首页 > 最新文献

Journal of Virology最新文献

英文 中文
Viroporin-like activity of the hairpin transmembrane domain of African swine fever virus B169L protein. 非洲猪瘟病毒 B169L 蛋白发夹跨膜结构域的类病毒素活性。
IF 4 2区 医学 Q2 VIROLOGY Pub Date : 2024-08-20 Epub Date: 2024-07-09 DOI: 10.1128/jvi.00231-24
Douglas P Gladue, Lidia Gomez-Lucas, Eneko Largo, Elizabeth Ramirez-Medina, Johana Torralba, Maria Queralt-Martín, Antonio Alcaraz, Lauro Velazquez-Salinas, Jose L Nieva, Manuel V Borca

African swine fever virus (ASFV) is the causative agent of a contagious disease affecting wild and domestic swine. The function of B169L protein, as a potential integral structural membrane protein, remains to be experimentally characterized. Using state-of-the-art bioinformatics tools, we confirm here earlier predictions indicating the presence of an integral membrane helical hairpin, and further suggest anchoring of this protein to the ER membrane, with both terminal ends facing the lumen of the organelle. Our evolutionary analysis confirmed the importance of purifying selection in the preservation of the identified domains during the evolution of B169L in nature. Also, we address the possible function of this hairpin transmembrane domain (HTMD) as a class IIA viroporin. Expression of GFP fusion proteins in the absence of a signal peptide supported B169L insertion into the ER as a Type III membrane protein and the formation of oligomers therein. Overlapping peptides that spanned the B169L HTMD were reconstituted into ER-like membranes and the adopted structures analyzed by infrared spectroscopy. Consistent with the predictions, B169L transmembrane sequences adopted α-helical conformations in lipid bilayers. Moreover, single vesicle permeability assays demonstrated the assembly of lytic pores in ER-like membranes by B169L transmembrane helices, a capacity confirmed by ion-channel activity measurements in planar bilayers. Emphasizing the relevance of these observations, pore-forming activities were not observed in the case of transmembrane helices derived from EP84R, another ASFV protein predicted to anchor to membranes through a α-helical HTMD. Overall, our results support predictions of viroporin-like function for the B169L HTMD.IMPORTANCEAfrican swine fever (ASF), a devastating disease affecting domestic swine, is widely spread in Eurasia, producing significant economic problems in the pork industry. Approaches to prevent/cure the disease are mainly restricted to the limited information concerning the role of most of the genes encoded by the large (160-170 kba) virus genome. In this report, we present the experimental data on the functional characterization of the African swine fever virus (ASFV) gene B169L. Data presented here indicates that the B169L gene encodes for an essential membrane-associated protein with a viroporin function.

非洲猪瘟病毒(ASFV)是一种影响野猪和家猪的传染性疾病的病原体。B169L 蛋白作为一种潜在的整体结构膜蛋白,其功能仍有待实验鉴定。利用最先进的生物信息学工具,我们在此证实了早先的预测,即存在一个整体膜螺旋发夹,并进一步表明该蛋白锚定在 ER 膜上,其两端面向细胞器的内腔。我们的进化分析证实了纯化选择对 B169L 在自然界进化过程中保留已确定结构域的重要性。此外,我们还探讨了这种发夹式跨膜结构域(HTMD)作为 IIA 类病毒蛋白可能具有的功能。在没有信号肽的情况下表达 GFP 融合蛋白支持 B169L 作为 III 型膜蛋白插入 ER 并在其中形成寡聚体。将跨越 B169L HTMD 的重叠肽重组为类似 ER 的膜,并通过红外光谱分析所采用的结构。与预测一致,B169L 跨膜序列在脂质双层膜中采用了 α-螺旋构象。此外,单个囊泡通透性试验表明,B169L 跨膜螺旋在类似 ER 的膜中组装了裂孔,平面双层膜中的离子通道活性测量也证实了这一能力。强调这些观察结果的相关性的是,在来自 EP84R 的跨膜螺旋的情况下没有观察到孔形成活性,EP84R 是另一种 ASFV 蛋白,预测其通过 α-helical HTMD 固定在膜上。重要意义非洲猪瘟(ASF)是一种影响家猪的毁灭性疾病,在欧亚大陆广泛传播,给猪肉行业带来了严重的经济问题。预防/治疗该疾病的方法主要局限于有关大型(160-170 kba)病毒基因组所编码的大多数基因的作用的有限信息。在本报告中,我们介绍了有关非洲猪瘟病毒(ASFV)基因 B169L 功能特征的实验数据。本文提供的数据表明,B169L 基因编码一种重要的膜相关蛋白,具有病毒蛋白功能。
{"title":"Viroporin-like activity of the hairpin transmembrane domain of African swine fever virus B169L protein.","authors":"Douglas P Gladue, Lidia Gomez-Lucas, Eneko Largo, Elizabeth Ramirez-Medina, Johana Torralba, Maria Queralt-Martín, Antonio Alcaraz, Lauro Velazquez-Salinas, Jose L Nieva, Manuel V Borca","doi":"10.1128/jvi.00231-24","DOIUrl":"10.1128/jvi.00231-24","url":null,"abstract":"<p><p>African swine fever virus (ASFV) is the causative agent of a contagious disease affecting wild and domestic swine. The function of B169L protein, as a potential integral structural membrane protein, remains to be experimentally characterized. Using state-of-the-art bioinformatics tools, we confirm here earlier predictions indicating the presence of an integral membrane helical hairpin, and further suggest anchoring of this protein to the ER membrane, with both terminal ends facing the lumen of the organelle. Our evolutionary analysis confirmed the importance of purifying selection in the preservation of the identified domains during the evolution of B169L in nature. Also, we address the possible function of this hairpin transmembrane domain (HTMD) as a class IIA viroporin. Expression of GFP fusion proteins in the absence of a signal peptide supported B169L insertion into the ER as a Type III membrane protein and the formation of oligomers therein. Overlapping peptides that spanned the B169L HTMD were reconstituted into ER-like membranes and the adopted structures analyzed by infrared spectroscopy. Consistent with the predictions, B169L transmembrane sequences adopted α-helical conformations in lipid bilayers. Moreover, single vesicle permeability assays demonstrated the assembly of lytic pores in ER-like membranes by B169L transmembrane helices, a capacity confirmed by ion-channel activity measurements in planar bilayers. Emphasizing the relevance of these observations, pore-forming activities were not observed in the case of transmembrane helices derived from EP84R, another ASFV protein predicted to anchor to membranes through a α-helical HTMD. Overall, our results support predictions of viroporin-like function for the B169L HTMD.IMPORTANCEAfrican swine fever (ASF), a devastating disease affecting domestic swine, is widely spread in Eurasia, producing significant economic problems in the pork industry. Approaches to prevent/cure the disease are mainly restricted to the limited information concerning the role of most of the genes encoded by the large (160-170 kba) virus genome. In this report, we present the experimental data on the functional characterization of the African swine fever virus (ASFV) gene B169L. Data presented here indicates that the B169L gene encodes for an essential membrane-associated protein with a viroporin function.</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/PMC11334534/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141559014","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
Vaccine strains of Rift Valley fever virus exhibit attenuation at the maternal-fetal placental interface. 裂谷热病毒疫苗株在母胎胎盘界面表现出衰减。
IF 4 2区 医学 Q2 VIROLOGY Pub Date : 2024-08-20 Epub Date: 2024-07-17 DOI: 10.1128/jvi.00983-24
Cynthia M McMillen, Christina Megli, Rebecca Radisic, Lauren B Skvarca, Ryan M Hoehl, Devin A Boyles, Jackson J McGaughey, Brian H Bird, Anita K McElroy, Amy L Hartman

Rift Valley fever virus (RVFV) infection causes abortions in ruminant livestock and is associated with an increased likelihood of miscarriages in women. Using sheep and human placenta explant cultures, we sought to identify tissues at the maternal-fetal interface targeted by RVFV. Sheep villi and fetal membranes were highly permissive to RVFV infection resulting in markedly higher virus titers than human cultures. Sheep cultures were most permissive to wild-type RVFV and ΔNSm infection, while live-attenuated RVFV vaccines (LAVs; MP-12, ΔNSs, and ΔNSs/ΔNSm) exhibited reduced replication. The human fetal membrane restricted wild-type and LAV replication, and when infection occurred, it was prominent on the maternal-facing side. Type I and type III interferons were induced in human villi exposed to LAVs lacking the NSs protein. This study supports the use of sheep and human placenta explants to understand vertical transmission of RVFV in mammals and whether LAVs are attenuated at the maternal-fetal interface.IMPORTANCEA direct comparison of replication of Rift Valley fever virus (RVFV) in sheep and human placental explants reveals comparative efficiencies and permissivity to infection and replication. Vaccine strains of RVFV demonstrated reduced infection and replication capacity in the mammalian placenta. This study represents the first direct cross-host comparison of the vertical transmission capacity of this high-priority emerging mosquito-transmitted virus.

