineered to reduce Fc receptor binding and to extend half-life, in a titration study in rhesus macaques to protect against ZIKV challenge. We show complete protection at a dose of 0.016 mg/kg ZIKV-117-LALA-YTE, which resulted in median serum concentrations of 0.13 µg/mL. The high potency of this mAb supports its potential clinical development as a novel biotherapeutic intervention for ZIKV.IMPORTANCEIn this study, we report the potency of the Zika virus (ZIKV)-specific neutralizing antibody ZIKV-117-LALA-YTE against ZIKV challenge in a titration study rhesus macaques. This high potency supports the further development of this monoclonal antibody for ZIKV.

寨卡病毒(ZIKV)是一种由蚊子传播的黄病毒,曾在 2016-2017 年引发全球大流行,并在多个国家持续低水平传播。在没有获得批准的寨卡病毒疫苗的情况下,中和单克隆抗体(mAbs)为预防和治疗寨卡病毒感染提供了一种选择。之前的研究发现了一种强效中和性人类 mAb ZIKV-117,它能减少小鼠在受到 ZIKV 挑战后的胎儿感染和死亡。在本研究中,我们报告了 ZIKV-117-LALA-YTE 在恒河猴滴定研究中的卓越功效,该抗体经过设计,可减少 Fc 受体结合并延长半衰期,从而抵御 ZIKV 挑战。我们的研究结果表明,ZIKV-117-LALA-YTE 的剂量为 0.016 mg/kg,血清浓度中值为 0.13 µg/mL,即可提供完全保护。重要意义在这项研究中,我们报告了寨卡病毒(ZIKV)特异性中和抗体 ZIKV-117-LALA-YTE 在猕猴滴定研究中抵御 ZIKV 挑战的效力。这种高效力支持进一步开发这种针对 ZIKV 的单克隆抗体。
{"title":"Protective threshold of a potent neutralizing Zika virus monoclonal antibody in rhesus macaques.","authors":"Joseph P Nkolola, David Hope, Ruoran Guan, Alessandro Colarusso, Malika Aid, Deborah Weiss, John Misamore, Hanne Andersen, Mark G Lewis, Lauren Williamson, Robert H Carnahan, James E Crowe, Dan H Barouch","doi":"10.1128/jvi.01429-24","DOIUrl":"https://doi.org/10.1128/jvi.01429-24","url":null,"abstract":"<p><p>Zika virus (ZIKV) is a mosquito-borne flavivirus that caused a global pandemic in 2016-2017 with continued ongoing transmission at low levels in several countries. In the absence of an approved ZIKV vaccine, neutralizing monoclonal antibodies (mAbs) provide an option for the prevention and treatment of ZIKV infection. Previous studies identified a potent neutralizing human mAb ZIKV-117 that reduced fetal infection and death in mice following ZIKV challenge. In this study, we report exquisite potency of ZIKV-117-LALA-YTE, which has been engineered to reduce Fc receptor binding and to extend half-life, in a titration study in rhesus macaques to protect against ZIKV challenge. We show complete protection at a dose of 0.016 mg/kg ZIKV-117-LALA-YTE, which resulted in median serum concentrations of 0.13 µg/mL. The high potency of this mAb supports its potential clinical development as a novel biotherapeutic intervention for ZIKV.IMPORTANCEIn this study, we report the potency of the Zika virus (ZIKV)-specific neutralizing antibody ZIKV-117-LALA-YTE against ZIKV challenge in a titration study rhesus macaques. This high potency supports the further development of this monoclonal antibody for ZIKV.</p>","PeriodicalId":17583,"journal":{"name":"Journal of Virology","volume":" ","pages":"e0142924"},"PeriodicalIF":4.0,"publicationDate":"2024-11-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142639244","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
Novel virulence determinants in VP1 regulate the assembly of enterovirus-A71. VP1 中的新型毒力决定因子调控肠病毒-A71 的组装。
IF 4 2区 医学 Q2 VIROLOGY Pub Date : 2024-11-13 DOI: 10.1128/jvi.01655-24
Wenjing Zhang, Quanjie Li, Dongrong Yi, Ruifang Zheng, Guihua Liu, Qian Liu, Saisai Guo, Jianyuan Zhao, Jing Wang, Ling Ma, Jiwei Ding, Rui Zhou, Yongcheng Ren, Tingting Sun, Ao Zhang, Xiaoyu Li, Yongxin Zhang, Shan Cen