裂谷热病毒(RVFV)感染会导致反刍家畜流产,并增加妇女流产的可能性。我们利用绵羊和人类胎盘外植体培养物,试图确定 RVFV 针对的母胎界面组织。绵羊绒毛和胎膜对 RVFV 感染的容许度很高,因此病毒滴度明显高于人类培养物。绵羊培养物最易受野生型 RVFV 和 ΔNSm 感染,而 RVFV 减毒活疫苗(LAVs;MP-12、ΔNSs 和 ΔNSs/ΔNSm)的复制能力则有所下降。人类胎膜限制了野生型和 LAV 的复制,当发生感染时,感染主要发生在面向母体的一侧。人绒毛暴露于缺乏 NSs 蛋白的 LAV 后,可诱导 I 型和 III 型干扰素。这项研究支持使用绵羊和人类胎盘外植体来了解 RVFV 在哺乳动物中的垂直传播,以及 LAVs 是否在母胎界面被减弱。RVFV 疫苗株在哺乳动物胎盘中的感染和复制能力均有所下降。这项研究首次直接跨宿主比较了这种高度优先的新兴蚊媒传播病毒的垂直传播能力。
{"title":"Vaccine strains of Rift Valley fever virus exhibit attenuation at the maternal-fetal placental interface.","authors":"Cynthia M McMillen, Christina Megli, Rebecca Radisic, Lauren B Skvarca, Ryan M Hoehl, Devin A Boyles, Jackson J McGaughey, Brian H Bird, Anita K McElroy, Amy L Hartman","doi":"10.1128/jvi.00983-24","DOIUrl":"10.1128/jvi.00983-24","url":null,"abstract":"<p><p>Rift Valley fever virus (RVFV) infection causes abortions in ruminant livestock and is associated with an increased likelihood of miscarriages in women. Using sheep and human placenta explant cultures, we sought to identify tissues at the maternal-fetal interface targeted by RVFV. Sheep villi and fetal membranes were highly permissive to RVFV infection resulting in markedly higher virus titers than human cultures. Sheep cultures were most permissive to wild-type RVFV and ΔNSm infection, while live-attenuated RVFV vaccines (LAVs; MP-12, ΔNSs, and ΔNSs/ΔNSm) exhibited reduced replication. The human fetal membrane restricted wild-type and LAV replication, and when infection occurred, it was prominent on the maternal-facing side. Type I and type III interferons were induced in human villi exposed to LAVs lacking the NSs protein. This study supports the use of sheep and human placenta explants to understand vertical transmission of RVFV in mammals and whether LAVs are attenuated at the maternal-fetal interface.IMPORTANCEA direct comparison of replication of Rift Valley fever virus (RVFV) in sheep and human placental explants reveals comparative efficiencies and permissivity to infection and replication. Vaccine strains of RVFV demonstrated reduced infection and replication capacity in the mammalian placenta. This study represents the first direct cross-host comparison of the vertical transmission capacity of this high-priority emerging mosquito-transmitted virus.</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/PMC11334480/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141627114","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
Structural determinants of spike infectivity in bat SARS-like coronaviruses RsSHC014 and WIV1. 蝙蝠 SARS 样冠状病毒 RsSHC014 和 WIV1 穗状病毒感染性的结构决定因素。
IF 4 2区 医学 Q2 VIROLOGY Pub Date : 2024-08-20 Epub Date: 2024-07-19 DOI: 10.1128/jvi.00342-24
Shuyuan Qiao, Xinquan Wang

The recurrent spillovers of coronaviruses (CoVs) have posed severe threats to public health and the global economy. Bat severe acute respiratory syndrome (SARS)-like CoVs RsSHC014 and WIV1, currently circulating in bat populations, are poised for human emergence. The trimeric spike (S) glycoprotein, responsible for receptor recognition and membrane fusion, plays a critical role in cross-species transmission and infection. Here, we determined the cryo-electron microscopy (EM) structures of the RsSHC014 S protein in the closed state at 2.9 Å, the WIV1 S protein in the closed state at 2.8 Å, and the intermediate state at 4.0 Å. In the intermediate state, one receptor-binding domain (RBD) is in the "down" position, while the other two RBDs exhibit poor density. We also resolved the complex structure of the WIV1 S protein bound to human ACE2 (hACE2) at 4.5 Å, which provides structural basis for the future emergence of WIV1 in humans. Through biochemical experiments, we found that despite strong binding affinities between the RBDs and both human and civet ACE2, the pseudoviruses of RsSHC014, but not WIV1, failed to infect 293T cells overexpressing either human or civet ACE2. Mutagenesis analysis revealed that the Y623H substitution, located in the SD2 region, significantly improved the cell entry efficiency of RsSHC014 pseudoviruses, which is likely accomplished by promoting the open conformation of spike glycoproteins. Our findings emphasize the necessity of both efficient RBD lifting and tight RBD-hACE2 binding for viral infection and underscore the significance of the 623 site of the spike glycoprotein for the infectivity of bat SARS-like CoVs.

Importance: The bat SARS-like CoVs RsSHC014 and WIV1 can use hACE2 for cell entry without further adaptation, indicating their potential risk of emergence in human populations. The S glycoprotein, responsible for receptor recognition and membrane fusion, plays a crucial role in cross-species transmission and infection. In this study, we determined the cryo-EM structures of the S glycoproteins of RsSHC014 and WIV1. Detailed comparisons revealed dynamic structural variations within spike proteins. We also elucidated the complex structure of WIV1 S-hACE2, providing structural evidence for the potential emergence of WIV1 in humans. Although RsSHC014 and WIV1 had similar hACE2-binding affinities, they exhibited distinct pseudovirus cell entry behavior. Through mutagenesis and cryo-EM analysis, we revealed that besides the structural variations, the 623 site in the SD2 region is another important structural determinant of spike infectivity.

冠状病毒(CoVs)的反复蔓延对公共卫生和全球经济构成了严重威胁。类似于蝙蝠严重急性呼吸系统综合征(SARS)的冠状病毒 RsSHC014 和 WIV1 目前在蝙蝠种群中流行,并有望在人类中出现。三聚体尖峰(S)糖蛋白负责受体识别和膜融合,在跨物种传播和感染中起着关键作用。在这里,我们测定了 RsSHC014 S 蛋白在封闭状态下 2.9 Å 的低温电子显微镜(EM)结构、WIV1 S 蛋白在封闭状态下 2.8 Å 的低温电子显微镜(EM)结构以及中间状态下 4.0 Å 的低温电子显微镜(EM)结构。我们还解析了 WIV1 S 蛋白与人类 ACE2(hACE2)结合的复合物结构,该结构达到 4.5 Å,为将来 WIV1 在人类中的出现提供了结构依据。通过生化实验,我们发现尽管 RBD 与人类和果子狸 ACE2 都有很强的结合亲和力,但 RsSHC014 的假病毒却不能感染过表达人类或果子狸 ACE2 的 293T 细胞,而 WIV1 则不能。突变分析表明,位于SD2区的Y623H取代显著提高了RsSHC014假病毒进入细胞的效率,这可能是通过促进尖峰糖蛋白的开放构象实现的。我们的发现强调了病毒感染需要有效的 RBD 提升和 RBD-hACE2 紧密结合,并突出了尖峰糖蛋白的 623 位点对蝙蝠 SARS 样 CoVs 感染性的重要性:蝙蝠SARS样CoVs RsSHC014和WIV1可利用hACE2进入细胞而无需进一步适应,这表明它们有可能出现在人类群体中。S糖蛋白负责受体识别和膜融合,在跨物种传播和感染中起着至关重要的作用。在这项研究中,我们测定了 RsSHC014 和 WIV1 的 S 糖蛋白的冷冻电镜结构。详细比较显示了尖峰蛋白内部的动态结构变化。我们还阐明了 WIV1 S-hACE2 的复合结构,为 WIV1 可能在人类中出现提供了结构证据。虽然 RsSHC014 和 WIV1 具有相似的 hACE2 结合亲和力,但它们却表现出不同的伪病毒细胞进入行为。通过诱变和低温电子显微镜分析,我们发现除了结构变异外,SD2区的623位点是决定尖峰感染性的另一个重要结构因素。
{"title":"Structural determinants of spike infectivity in bat SARS-like coronaviruses RsSHC014 and WIV1.","authors":"Shuyuan Qiao, Xinquan Wang","doi":"10.1128/jvi.00342-24","DOIUrl":"10.1128/jvi.00342-24","url":null,"abstract":"<p><p>The recurrent spillovers of coronaviruses (CoVs) have posed severe threats to public health and the global economy. Bat severe acute respiratory syndrome (SARS)-like CoVs RsSHC014 and WIV1, currently circulating in bat populations, are poised for human emergence. The trimeric spike (S) glycoprotein, responsible for receptor recognition and membrane fusion, plays a critical role in cross-species transmission and infection. Here, we determined the cryo-electron microscopy (EM) structures of the RsSHC014 S protein in the closed state at 2.9 Å, the WIV1 S protein in the closed state at 2.8 Å, and the intermediate state at 4.0 Å. In the intermediate state, one receptor-binding domain (RBD) is in the \"down\" position, while the other two RBDs exhibit poor density. We also resolved the complex structure of the WIV1 S protein bound to human ACE2 (hACE2) at 4.5 Å, which provides structural basis for the future emergence of WIV1 in humans. Through biochemical experiments, we found that despite strong binding affinities between the RBDs and both human and civet ACE2, the pseudoviruses of RsSHC014, but not WIV1, failed to infect 293T cells overexpressing either human or civet ACE2. Mutagenesis analysis revealed that the Y623H substitution, located in the SD2 region, significantly improved the cell entry efficiency of RsSHC014 pseudoviruses, which is likely accomplished by promoting the open conformation of spike glycoproteins. Our findings emphasize the necessity of both efficient RBD lifting and tight RBD-hACE2 binding for viral infection and underscore the significance of the 623 site of the spike glycoprotein for the infectivity of bat SARS-like CoVs.</p><p><strong>Importance: </strong>The bat SARS-like CoVs RsSHC014 and WIV1 can use hACE2 for cell entry without further adaptation, indicating their potential risk of emergence in human populations. The S glycoprotein, responsible for receptor recognition and membrane fusion, plays a crucial role in cross-species transmission and infection. In this study, we determined the cryo-EM structures of the S glycoproteins of RsSHC014 and WIV1. Detailed comparisons revealed dynamic structural variations within spike proteins. We also elucidated the complex structure of WIV1 S-hACE2, providing structural evidence for the potential emergence of WIV1 in humans. Although RsSHC014 and WIV1 had similar hACE2-binding affinities, they exhibited distinct pseudovirus cell entry behavior. Through mutagenesis and cryo-EM analysis, we revealed that besides the structural variations, the 623 site in the SD2 region is another important structural determinant of spike infectivity.</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/PMC11334503/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141723825","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
Effective inhibition of PDCoV infection in chimeric APN gene-edited neonatal pigs. 有效抑制嵌合 APN 基因编辑的新生猪感染 PDCoV。
IF 4 2区 医学 Q2 VIROLOGY Pub Date : 2024-08-20 Epub Date: 2024-07-30 DOI: 10.1128/jvi.00611-24
Jizong Li, Jian Zhou, Tianyi Zhang, Heyong Wu, Feng Li, Chunyun Qi, Liyuan Fan, Xuesong Yuan, Wei Wang, Rongli Guo, Baochao Fan, Xiaochun Tang, Daxin Pang, Hongsheng Ouyang, Zicong Xie, Bin Li

Porcine deltacoronavirus (PDCoV), an emerging enteropathogenic coronavirus, is a serious threat to piglets and has zoonotic potential. Here, we aimed to further explore the role of aminopeptidase N (APN) as a receptor for PDCoV and test the inhibitory effect of a chimeric APN protein strategy on PDCoV infection. PK-15 cells and LLC-PK1 cells expressing chimeric APN were selected and infected with PDCoV. Viral replication was significantly decreased in these chimeric APN cells compared with that in control group cells. To further characterize the effect of the chimeric APN strategy on PDCoV infection in vitro, primary intestinal epithelial cells isolated from chimeric APN pigs were inoculated with PDCoV. Viral challenge of these cells led to decreased PDCoV infection. More importantly, virally challenged chimeric APN neonatal piglets displayed reduced viral load, significantly fewer microscopic lesions in the intestinal tissue, and no diarrhea. Taken together, these findings deepen our understanding of the mechanism of PDCoV infection and provide a valuable model for the production of disease-resistant animals.

Importance: Porcine deltacoronavirus (PDCoV), an emerging enteropathogenic coronavirus, causes diarrhea in piglets and possesses the potential to infect humans. However, there are currently no effective measures for the prevention or control of PDCoV infection. Here, we have developed PK-15 cells, LLC-PK1 cells, and primary intestinal epithelial cells expressing chimeric APN, and viral challenge of these cells led to decreased PDCoV infection. Furthermore, virally challenged chimeric APN neonatal piglets displayed reduced viral load, significantly fewer microscopic lesions in the intestinal tissue, and no diarrhea. These data show that chimeric APN is a promising strategy to combat PDCoV infection.

猪三角冠状病毒(PDCoV)是一种新出现的肠道致病性冠状病毒,对仔猪构成严重威胁,并有可能引起人畜共患病。在此,我们旨在进一步探索氨基肽酶 N(APN)作为 PDCoV 受体的作用,并测试嵌合 APN 蛋白策略对 PDCoV 感染的抑制作用。筛选出表达嵌合 APN 的 PK-15 细胞和 LLC-PK1 细胞并用 PDCoV 感染。与对照组细胞相比,这些嵌合 APN 细胞中的病毒复制明显减少。为了进一步确定嵌合 APN 策略对体外 PDCoV 感染的影响,用 PDCoV 接种了从嵌合 APN 猪分离出来的原代肠上皮细胞。对这些细胞进行病毒挑战可减少 PDCoV 感染。更重要的是,受到病毒挑战的嵌合 APN 新生仔猪的病毒载量降低,肠道组织的显微病变明显减少,而且没有腹泻。总之,这些发现加深了我们对 PDCoV 感染机制的理解,并为生产抗病动物提供了一个宝贵的模型:猪三角冠状病毒(PDCoV)是一种新出现的肠道致病性冠状病毒,会导致仔猪腹泻,并有可能感染人类。然而,目前还没有预防或控制 PDCoV 感染的有效措施。在这里,我们开发了表达嵌合 APN 的 PK-15 细胞、LLC-PK1 细胞和原代肠上皮细胞,病毒挑战这些细胞可减少 PDCoV 感染。此外,受到病毒挑战的嵌合 APN 新生仔猪的病毒载量减少,肠道组织的显微病变明显减少,而且没有腹泻。这些数据表明,嵌合 APN 是一种很有前景的抗 PDCoV 感染策略。
{"title":"Effective inhibition of PDCoV infection in chimeric APN gene-edited neonatal pigs.","authors":"Jizong Li, Jian Zhou, Tianyi Zhang, Heyong Wu, Feng Li, Chunyun Qi, Liyuan Fan, Xuesong Yuan, Wei Wang, Rongli Guo, Baochao Fan, Xiaochun Tang, Daxin Pang, Hongsheng Ouyang, Zicong Xie, Bin Li","doi":"10.1128/jvi.00611-24","DOIUrl":"10.1128/jvi.00611-24","url":null,"abstract":"<p><p>Porcine deltacoronavirus (PDCoV), an emerging enteropathogenic coronavirus, is a serious threat to piglets and has zoonotic potential. Here, we aimed to further explore the role of aminopeptidase N (APN) as a receptor for PDCoV and test the inhibitory effect of a chimeric APN protein strategy on PDCoV infection. PK-15 cells and LLC-PK1 cells expressing chimeric APN were selected and infected with PDCoV. Viral replication was significantly decreased in these chimeric APN cells compared with that in control group cells. To further characterize the effect of the chimeric APN strategy on PDCoV infection <i>in vitro</i>, primary intestinal epithelial cells isolated from chimeric APN pigs were inoculated with PDCoV. Viral challenge of these cells led to decreased PDCoV infection. More importantly, virally challenged chimeric APN neonatal piglets displayed reduced viral load, significantly fewer microscopic lesions in the intestinal tissue, and no diarrhea. Taken together, these findings deepen our understanding of the mechanism of PDCoV infection and provide a valuable model for the production of disease-resistant animals.</p><p><strong>Importance: </strong>Porcine deltacoronavirus (PDCoV), an emerging enteropathogenic coronavirus, causes diarrhea in piglets and possesses the potential to infect humans. However, there are currently no effective measures for the prevention or control of PDCoV infection. Here, we have developed PK-15 cells, LLC-PK1 cells, and primary intestinal epithelial cells expressing chimeric APN, and viral challenge of these cells led to decreased PDCoV infection. Furthermore, virally challenged chimeric APN neonatal piglets displayed reduced viral load, significantly fewer microscopic lesions in the intestinal tissue, and no diarrhea. These data show that chimeric APN is a promising strategy to combat 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/PMC11334500/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141792790","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
rAAV expressing a COBRA-designed influenza hemagglutinin generates a protective and durable adaptive immune response with a single dose. 表达 COBRA 设计的流感血凝素的 rAAV 只需一次给药就能产生保护性和持久的适应性免疫反应。
IF 4 2区 医学 Q2 VIROLOGY Pub Date : 2024-08-20 Epub Date: 2024-07-30 DOI: 10.1128/jvi.00781-24
Kristin B Wiggins, Stephen M Winston, Isaiah L Reeves, Jessica Gaevert, Yunyu Spence, Mark A Brimble, Brandi Livingston, Christopher L Morton, Paul G Thomas, Andrea J Sant, Ted M Ross, Andrew M Davidoff, Stacey Schultz-Cherry