Enterovirus-A71 (EV-A71) is the second most common causative agent after coxsackievirus A16 of hand, foot, and mouth disease. The capsids of EV-A71 consist of 60 copies of each of the four viral structural proteins (VP1-VP4). VP1 is highly exposed and surface accessible, playing a central role in virus particle assembly, attachment, and entry. To gain insight into the role of highly conserved residues at positions 75, 78, and 88 in the capsid protein VP1 in these processes, an alanine-scanning analysis was performed using an infectious cDNA clone of EV-A71. Our study revealed that the substitutions of VP1-T75A, VP1-T78A, and VP1-G88A could affect the assembly of the virus capsid proteins, resulting in the production of abnormal virions with reduced infectivity. Specifically, the substitution of VP1-T75A affected the maturation cleavage of the VP0 precursor, leading to deficiencies in binding to receptor scavenger receptor class B2 (SCARB2), viral attachment, internalization, and even uncoating. For the mutants of T78A and G88A, a significant reduction in virion-associated genomic RNA was observed, suggesting that more noninfectious empty particles were produced during viral assembly. Interestingly, the VP1-T75A variant showed weak replication in cell cultures but demonstrated increased virulence in BALB/c neonatal mice, which might be due to the difference in viral receptors among mammalian species. Taken together, our data revealed the important role of the highly conserved residues T75, T78, and G88 in VP1 protein in the infectivity of EV-A71. Characterizing these novel determinants of EV-A71 virulence would contribute to rationally developing effective treatments and broadly protective vaccine candidates.

Importance: EV-A71 causes hand, foot, and mouth disease in children. In this study, we discovered three highly conserved residues at positions 75, 78, and 88 of the capsid protein VP1 as the potential virulence determinants of EV-A71, which can influence viral replication by regulating the assembly of EV-A71. Mechanistic studies revealed that VP1-T75A could affect the maturation cleavage of the VP0 precursor, resulting in deficiencies in binding to the receptor SCARB2, viral attachment, internalization, and even uncoating. For the mutants of T78A and G88A, more noninfectious empty particles were produced during viral assembly. The discovery of these novel determinants of EV-A71 virulence will promote the study of the pathogenesis of enteroviruses.

肠道病毒-A71(EV-A71)是仅次于柯萨奇病毒 A16 的第二大手足口病致病病毒。EV-A71 的囊壳由四种病毒结构蛋白(VP1-VP4)各 60 个拷贝组成。VP1 高度暴露于病毒表面,在病毒粒子的组装、附着和进入过程中发挥着核心作用。为了深入了解荚膜蛋白 VP1 第 75、78 和 88 位高度保守残基在这些过程中的作用,我们使用 EV-A71 的感染性 cDNA 克隆进行了丙氨酸扫描分析。我们的研究发现,VP1-T75A、VP1-T78A 和 VP1-G88A 的置换会影响病毒帽蛋白的组装,导致产生感染力降低的异常病毒。具体来说,VP1-T75A 的替代影响了 VP0 前体的成熟裂解,导致与清道夫受体 B2 类(SCARB2)结合、病毒附着、内化甚至解衣壳等方面的缺陷。在 T78A 和 G88A 突变体中,观察到与病毒相关的基因组 RNA 显著减少,这表明在病毒组装过程中产生了更多的非感染性空颗粒。有趣的是,VP1-T75A 变体在细胞培养物中的复制能力较弱,但在 BALB/c 新生小鼠中的毒力却有所增强,这可能是由于哺乳动物物种间病毒受体的差异造成的。总之,我们的数据揭示了 VP1 蛋白中高度保守的残基 T75、T78 和 G88 在 EV-A71 感染性中的重要作用。研究这些决定 EV-A71 致病性的新因素将有助于合理开发有效的治疗方法和具有广泛保护作用的候选疫苗:重要性:EV-A71 会导致儿童手足口病。在这项研究中,我们发现了位于荚膜蛋白质 VP1 第 75、78 和 88 位的三个高度保守残基是 EV-A71 的潜在毒力决定因子,它们可以通过调节 EV-A71 的组装来影响病毒复制。机理研究发现,VP1-T75A可影响VP0前体的成熟裂解,导致与受体SCARB2结合、病毒附着、内化甚至解衣等方面的缺陷。对于 T78A 和 G88A 突变体,在病毒组装过程中会产生更多的非感染性空颗粒。EV-A71 毒力的这些新决定因素的发现将促进对肠道病毒发病机制的研究。
{"title":"Novel virulence determinants in VP1 regulate the assembly of enterovirus-A71.","authors":"Wenjing Zhang, Quanjie Li, Dongrong Yi, Ruifang Zheng, Guihua Liu, Qian Liu, Saisai Guo, Jianyuan Zhao, Jing Wang, Ling Ma, Jiwei Ding, Rui Zhou, Yongcheng Ren, Tingting Sun, Ao Zhang, Xiaoyu Li, Yongxin Zhang, Shan Cen","doi":"10.1128/jvi.01655-24","DOIUrl":"https://doi.org/10.1128/jvi.01655-24","url":null,"abstract":"<p><p>Enterovirus-A71 (EV-A71) is the second most common causative agent after coxsackievirus A16 of hand, foot, and mouth disease. The capsids of EV-A71 consist of 60 copies of each of the four viral structural proteins (VP1-VP4). VP1 is highly exposed and surface accessible, playing a central role in virus particle assembly, attachment, and entry. To gain insight into the role of highly conserved residues at positions 75, 78, and 88 in the capsid protein VP1 in