Influenza remains a worldwide public health threat. Although seasonal influenza vaccines are currently the best means of preventing severe disease, the standard-of-care vaccines require frequent updating due to antigenic drift and can have low efficacy, particularly in vulnerable populations. Here, we demonstrate that a single administration of a recombinant adenovirus-associated virus (rAAV) vector expressing a computationally optimized broadly reactive antigen (COBRA)-derived influenza H1 hemagglutinin (HA) induces strongly neutralizing and broadly protective antibodies in naïve mice and ferrets with pre-existing influenza immunity. Following a lethal viral challenge, the rAAV-COBRA vaccine allowed for significantly reduced viral loads in the upper and lower respiratory tracts and complete protection from morbidity and mortality that lasted for at least 5 months post-vaccination. We observed no signs of antibody waning during this study. CpG motif enrichment of the antigen can act as an internal adjuvant to further enhance the immune responses to allow for lower vaccine dosages with the induction of unique interferon-producing CD4+ and CD8+ T cells specific to HA head and stem peptide sequences. Our studies highlight the utility of rAAV as an effective platform to improve seasonal influenza vaccines.

Importance: Developing an improved seasonal influenza vaccine remains an ambitious goal of researchers and clinicians alike. With influenza routinely causing severe epidemics with the potential to rise to pandemic levels, it is critical to create an effective, broadly protective, and durable vaccine to improve public health worldwide. As a potential solution, we created a rAAV viral vector expressing a COBRA-optimized influenza hemagglutinin antigen with modestly enriched CpG motifs to evoke a robust and long-lasting immune response after a single intramuscular dose without needing boosts or adjuvants. Importantly, the rAAV vaccine boosted antibody breadth to future strains in ferrets with pre-existing influenza immunity. Together, our data support further investigation into the utility of viral vectors as a potential avenue to improve our seasonal influenza vaccines.

流感仍然是一个全球性的公共卫生威胁。尽管季节性流感疫苗是目前预防严重疾病的最佳手段,但由于抗原漂移,标准疫苗需要经常更新,而且效力可能很低,尤其是在易感人群中。在这里,我们证明了单次给药重组腺病毒相关病毒(rAAV)载体表达计算优化的广泛反应抗原(COBRA)衍生的 H1 流感血凝素(HA),可在已有流感免疫力的天真小鼠和雪貂中诱导强中和抗体和广泛保护性抗体。在一次致命的病毒挑战后,rAAV-COBRA 疫苗可显著减少上呼吸道和下呼吸道的病毒载量,并在接种后至少 5 个月内完全避免发病和死亡。在这项研究中,我们没有观察到抗体减弱的迹象。抗原的 CpG 基序富集可作为一种内佐剂,进一步增强免疫反应,从而降低疫苗剂量,诱导出对 HA 头部和干肽序列具有特异性的产生干扰素的 CD4+ 和 CD8+ T 细胞。我们的研究强调了 rAAV 作为改进季节性流感疫苗有效平台的实用性:开发改良的季节性流感疫苗仍然是研究人员和临床医生的宏伟目标。由于流感经常引起严重的流行病,并有可能上升到大流行病的水平,因此研制一种有效、具有广泛保护性和持久性的疫苗对于改善全球公共卫生至关重要。作为一种潜在的解决方案,我们创建了一种表达 COBRA 优化的流感血凝素抗原的 rAAV 病毒载体,该抗原具有适度富集的 CpG 基序,只需肌肉注射一次,就能唤起强大而持久的免疫反应,而无需增强剂或佐剂。重要的是,rAAV 疫苗提高了原有流感免疫力的雪貂对未来毒株的抗体广度。总之,我们的数据支持进一步研究病毒载体作为改进季节性流感疫苗的潜在途径的效用。
{"title":"rAAV expressing a COBRA-designed influenza hemagglutinin generates a protective and durable adaptive immune response with a single dose.","authors":"Kristin B Wiggins, Stephen M Winston, Isaiah L Reeves, Jessica Gaevert, Yunyu Spence, Mark A Brimble, Brandi Livingston, Christopher L Morton, Paul G Thomas, Andrea J Sant, Ted M Ross, Andrew M Davidoff, Stacey Schultz-Cherry","doi":"10.1128/jvi.00781-24","DOIUrl":"10.1128/jvi.00781-24","url":null,"abstract":"<p><p>Influenza remains a worldwide public health threat. Although seasonal influenza vaccines are currently the best means of preventing severe disease, the standard-of-care vaccines require frequent updating due to antigenic drift and can have low efficacy, particularly in vulnerable populations. Here, we demonstrate that a single administration of a recombinant adenovirus-associated virus (rAAV) vector expressing a computationally optimized broadly reactive antigen (COBRA)-derived influenza H1 hemagglutinin (HA) induces strongly neutralizing and broadly protective antibodies in naïve mice and ferrets with pre-existing influenza immunity. Following a lethal viral challenge, the rAAV-COBRA vaccine allowed for significantly reduced viral loads in the upper and lower respiratory tracts and complete protection from morbidity and mortality that lasted for at least 5 months post-vaccination. We observed no signs of antibody waning during this study. CpG motif enrichment of the antigen can act as an internal adjuvant to further enhance the immune responses to allow for lower vaccine dosages with the induction of unique interferon-producing CD4+ and CD8+ T cells specific to HA head and stem peptide sequences. Our studies highlight the utility of rAAV as an effective platform to improve seasonal influenza vaccines.</p><p><strong>Importance: </strong>Developing an improved seasonal influenza vaccine remains an ambitious goal of researchers and clinicians alike. With influenza routinely causing severe epidemics with the potential to rise to pandemic levels, it is critical to create an effective, broadly protective, and durable vaccine to improve public health worldwide. As a potential solution, we created a rAAV viral vector expressing a COBRA-optimized influenza hemagglutinin antigen with modestly enriched CpG motifs to evoke a robust and long-lasting immune response after a single intramuscular dose without needing boosts or adjuvants. Importantly, the rAAV vaccine boosted antibody breadth to future strains in ferrets with pre-existing influenza immunity. Together, our data support further investigation into the utility of viral vectors as a potential avenue to improve our seasonal influenza vaccines.</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/PMC11338075/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141792832","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
Chikungunya virus release is reduced by TIM-1 receptors through binding of envelope phosphatidylserine. TIM-1受体通过结合包膜磷脂酰丝氨酸减少基孔肯雅病毒的释放。
IF 4 2区 医学 Q2 VIROLOGY Pub Date : 2024-08-20 Epub Date: 2024-07-15 DOI: 10.1128/jvi.00775-24
Judith M Reyes Ballista, Ashley J Hoover, Joseph T Noble, Marissa D Acciani, Kerri L Miazgowicz, Sarah A Harrison, Grace Andrea L Tabscott, Avery Duncan, Don N Barnes, Ariana R Jimenez, Melinda A Brindley

T-cell immunoglobin and mucin domain protein-1 (TIM-1) mediates entry of chikungunya virus (CHIKV) into some mammalian cells through the interaction with envelope phospholipids. While this interaction enhances entry, TIM-1 has been shown to tether newly formed HIV and Ebola virus particles, limiting their efficient release. In this study, we investigate the ability of surface receptors such as TIM-1 to sequester newly budded virions on the surface of infected cells. We established a luminescence reporter system to produce chikungunya viral particles that integrate nano-luciferase and easily quantify viral particles. We found that TIM-1 on the surface of host cells significantly reduced CHIKV release efficiency in comparison to other entry factors. Removal of cell surface TIM-1 through direct cellular knock-out or altering the cellular lipid distribution enhanced CHIKV release. Over the course of infection, CHIKV was able to counteract the tethering effect by gradually decreasing the surface levels of TIM-1 in a process mediated by the nonstructural protein 2. This study highlights the importance of phosphatidylserine receptors in mediating not only the entry of CHIKV but also its release and could aid in developing cell lines capable of enhanced vaccine production.

Importance: Chikungunya virus (CHIKV) is an enveloped alphavirus transmitted by the bites of infectious mosquitoes. Infection with CHIKV results in the development of fever, joint pain, and arthralgia that can become chronic and last for months after infection. Prevention of this disease is still highly focused on vector control strategies. In December 2023, a new live attenuated vaccine against CHIKV was approved by the FDA. We aimed to study the cellular factors involved in CHIKV release, to better understand CHIKV's ability to efficiently infect and spread among a wide variety of cell lines. We found that TIM-1 receptors can significantly abrogate CHIKV's ability to efficiently exit infected cells. This information can be beneficial for maximizing viral particle production in laboratory settings and during vaccine manufacturing.

T细胞免疫球蛋白和粘蛋白结构域蛋白-1(TIM-1)通过与包膜磷脂相互作用,介导基孔肯雅病毒(CHIKV)进入一些哺乳动物细胞。虽然这种相互作用能增强病毒的进入,但 TIM-1 已被证明能拴住新形成的 HIV 和埃博拉病毒颗粒,限制它们的有效释放。在本研究中,我们研究了 TIM-1 等表面受体在受感染细胞表面封存新萌发病毒的能力。我们建立了一个发光报告系统,以产生整合了纳米荧光素酶的基孔肯雅病毒颗粒,并轻松地量化病毒颗粒。我们发现,与其他进入因子相比,宿主细胞表面的 TIM-1 能显著降低 CHIKV 的释放效率。通过直接敲除细胞或改变细胞脂质分布来去除细胞表面的 TIM-1,可以提高 CHIKV 的释放效率。在感染过程中,CHIKV 能够通过在非结构蛋白 2 的介导下逐渐降低 TIM-1 的表面水平来抵消系留效应。 这项研究强调了磷脂酰丝氨酸受体在介导 CHIKV 进入和释放方面的重要性,并有助于开发能够增强疫苗生产的细胞系:基孔肯雅病毒(CHIKV)是一种包膜α病毒,通过有传染性的蚊子叮咬传播。感染基孔肯雅病毒后会出现发热、关节疼痛和关节痛,感染后可转为慢性并持续数月。这种疾病的预防仍然高度集中于病媒控制策略。2023 年 12 月,美国食品和药物管理局批准了一种新型 CHIKV 减毒活疫苗。我们的目标是研究参与CHIKV释放的细胞因素,以更好地了解CHIKV在各种细胞系中高效感染和传播的能力。我们发现,TIM-1 受体能显著削弱 CHIKV 有效排出感染细胞的能力。这些信息有助于在实验室环境和疫苗生产过程中最大限度地提高病毒粒子的产量。
{"title":"Chikungunya virus release is reduced by TIM-1 receptors through binding of envelope phosphatidylserine.","authors":"Judith M Reyes Ballista, Ashley J Hoover, Joseph T Noble, Marissa D Acciani, Kerri L Miazgowicz, Sarah A Harrison, Grace Andrea L Tabscott, Avery Duncan, Don N Barnes, Ariana R Jimenez, Melinda A Brindley","doi":"10.1128/jvi.00775-24","DOIUrl":"10.1128/jvi.00775-24","url":null,"abstract":"<p><p>T-cell immunoglobin and mucin domain protein-1 (TIM-1) mediates entry of chikungunya virus (CHIKV) into some mammalian cells through the interaction with envelope phospholipids. While this interaction enhances entry, TIM-1 has been shown to tether newly formed HIV and Ebola virus particles, limiting their efficient release. In this study, we investigate the ability of surface receptors such as TIM-1 to sequester newly budded virions on the surface of infected cells. We established a luminescence reporter system to produce chikungunya viral particles that integrate nano-luciferase and easily quantify viral particles. We found that TIM-1 on the surface of host cells significantly reduced CHIKV release efficiency in comparison to other entry factors. Removal of cell surface TIM-1 through direct cellular knock-out or altering the cellular lipid distribution enhanced CHIKV release. Over the course of infection, CHIKV was able to counteract the tethering effect by gradually decreasing the surface levels of TIM-1 in a process mediated by the nonstructural protein 2. This study highlights the importance of phosphatidylserine receptors in mediating not only the entry of CHIKV but also its release and could aid in developing cell lines capable of enhanced vaccine production.</p><p><strong>Importance: </strong>Chikungunya virus (CHIKV) is an enveloped alphavirus transmitted by the bites of infectious mosquitoes. Infection with CHIKV results in the development of fever, joint pain, and arthralgia that can become chronic and last for months after infection. Prevention of this disease is still highly focused on vector control strategies. In December 2023, a new live attenuated vaccine against CHIKV was approved by the FDA. We aimed to study the cellular factors involved in CHIKV release, to better understand CHIKV's ability to efficiently infect and spread among a wide variety of cell lines. We found that TIM-1 receptors can significantly abrogate CHIKV's ability to efficiently exit infected cells. This information can be beneficial for maximizing viral particle production in laboratory settings and during vaccine manufacturing.</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/PMC11334481/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141616750","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
In vitro higher-order oligomeric assembly of the respiratory syncytial virus M2-1 protein with longer RNAs. 呼吸道合胞病毒 M2-1 蛋白与长 RNA 的体外高阶寡聚组装。
IF 4 2区 医学 Q2 VIROLOGY Pub Date : 2024-08-20 Epub Date: 2024-07-17 DOI: 10.1128/jvi.01046-24
Yunrong Gao, Anirudh Raghavan, Sara Andrea Espinosa Garcia, Bowei Deng, Diego Hurtado de Mendoza, Bo Liang

The respiratory syncytial virus (RSV) M2-1 protein is a transcriptional antitermination factor crucial for efficiently synthesizing multiple full-length viral mRNAs. During RSV infection, M2-1 exists in a complex with mRNA within cytoplasmic compartments called inclusion body-associated granules (IBAGs). Prior studies showed that M2-1 can bind along the entire length of viral mRNAs instead of just gene-end (GE) sequences, suggesting that M2-1 has more sophisticated RNA recognition and binding characteristics. Here, we analyzed the higher oligomeric complexes formed by M2-1 and RNAs in vitro using size exclusion chromatography (SEC), electrophoretic mobility shift assays (EMSA), negative stain electron microscopy (EM), and mutagenesis. We observed that the minimal RNA length for such higher oligomeric assembly is about 14 nucleotides for polyadenine sequences, and longer RNAs exhibit distinct RNA-induced binding modality to M2-1, leading to enhanced particle formation frequency and particle homogeneity as the local RNA concentration increases. We showed that particular cysteine residues of the M2-1 cysteine-cysteine-cystine-histidine (CCCH) zinc-binding motif are essential for higher oligomeric assembly. Furthermore, complexes assembled with long polyadenine sequences remain unaffected when co-incubated with ribonucleases or a zinc chelation agent. Our study provided new insights into the higher oligomeric assembly of M2-1 with longer RNA.IMPORTANCERespiratory syncytial virus (RSV) causes significant respiratory infections in infants, the elderly, and immunocompromised individuals. The virus forms specialized compartments to produce genetic material, with the M2-1 protein playing a pivotal role. M2-1 acts as an anti-terminator in viral transcription, ensuring the creation of complete viral mRNA and associating with both viral and cellular mRNA. Our research focuses on understanding M2-1's function in viral mRNA synthesis by modeling interactions in a controlled environment. This approach is crucial due to the challenges of studying these compartments in vivo. Reconstructing the system in vitro uncovers structural and biochemical aspects and reveals the potential functions of M2-1 and its homologs in related viruses. Our work may contribute to identifying targets for antiviral inhibitors and advancing RSV infection treatment.