these processes, an alanine-scanning analysis was performed using an infectious cDNA clone of EV-A71. Our study revealed that the substitutions of VP1-T75A, VP1-T78A, and VP1-G88A could affect the assembly of the virus capsid proteins, resulting in the production of abnormal virions with reduced infectivity. Specifically, the substitution of VP1-T75A affected the maturation cleavage of the VP0 precursor, leading to deficiencies in binding to receptor scavenger receptor class B2 (SCARB2), viral attachment, internalization, and even uncoating. For the mutants of T78A and G88A, a significant reduction in virion-associated genomic RNA was observed, suggesting that more noninfectious empty particles were produced during viral assembly. Interestingly, the VP1-T75A variant showed weak replication in cell cultures but demonstrated increased virulence in BALB/c neonatal mice, which might be due to the difference in viral receptors among mammalian species. Taken together, our data revealed the important role of the highly conserved residues T75, T78, and G88 in VP1 protein in the infectivity of EV-A71. Characterizing these novel determinants of EV-A71 virulence would contribute to rationally developing effective treatments and broadly protective vaccine candidates.</p><p><strong>Importance: </strong>EV-A71 causes hand, foot, and mouth disease in children. In this study, we discovered three highly conserved residues at positions 75, 78, and 88 of the capsid protein VP1 as the potential virulence determinants of EV-A71, which can influence viral replication by regulating the assembly of EV-A71. Mechanistic studies revealed that VP1-T75A could affect the maturation cleavage of the VP0 precursor, resulting in deficiencies in binding to the receptor SCARB2, viral attachment, internalization, and even uncoating. For the mutants of T78A and G88A, more noninfectious empty particles were produced during viral assembly. The discovery of these novel determinants of EV-A71 virulence will promote the study of the pathogenesis of enteroviruses.</p>","PeriodicalId":17583,"journal":{"name":"Journal of Virology","volume":" ","pages":"e0165524"},"PeriodicalIF":4.0,"publicationDate":"2024-11-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142622794","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
SRCAP is involved in porcine reproductive and respiratory syndrome virus activated Notch signaling pathway. SRCAP 参与猪繁殖与呼吸综合征病毒激活的 Notch 信号通路。
IF 4 2区 医学 Q2 VIROLOGY Pub Date : 2024-11-12 DOI: 10.1128/jvi.01216-24
Guofei Ding, Yingchao Li, Dexin Li, Mingyu Dou, Chaolun Fu, Ting Chen, Xinyu Cui, Qin Zhang, Pingping Yang, Yanmeng Hou, Sidang Liu, Yihong Xiao

Porcine reproductive and respiratory syndrome virus (PRRSV) is the cause of porcine reproductive and respiratory syndrome (PRRS); a disease of pigs, which results in great economic losses in the pork industry. The non-structural protein 4 (Nsp4), a 3C-like serine protease responsible for most non-structural protein processing, plays an essential role in PRRSV infection. We used label-free quantitative proteomics to elucidate the Nsp4 interactome and SRCAP was identified as one of the interactors. SRCAP facilitated PRRSV infection by activating non-canonical Notch signaling. The ATPase I-IV domain in SRCAP and the 122VITEA126 in Nsp4 were identified as the interacting sites. The infection of recovered mutant rTA-12/5A (122AAAAA126) could not activate Notch signaling. The results indicated that 122VITEA126 in Nsp4 were key sites to determine the function of SRCAP and their interaction. A function of Nsp4 in activating the Notch signaling pathway was discovered. Block Notch signaling pathway could inhibit PRRSV infection both in vitro and in vivo which may lead to the development of novel therapeutic antiviral strategies.