呼吸道合胞病毒(RSV)M2-1 蛋白是一种转录反终止因子,对有效合成多种全长病毒 mRNA 至关重要。在 RSV 感染过程中,M2-1 与 mRNA 复合存在于称为包涵体相关颗粒(IBAGs)的细胞质区室中。之前的研究表明,M2-1 能与病毒 mRNA 的整个长度而不仅仅是基因末端(GE)序列结合,这表明 M2-1 具有更复杂的 RNA 识别和结合特性。在这里,我们使用尺寸排阻色谱(SEC)、电泳迁移试验(EMSA)、负染色电子显微镜(EM)和诱变等方法分析了 M2-1 与 RNA 在体外形成的高寡聚复合物。我们观察到,对于多腺嘌呤序列来说,这种较高的寡聚体组装所需的最小 RNA 长度约为 14 个核苷酸,而较长的 RNA 会表现出与 M2-1 不同的 RNA 诱导结合模式,从而随着局部 RNA 浓度的增加而提高粒子形成频率和粒子均匀性。我们的研究表明,M2-1半胱氨酸-半胱氨酸-胱氨酸-组氨酸(CCCH)锌结合基团的特定半胱氨酸残基对于更高的寡聚体组装至关重要。此外,当与核糖核酸酶或锌螯合剂共孵育时,与长多聚腺嘌呤序列组装的复合物不受影响。重要意义呼吸道合胞病毒(RSV)会导致婴儿、老人和免疫力低下的人发生严重的呼吸道感染。病毒形成专门的分区以产生遗传物质,其中 M2-1 蛋白起着关键作用。M2-1 在病毒转录过程中充当反终止子,确保产生完整的病毒 mRNA,并与病毒和细胞 mRNA 结合。我们的研究重点是通过模拟受控环境中的相互作用,了解 M2-1 在病毒 mRNA 合成中的功能。由于在体内研究这些区室所面临的挑战,这种方法至关重要。体外系统的重建揭示了结构和生化方面的问题,并揭示了 M2-1 及其同源物在相关病毒中的潜在功能。我们的工作可能有助于确定抗病毒抑制剂的靶点,并推进 RSV 感染的治疗。
{"title":"<i>In vitro</i> higher-order oligomeric assembly of the respiratory syncytial virus M2-1 protein with longer RNAs.","authors":"Yunrong Gao, Anirudh Raghavan, Sara Andrea Espinosa Garcia, Bowei Deng, Diego Hurtado de Mendoza, Bo Liang","doi":"10.1128/jvi.01046-24","DOIUrl":"10.1128/jvi.01046-24","url":null,"abstract":"<p><p>The respiratory syncytial virus (RSV) M2-1 protein is a transcriptional antitermination factor crucial for efficiently synthesizing multiple full-length viral mRNAs. During RSV infection, M2-1 exists in a complex with mRNA within cytoplasmic compartments called inclusion body-associated granules (IBAGs). Prior studies showed that M2-1 can bind along the entire length of viral mRNAs instead of just gene-end (GE) sequences, suggesting that M2-1 has more sophisticated RNA recognition and binding characteristics. Here, we analyzed the higher oligomeric complexes formed by M2-1 and RNAs <i>in vitro</i> using size exclusion chromatography (SEC), electrophoretic mobility shift assays (EMSA), negative stain electron microscopy (EM), and mutagenesis. We observed that the minimal RNA length for such higher oligomeric assembly is about 14 nucleotides for polyadenine sequences, and longer RNAs exhibit distinct RNA-induced binding modality to M2-1, leading to enhanced particle formation frequency and particle homogeneity as the local RNA concentration increases. We showed that particular cysteine residues of the M2-1 cysteine-cysteine-cystine-histidine (CCCH) zinc-binding motif are essential for higher oligomeric assembly. Furthermore, complexes assembled with long polyadenine sequences remain unaffected when co-incubated with ribonucleases or a zinc chelation agent. Our study provided new insights into the higher oligomeric assembly of M2-1 with longer RNA.IMPORTANCERespiratory syncytial virus (RSV) causes significant respiratory infections in infants, the elderly, and immunocompromised individuals. The virus forms specialized compartments to produce genetic material, with the M2-1 protein playing a pivotal role. M2-1 acts as an anti-terminator in viral transcription, ensuring the creation of complete viral mRNA and associating with both viral and cellular mRNA. Our research focuses on understanding M2-1's function in viral mRNA synthesis by modeling interactions in a controlled environment. This approach is crucial due to the challenges of studying these compartments <i>in vivo</i>. Reconstructing the system <i>in vitro</i> uncovers structural and biochemical aspects and reveals the potential functions of M2-1 and its homologs in related viruses. Our work may contribute to identifying targets for antiviral inhibitors and advancing RSV infection treatment.</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/PMC11334520/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141627112","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
AcMNPV-miR-2 affects Autographa californica nucleopolyhedrovirus infection by regulating the expression of ac28 and several other viral early genes. AcMNPV-miR-2通过调控ac28和其他几个病毒早期基因的表达来影响加利福尼亚核多角体病毒的感染。
IF 4 2区 医学 Q2 VIROLOGY Pub Date : 2024-08-20 Epub Date: 2024-07-18 DOI: 10.1128/jvi.00570-24
Xinghua Yu, Tingkai Teng, Zhuowen Duan, Jinwen Wang

Virus-encoded microRNAs (miRNAs) exert diverse regulatory roles in the biological processes of both viruses and hosts. This study delves into the functions of AcMNPV-miR-2, an early miRNA encoded by Autographa californica multiple nucleopolyhedrovirus (AcMNPV). AcMNPV-miR-2 targets viral early genes ac28 (lef-6), ac37 (lef-11), ac49, and ac63. Overexpression of AcMNPV-miR-2 leads to reduced production of infectious budded virions (BVs) and diminished viral DNA replication. Delayed polyhedron formation was observed through light and transmission electron microscopy, and the larval lifespan extended in oral infection assays. Moreover, the mRNA expression levels of two Lepidoptera-specific immune-related proteins, Gloverin and Spod-11-tox, significantly decreased. These findings indicate that AcMNPV-miR-2 restrains viral load, reducing host immune sensitivity. This beneficial effect enables the virus to combat host defense mechanisms and reside within the host for an extended duration.

Importance: Virus-encoded miRNAs have been extensively studied for their pivotal roles in finetuning viral infections. Baculoviruses, highly pathogenic in insects, remain underexplored concerning their encoded miRNAs. Previous reports outlined three AcMNPV-encoded miRNAs, AcMNPV-miR-1, -miR-3, and -miR-4. This study delves into the functions of another AcMNPV-encoded miRNA, AcMNPV-miR-2 (Ac-miR-2). Through a comprehensive analysis of target gene expression, the impact on larvae, and variations in host immune-related gene expression, we elucidate a functional pathway for Ac-miR-2. This miRNA suppresses viral load and infectivity and prolongs lifespans of infected larva by downregulating specific viral early genes and host immune-related genes. These mechanisms ultimately serve the virus's primary goal of enhanced propagation. Our study significantly contributes to understanding of the intricate regulatory mechanisms of virus-encoded miRNAs in baculovirus infections.

病毒编码的微RNA(miRNA)在病毒和宿主的生物过程中发挥着多种调控作用。本研究深入探讨了AcMNPV-miR-2的功能,这是一种由Autographa californica multiple nucleopolyhedrovirus(AcMNPV)编码的早期miRNA。AcMNPV-miR-2靶向病毒早期基因ac28(lef-6)、ac37(lef-11)、ac49和ac63。过量表达 AcMNPV-miR-2 会导致感染性芽生病毒(BV)产生减少,病毒 DNA 复制减弱。光镜和透射电子显微镜观察到多面体形成延迟,口腔感染试验中幼虫寿命延长。此外,两种鳞翅目特异性免疫相关蛋白 Gloverin 和 Spod-11-tox 的 mRNA 表达水平显著下降。这些发现表明,AcMNPV-miR-2 能抑制病毒载量,降低宿主免疫敏感性。这种有利影响使病毒能够对抗宿主防御机制,并在宿主体内长期存在:病毒编码的 miRNA 因其在微调病毒感染中的关键作用而被广泛研究。杆状病毒是昆虫中的高致病性病毒,但对其编码的 miRNA 的研究仍然不足。以前的报告概述了三种 AcMNPV 编码的 miRNA,即 AcMNPV-miR-1、-miR-3 和 -miR-4。本研究深入研究了另一种 AcMNPV 编码的 miRNA--AcMNPV-miR-2(Ac-miR-2)的功能。通过全面分析靶基因表达、对幼虫的影响以及宿主免疫相关基因表达的变化,我们阐明了 Ac-miR-2 的功能途径。这种 miRNA 通过下调特定的病毒早期基因和宿主免疫相关基因,抑制病毒载量和感染性,延长受感染幼虫的寿命。这些机制最终服务于病毒的主要目标--增强繁殖。我们的研究大大有助于人们了解病毒编码的 miRNA 在杆状病毒感染中的复杂调控机制。
{"title":"AcMNPV-miR-2 affects <i>Autographa californica</i> nucleopolyhedrovirus infection by regulating the expression of <i>ac28</i> and several other viral early genes.","authors":"Xinghua Yu, Tingkai Teng, Zhuowen Duan, Jinwen Wang","doi":"10.1128/jvi.00570-24","DOIUrl":"10.1128/jvi.00570-24","url":null,"abstract":"<p><p>Virus-encoded microRNAs (miRNAs) exert diverse regulatory roles in the biological processes of both viruses and hosts. This study delves into the functions of AcMNPV-miR-2, an early miRNA encoded by <i>Autographa californica</i> multiple nucleopolyhedrovirus (AcMNPV). AcMNPV-miR-2 targets viral early genes <i>ac28</i> (<i>lef-6</i>), <i>ac37</i> (<i>lef-11</i>), <i>ac49</i>, and <i>ac63</i>. Overexpression of AcMNPV-miR-2 leads to reduced production of infectious budded virions (BVs) and diminished viral DNA replication. Delayed polyhedron formation was observed through light and transmission electron microscopy, and the larval lifespan extended in oral infection assays. Moreover, the mRNA expression levels of two Lepidoptera-specific immune-related proteins, Gloverin and Spod-11-tox, significantly decreased. These findings indicate that AcMNPV-miR-2 restrains viral load, reducing host immune sensitivity. This beneficial effect enables the virus to combat host defense mechanisms and reside within the host for an extended duration.</p><p><strong>Importance: </strong>Virus-encoded miRNAs have been extensively studied for their pivotal roles in finetuning viral infections. Baculoviruses, highly pathogenic in insects, remain underexplored concerning their encoded miRNAs. Previous reports outlined three AcMNPV-encoded miRNAs, AcMNPV-miR-1, -miR-3, and -miR-4. This study delves into the functions of another AcMNPV-encoded miRNA, AcMNPV-miR-2 (Ac-miR-2). Through a comprehensive analysis of target gene expression, the impact on larvae, and variations in host immune-related gene expression, we elucidate a functional pathway for Ac-miR-2. This miRNA suppresses viral load and infectivity and prolongs lifespans of infected larva by downregulating specific viral early genes and host immune-related genes. These mechanisms ultimately serve the virus's primary goal of enhanced propagation. Our study significantly contributes to understanding of the intricate regulatory mechanisms of virus-encoded miRNAs in baculovirus infections.</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/PMC11334470/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141633823","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
Japanese encephalitis virus NS5 protein interacts with nucleolin to enhance the virus replication. 日本脑炎病毒 NS5 蛋白与核蛋白相互作用,促进病毒复制。
IF 4 2区 医学 Q2 VIROLOGY Pub Date : 2024-08-20 Epub Date: 2024-07-30 DOI: 10.1128/jvi.00858-24
Arundhati Deb, Shilpi Nagpal, Rajnesh Kumari Yadav, Harsh Thakur, Deepak Nair, Vengadesan Krishnan, Sudhanshu Vrati

Japanese encephalitis virus (JEV) is an arthropod-borne, plus-strand flavivirus causing viral encephalitis in humans with a high case fatality rate. The JEV non-structural protein 5 (NS5) with the RNA-dependent RNA polymerase activity interacts with the viral and host proteins to constitute the replication complex. We have identified the multifunctional protein Nucleolin (NCL) as one of the several NS5-interacting host proteins. We demonstrate the interaction and colocalization of JEV NS5 with NCL in the virus-infected HeLa cells. The siRNA-mediated knockdown of NCL indicated that it was required for efficient viral replication. Importantly, JEV grew to higher titers in cells over-expressing exogenous NCL, demonstrating its pro-viral role. We demonstrated that NS5 interacted with the RRM and GAR domains of NCL. We show that the NCL-binding aptamer AS1411 containing the G-quadruplex (GQ) structure and the GQ ligand BRACO-19 caused significant inhibition of JEV replication. The antiviral effect of AS1411 and BRACO-19 could be overcome in HeLa cells by the overexpression of exogenous NCL. We demonstrated that the synthetic RNAs derived from the 3'-NCR of JEV genomic RNA containing the GQ sequence could bind NCL in vitro. The replication complex binding to the 3'-NCR is required for the viral RNA synthesis. It is likely that NCL present in the replication complex destabilizes the GQ structures in the genomic RNA, thus facilitating the movement of the replication complex resulting in efficient virus replication.IMPORTANCEJapanese encephalitis virus (JEV) is endemic in most parts of South-East Asia and the Western Pacific region, causing epidemics of encephalitis with a high case fatality rate. While a tissue culture-derived JEV vaccine is available, no antiviral therapy exists. The JEV NS5 protein has RNA-dependent RNA polymerase activity. Together with several host and viral proteins, it constitutes the replication complex necessary for virus replication. Understanding the interaction of NS5 with the host proteins could help design novel antivirals. We identified Nucleolin (NCL) as a crucial host protein interactor of JEV NS5 having a pro-viral role in virus replication. The NS5-interacting NCL binds to the G-quadruplex (GQ) structure sequence in the 3'-NCR of JEV RNA. This may smoothen the movement of the replication complex along the genomic RNA, thereby facilitating the virus replication. This study is the first report on how NCL, a host protein, helps in JEV replication through GQ-binding.

日本脑炎病毒(JEV)是一种节肢动物传播的加链黄病毒,可导致人类病毒性脑炎,病死率很高。具有 RNA 依赖性 RNA 聚合酶活性的 JEV 非结构蛋白 5(NS5)与病毒蛋白和宿主蛋白相互作用,构成复制复合物。我们发现多功能蛋白 Nucleolin (NCL) 是与 NS5 相互作用的几种宿主蛋白之一。我们证明了病毒感染的 HeLa 细胞中 JEV NS5 与 NCL 的相互作用和共定位。siRNA 介导的 NCL 敲除表明,病毒的有效复制需要 NCL。重要的是,JEV在过度表达外源NCL的细胞中生长出更高的滴度,这证明了NCL的促病毒作用。我们证明了 NS5 与 NCL 的 RRM 和 GAR 结构域相互作用。我们的研究表明,含有 G-quadruplex (GQ) 结构的 NCL 结合适配体 AS1411 和 GQ 配体 BRACO-19 能显著抑制 JEV 的复制。在 HeLa 细胞中,外源 NCL 的过表达可以克服 AS1411 和 BRACO-19 的抗病毒作用。我们证实,从含有 GQ 序列的 JEV 基因组 RNA 的 3'-NCR 提取的合成 RNA 可在体外与 NCL 结合。病毒 RNA 合成需要与 3'-NCR 结合的复制复合物。重要意义日本脑炎病毒(JEV)在东南亚和西太平洋地区的大部分地方流行,引起脑炎流行,病死率很高。虽然有组织培养的 JEV 疫苗,但目前还没有抗病毒疗法。JEV NS5 蛋白具有 RNA 依赖性 RNA 聚合酶活性。它与几种宿主蛋白和病毒蛋白一起构成了病毒复制所需的复制复合物。了解 NS5 与宿主蛋白的相互作用有助于设计新型抗病毒药物。我们发现 Nucleolin(NCL)是 JEV NS5 的一个重要宿主蛋白相互作用体,在病毒复制过程中具有促进病毒复制的作用。与 NS5 相互作用的 NCL 与 JEV RNA 的 3'-NCR 中的 G-quadruplex (GQ) 结构序列结合。这可能会使复制复合物沿着基因组 RNA 平滑移动,从而促进病毒复制。这项研究首次报道了宿主蛋白 NCL 如何通过 GQ 结合帮助 JEV 复制。
{"title":"Japanese encephalitis virus NS5 protein interacts with nucleolin to enhance the virus replication.","authors":"Arundhati Deb, Shilpi Nagpal, Rajnesh Kumari Yadav, Harsh Thakur, Deepak Nair, Vengadesan Krishnan, Sudhanshu Vrati","doi":"10.1128/jvi.00858-24","DOIUrl":"10.1128/jvi.00858-24","url":null,"abstract":"<p><p>Japanese encephalitis virus (JEV) is an arthropod-borne, plus-strand flavivirus causing viral encephalitis in humans with a high case fatality rate. The JEV non-structural protein 5 (NS5) with the RNA-dependent RNA polymerase activity interacts with the viral and host proteins to constitute the replication complex. We have identified the multifunctional protein Nucleolin (NCL) as one of the several NS5-interacting host proteins. We demonstrate the interaction and colocalization of JEV NS5 with NCL in the virus-infected HeLa cells. The siRNA-mediated knockdown of NCL indicated that it was required for efficient viral replication. Importantly, JEV grew to higher titers in cells over-expressing exogenous NCL, demonstrating its pro-viral role. We demonstrated that NS5 interacted with the RRM and GAR domains of NCL. We show that the NCL-binding aptamer AS1411 containing the G-quadruplex (GQ) structure and the GQ ligand BRACO-19 caused significant inhibition of JEV replication. The antiviral effect of AS1411 and BRACO-19 could be overcome in HeLa cells by the overexpression of exogenous NCL. We demonstrated that the synthetic RNAs derived from the 3'-NCR of JEV genomic RNA containing the GQ sequence could bind NCL <i>in vitro</i>. The replication complex binding to the 3'-NCR is required for the viral RNA synthesis. It is likely that NCL present in the replication complex destabilizes the GQ structures in the genomic RNA, thus facilitating the movement of the replication complex resulting in efficient virus replication.IMPORTANCEJapanese encephalitis virus (JEV) is endemic in most parts of South-East Asia and the Western Pacific region, causing epidemics of encephalitis with a high case fatality rate. While a tissue culture-derived JEV vaccine is available, no antiviral therapy exists. The JEV NS5 protein has RNA-dependent RNA polymerase activity. Together with several host and viral proteins, it constitutes the replication complex necessary for virus replication. Understanding the interaction of NS5 with the host proteins could help design novel antivirals. We identified Nucleolin (NCL) as a crucial host protein interactor of JEV NS5 having a pro-viral role in virus replication. The NS5-interacting NCL binds to the G-quadruplex (GQ) structure sequence in the 3'-NCR of JEV RNA. This may smoothen the movement of the replication complex along the genomic RNA, thereby facilitating the virus replication. This study is the first report on how NCL, a host protein, helps in JEV replication through GQ-binding.</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/PMC11334521/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141792830","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
Delayed treatment of cynomolgus macaques with a FVM04/CA45 monoclonal antibody cocktail provides complete protection against lethal Sudan virus infection. 用 FVM04/CA45 单克隆鸡尾酒抗体延迟治疗猕猴,可完全防止致命性苏丹病毒感染。
IF 4 2区 医学 Q2 VIROLOGY Pub Date : 2024-08-20 Epub Date: 2024-07-16 DOI: 10.1128/jvi.01242-23
Mable Chan, Bryce M Warner, Jonathan Audet, Douglas Barker, Nikesh Tailor, Robert Vendramelli, Thang Truong, Kevin Tierney, Amrit S Boese, Honguy Qiu, Frederick W Holtsberg, Javad Aman, Shantha Kodihalli, Darwyn Kobasa