Importance: In the present study, the interactome of the NSP4 originating from PRRSV was studied and SRCAP was confirmed as one of the interactors. Mechanism study showed the interaction of Nsp4 and SRCAP was found to facilitate PRRSV infection by activating non-canonical Notch signaling. ATPase Ⅰ-Ⅳ domain in SRCAP and the 122VITEA126 in Nsp4 were identified as the interacting sites that demined the activating of Notch signaling. Block Notch signaling pathway could inhibit PRRSV infection in vitro and in vivo which may be a new target for antiviral drug development.

猪繁殖与呼吸综合征病毒(PRRSV)是猪繁殖与呼吸综合征(PRRS)的病原体,这种猪病给猪肉行业造成了巨大的经济损失。非结构蛋白 4(Nsp4)是一种 3C 类丝氨酸蛋白酶,负责大部分非结构蛋白的加工,在 PRRSV 感染中起着至关重要的作用。我们利用无标记定量蛋白质组学阐明了 Nsp4 的相互作用组,发现 SRCAP 是其中的一个相互作用因子。SRCAP通过激活非典型Notch信号转导促进了PRRSV感染。SRCAP的ATPase I-IV结构域和Nsp4的122VITEA126被确定为相互作用位点。感染恢复突变体rTA-12/5A(122AAAAAA126)不能激活Notch信号。结果表明,Nsp4中的122VITEA126是决定SRCAP功能及其相互作用的关键位点。发现了Nsp4激活Notch信号通路的功能。阻断Notch信号通路可抑制体外和体内的PRRSV感染,从而开发出新型的抗病毒治疗策略:本研究对源自 PRRSV 的 NSP4 的相互作用组进行了研究,并证实 SRCAP 是其中的一个相互作用因子。机理研究表明,Nsp4与SRCAP的相互作用可通过激活非典型Notch信号来促进PRRSV感染。SRCAP中的ATPase Ⅰ-Ⅳ结构域和Nsp4中的122VITEA126结构域被确定为抑制Notch信号激活的相互作用位点。阻断Notch信号通路可抑制PRRSV的体外和体内感染,这可能是抗病毒药物开发的一个新靶点。
{"title":"SRCAP is involved in porcine reproductive and respiratory syndrome virus activated Notch signaling pathway.","authors":"Guofei Ding, Yingchao Li, Dexin Li, Mingyu Dou, Chaolun Fu, Ting Chen, Xinyu Cui, Qin Zhang, Pingping Yang, Yanmeng Hou, Sidang Liu, Yihong Xiao","doi":"10.1128/jvi.01216-24","DOIUrl":"https://doi.org/10.1128/jvi.01216-24","url":null,"abstract":"<p><p>Porcine reproductive and respiratory syndrome viru<i>s</i> (PRRSV) is the cause of porcine reproductive and respiratory syndrome (PRRS); a disease of pigs, which results in great economic losses in the pork industry. The non-structural protein 4 (Nsp4), a 3C-like serine protease responsible for most non-structural protein processing, plays an essential role in PRRSV infection. We used label-free quantitative proteomics to elucidate the Nsp4 interactome and SRCAP was identified as one of the interactors. SRCAP facilitated PRRSV infection by activating non-canonical Notch signaling. The ATPase I-IV domain in SRCAP and the <sup>122</sup>VITEA<sup>126</sup> in Nsp4 were identified as the interacting sites. The infection of recovered mutant rTA-12/5A (<sup>122</sup>AAAAA<sup>126</sup>) could not activate Notch signaling. The results indicated that <sup>122</sup>VITEA<sup>126</sup> in Nsp4 were key sites to determine the function of SRCAP and their interaction. A function of Nsp4 in activating the Notch signaling pathway was discovered. Block Notch signaling pathway could inhibit PRRSV infection both <i>in vitro</i> and <i>in vivo</i> which may lead to the development of novel therapeutic antiviral strategies.</p><p><strong>Importance: </strong>In the present study, the interactome of the NSP4 originating from PRRSV was studied and SRCAP was confirmed as one of the interactors. Mechanism study showed the interaction of Nsp4 and SRCAP was found to facilitate PRRSV infection by activating non-canonical Notch signaling. ATPase Ⅰ-Ⅳ domain in SRCAP and the <sup>122</sup>VITEA<sup>126</sup> in Nsp4 were identified as the interacting sites that demined the activating of Notch signaling. Block Notch signaling pathway could inhibit PRRSV infection <i>in vitro</i> and <i>in vivo</i> which may be a new target for antiviral drug development.</p>","PeriodicalId":17583,"journal":{"name":"Journal of Virology","volume":" ","pages":"e0121624"},"PeriodicalIF":4.0,"publicationDate":"2024-11-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142622805","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
Quantifying the impact of vaccination on transmission and diversity of influenza A variants in pigs. 量化疫苗接种对猪体内甲型流感变种传播和多样性的影响。
IF 4 2区 医学 Q2 VIROLOGY Pub Date : 2024-11-12 DOI: 10.1128/jvi.01245-24
Chong Li, Marie R Culhane, Declan C Schroeder, Maxim C-J Cheeran, Lucina Galina Pantoja, Micah L Jansen, Montserrat Torremorell

Global evolutionary dynamics of influenza A virus (IAV) are fundamentally driven by the extent of virus diversity generated, transmitted, and shaped in individual hosts. How vaccination affects the degree of IAV genetic diversity that can be transmitted and expanded in pigs is unknown. To evaluate the effect of vaccination on the transmission of genetically distinct IAV variants and their diversity after transmission in pigs, we examined the whole genome of IAV recovered from the nasal cavities of pigs vaccinated with different influenza immunization regimens after being infected simultaneously by H1N1 and H3N2 IAVs using a seeder pig model. We found that the seeder pigs harbored more diversified virus populations than the contact pigs. Among contact pigs, H3N2 and H1N1 viruses recovered from pigs vaccinated with a single dose of an unmatched modified live vaccine generally accumulated more extensive genetic mutations than non-vaccinated pigs. Furthermore, the non-sterilizing immunity elicited by the single-dose-modified live vaccine may have exerted positive selection on H1 antigenic regions as we detected significantly higher nonsynonymous but lower synonymous evolutionary rates in H1 antigenic regions than non-antigenic regions. In addition, we observed that the vaccinated pigs shared significantly less proportion of H3N2 variants with seeder pigs than unvaccinated pigs. These results indicated that vaccination might reduce the impact of transmitted influenza variants on the overall diversity of IAV populations harbored in recipient pigs and that within-host genetic selection of IAV is more likely to occur in pigs vaccinated with improperly matched vaccines.IMPORTANCEUnderstanding how vaccination shapes the diversity of influenza variants that transmit and propagate among pigs is essential for designing effective IAV surveillance and control programs. Current knowledge about the transmission of IAV variants has primarily been explored in humans during natural infection. However, how immunity elicited by improperly matched vaccines affects the degree of IAV genetic diversity that can be transmitted and expanded in pigs at the whole-genome level is unknown. We analyzed IAV sequences from samples collected daily from experimentally infected pigs vaccinated with various protocols in a field-represented IAV co-infection model. We found that vaccine-induced non-sterilizing immunity might promote genetic variation on the IAV genome and drive positive selection at antigenic sites during infection. In addition, a smaller proportion of H3N2 viral variants were shared between seeder pigs and vaccinated pigs, suggesting the influence of vaccination on shaping the virus genomic diversity in recipient pigs during the transmission events.