Sudan ebolavirus (SUDV) is a member of the genus Ebolavirus (Family Filoviridae) and has caused sporadic outbreaks of Ebola disease (EBOD), or more specifically Sudan virus disease (SVD), with high mortality rates in Africa. Current vaccines and therapies that have been developed for filoviruses are almost all specific for Ebola virus (EBOV; of the species Zaire ebolavirus), and there is a current lack of therapeutics specific for SUDV. The recent SUDV outbreak in Uganda, which was distributed across multiple districts, including Kampala, a densely populated urban center, highlights the critical need for the development of novel SUDV-specific or pan-Ebola virus therapeutics. Previous work has characterized two monoclonal antibodies, FVM04 and CA45, which have neutralization capabilities against both EBOV and SUDV and have shown protective efficacy in animal challenge studies. Here, we expand upon this work, showing that treatment with a monoclonal antibody cocktail consisting of FVM04 and CA45 provides full protection against lethal SUDV infection in cynomolgus macaques. Studies that evaluate outcomes at late time points after infection, once clinical signs of illness are apparent, are vital for assessing the therapeutic efficacy of antibody therapeutics. We have shown that when treatment is initiated as late as 5 days after infection, with a second dose given on day 8, that treated groups showed few clinical signs or morbidity, with complete survival. This work provides further evidence that FVM04 and CA45 have strong therapeutic potential against SUDV and their development as a pan-Ebola virus therapeutic should be pursued.

Importance: There are currently no approved vaccines or therapeutics for Sudan virus, a filovirus which is highly related to Ebola virus and causes similar disease and outbreaks. In this study, a cocktail of two potent monoclonal antibodies that effectively neutralize Sudan virus was tested in a nonhuman primate model of Sudan virus disease. Treatment was highly effective, even when initiated as late as 5 days after infection, when clinical signs of infection were already evident. All treated animals showed complete recovery from infection, with little evidence of disease, while all animals that received a control treatment succumbed to infection within 8 days. The study further demonstrated the strong therapeutic potential of the antibody treatment and supported further development for use in Sudan virus outbreaks.

苏丹埃博拉病毒(SUDV)是埃博拉病毒属(丝状病毒科)的一员,曾在非洲引起埃博拉病(EBOD)或更确切地说是苏丹病毒病(SVD)的零星爆发,死亡率很高。目前针对丝状病毒开发的疫苗和疗法几乎都是针对埃博拉病毒(EBOV,属于扎伊尔伊波拉病毒)的,而目前还缺乏针对苏丹病毒的特效疗法。乌干达最近爆发的 SUDV 疫情遍及多个地区,包括人口稠密的城市中心坎帕拉,这凸显了开发新型 SUDV 特异性或泛埃博拉病毒疗法的迫切需要。此前的研究已经确定了两种单克隆抗体(FVM04 和 CA45)的特性,这两种抗体具有中和 EBOV 和 SUDV 的能力,并在动物挑战研究中显示出保护效力。在此,我们对这一研究成果进行了进一步扩展,结果表明,使用由 FVM04 和 CA45 组成的单克隆抗体鸡尾酒治疗可为猕猴提供全面保护,使其免受致命性 SUDV 感染。在感染后的晚期,即疾病的临床症状显现后,对结果进行评估的研究对于评估抗体疗法的疗效至关重要。我们的研究表明,在感染后 5 天开始治疗,并在第 8 天注射第二剂时,治疗组几乎没有出现临床症状或发病,而且完全存活。这项工作进一步证明,FVM04 和 CA45 对 SUDV 具有强大的治疗潜力,应将其开发为泛埃博拉病毒疗法:苏丹病毒是一种丝状病毒,与埃博拉病毒有很高的亲缘关系,会引起类似的疾病和疫情爆发。本研究在苏丹病毒病的非人灵长类动物模型中测试了能有效中和苏丹病毒的两种强效单克隆抗体鸡尾酒。即使在感染后 5 天,即感染的临床症状已经很明显时才开始治疗,治疗效果也很好。所有接受治疗的动物都从感染中完全恢复,几乎没有发病迹象,而所有接受对照组治疗的动物都在 8 天内因感染而死亡。这项研究进一步证明了抗体疗法的强大治疗潜力,并支持进一步开发用于苏丹病毒爆发的抗体疗法。
{"title":"Delayed treatment of cynomolgus macaques with a FVM04/CA45 monoclonal antibody cocktail provides complete protection against lethal Sudan virus infection.","authors":"Mable Chan, Bryce M Warner, Jonathan Audet, Douglas Barker, Nikesh Tailor, Robert Vendramelli, Thang Truong, Kevin Tierney, Amrit S Boese, Honguy Qiu, Frederick W Holtsberg, Javad Aman, Shantha Kodihalli, Darwyn Kobasa","doi":"10.1128/jvi.01242-23","DOIUrl":"10.1128/jvi.01242-23","url":null,"abstract":"<p><p>Sudan ebolavirus (SUDV) is a member of the genus <i>Ebolavirus</i> (Family <i>Filoviridae</i>) and has caused sporadic outbreaks of Ebola disease (EBOD), or more specifically Sudan virus disease (SVD), with high mortality rates in Africa. Current vaccines and therapies that have been developed for filoviruses are almost all specific for Ebola virus (EBOV; of the species <i>Zaire ebolavirus</i>), and there is a current lack of therapeutics specific for SUDV. The recent SUDV outbreak in Uganda, which was distributed across multiple districts, including Kampala, a densely populated urban center, highlights the critical need for the development of novel SUDV-specific or pan-Ebola virus therapeutics. Previous work has characterized two monoclonal antibodies, FVM04 and CA45, which have neutralization capabilities against both EBOV and SUDV and have shown protective efficacy in animal challenge studies. Here, we expand upon this work, showing that treatment with a monoclonal antibody cocktail consisting of FVM04 and CA45 provides full protection against lethal SUDV infection in cynomolgus macaques. Studies that evaluate outcomes at late time points after infection, once clinical signs of illness are apparent, are vital for assessing the therapeutic efficacy of antibody therapeutics. We have shown that when treatment is initiated as late as 5 days after infection, with a second dose given on day 8, that treated groups showed few clinical signs or morbidity, with complete survival. This work provides further evidence that FVM04 and CA45 have strong therapeutic potential against SUDV and their development as a pan-Ebola virus therapeutic should be pursued.</p><p><strong>Importance: </strong>There are currently no approved vaccines or therapeutics for Sudan virus, a filovirus which is highly related to Ebola virus and causes similar disease and outbreaks. In this study, a cocktail of two potent monoclonal antibodies that effectively neutralize Sudan virus was tested in a nonhuman primate model of Sudan virus disease. Treatment was highly effective, even when initiated as late as 5 days after infection, when clinical signs of infection were already evident. All treated animals showed complete recovery from infection, with little evidence of disease, while all animals that received a control treatment succumbed to infection within 8 days. The study further demonstrated the strong therapeutic potential of the antibody treatment and supported further development for use in Sudan virus outbreaks.</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/PMC11334508/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141620281","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