甲型流感病毒(IAV)的全球进化动态从根本上说是由病毒在宿主个体中产生、传播和形成的多样性程度所驱动的。疫苗接种如何影响可在猪体内传播和扩展的 IAV 遗传多样性程度尚不清楚。为了评估疫苗接种对基因不同的 IAV 变种的传播及其在猪体内传播后的多样性的影响,我们使用播种猪模型研究了同时感染 H1N1 和 H3N2 IAV 后接种不同流感免疫方案的猪从鼻腔中回收的 IAV 的全基因组。我们发现,与接触猪相比,播种猪体内的病毒群更加多样化。在接触猪中,从接种过一剂非匹配改良活疫苗的猪身上回收的 H3N2 和 H1N1 病毒通常比未接种过疫苗的猪积累了更多的基因突变。此外,单剂改良活疫苗引起的非灭菌免疫可能对 H1 抗原区产生了正选择,因为我们发现 H1 抗原区的非同义进化率明显高于非抗原区,但同义进化率却低于非抗原区。此外,我们还观察到,接种疫苗的猪与播种猪共享 H3N2 变体的比例明显低于未接种疫苗的猪。这些结果表明,接种疫苗可能会减少传播的流感变种对受种猪所蕴藏的 IAV 群体总体多样性的影响,而且接种了匹配不当的疫苗的猪更有可能发生 IAV 的宿主内遗传选择。重要意义 了解接种疫苗如何影响猪之间传播和繁殖的流感变种的多样性,对于设计有效的 IAV 监控计划至关重要。目前有关 IAV 变种传播的知识主要是在人类自然感染过程中探索的。然而,匹配不当的疫苗所引起的免疫力如何影响 IAV 基因多样性在猪体内全基因组水平上的传播和扩展程度尚不清楚。我们分析了每天从实验性感染猪只采集的样本中获得的 IAV 序列,这些猪只在现场代表的 IAV 共感染模型中接种了不同方案的疫苗。我们发现,疫苗诱导的非灭菌免疫可能会促进 IAV 基因组的遗传变异,并在感染过程中推动抗原位点的正选择。此外,播种猪和疫苗接种猪之间共享的 H3N2 病毒变体比例较小,这表明疫苗接种在传播过程中对受种猪病毒基因组多样性的形成产生了影响。
{"title":"Quantifying the impact of vaccination on transmission and diversity of influenza A variants in pigs.","authors":"Chong Li, Marie R Culhane, Declan C Schroeder, Maxim C-J Cheeran, Lucina Galina Pantoja, Micah L Jansen, Montserrat Torremorell","doi":"10.1128/jvi.01245-24","DOIUrl":"https://doi.org/10.1128/jvi.01245-24","url":null,"abstract":"<p><p>Global evolutionary dynamics of influenza A virus (IAV) are fundamentally driven by the extent of virus diversity generated, transmitted, and shaped in individual hosts. How vaccination affects the degree of IAV genetic diversity that can be transmitted and expanded in pigs is unknown. To evaluate the effect of vaccination on the transmission of genetically distinct IAV variants and their diversity after transmission in pigs, we examined the whole genome of IAV recovered from the nasal cavities of pigs vaccinated with different influenza immunization regimens after being infected simultaneously by H1N1 and H3N2 IAVs using a seeder pig model. We found that the seeder pigs harbored more diversified virus populations than the contact pigs. Among contact pigs, H3N2 and H1N1 viruses recovered from pigs vaccinated with a single dose of an unmatched modified live vaccine generally accumulated more extensive genetic mutations than non-vaccinated pigs. Furthermore, the non-sterilizing immunity elicited by the single-dose-modified live vaccine may have exerted positive selection on H1 antigenic regions as we detected significantly higher nonsynonymous but lower synonymous evolutionary rates in H1 antigenic regions than non-antigenic regions. In addition, we observed that the vaccinated pigs shared significantly less proportion of H3N2 variants with seeder pigs than unvaccinated pigs. These results indicated that vaccination might reduce the impact of transmitted influenza variants on the overall diversity of IAV populations harbored in recipient pigs and that within-host genetic selection of IAV is more likely to occur in pigs vaccinated with improperly matched vaccines.IMPORTANCEUnderstanding how vaccination shapes the diversity of influenza variants that transmit and propagate among pigs is essential for designing effective IAV surveillance and control programs. Current knowledge about the transmission of IAV variants has primarily been explored in humans during natural infection. However, how immunity elicited by improperly matched vaccines affects the degree of IAV genetic diversity that can be transmitted and expanded in pigs at the whole-genome level is unknown. We analyzed IAV sequences from samples collected daily from experimentally infected pigs vaccinated with various protocols in a field-represented IAV co-infection model. We found that vaccine-induced non-sterilizing immunity might promote genetic variation on the IAV genome and drive positive selection at antigenic sites during infection. In addition, a smaller proportion of H3N2 viral variants were shared between seeder pigs and vaccinated pigs, suggesting the influence of vaccination on shaping the virus genomic diversity in recipient pigs during the transmission events.</p>","PeriodicalId":17583,"journal":{"name":"Journal of Virology","volume":" ","pages":"e0124524"},"PeriodicalIF":4.0,"publicationDate":"2024-11-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142622800","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