1
|
Prasad BVV, Atmar RL, Ramani S, Palzkill T, Song Y, Crawford SE, Estes MK. Norovirus replication, host interactions and vaccine advances. Nat Rev Microbiol 2025; 23:385-401. [PMID: 39824927 DOI: 10.1038/s41579-024-01144-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/12/2024] [Indexed: 01/20/2025]
Abstract
Human noroviruses (HuNoVs) are the leading cause of acute gastroenteritis worldwide in all age groups and cause significant disease and economic burden globally. To date, no approved vaccines or antiviral therapies are available to treat or prevent HuNoV illness. Several candidate vaccines are in clinical trials, although potential barriers to successful development must be overcome. Recently, significant advances have been made in understanding HuNoV biology owing to breakthroughs in virus cultivation using human intestinal tissue-derived organoid (or enteroid) cultures, advances in structural biology technology combined with epitope mapping and increased metagenomic sequencing. New and unexpected strain-specific differences in pandemic versus non-pandemic virus structures, replication properties and virus-host interactions, including host factors required for susceptibility to infection and pathogenesis, are discussed.
Collapse
Affiliation(s)
- B V Venkataram Prasad
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Robert L Atmar
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Sasirekha Ramani
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Timothy Palzkill
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Yongcheng Song
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, TX, USA
| | - Sue E Crawford
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Mary K Estes
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA.
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
2
|
Asif S, Yingkun D, Meng C. Unlocking the secrets of Feline calicivirus: advances in structural and nonstructural proteins and its role as a key model for other Caliciviruses. Virol J 2025; 22:152. [PMID: 40399981 PMCID: PMC12096530 DOI: 10.1186/s12985-025-02750-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2025] [Accepted: 04/21/2025] [Indexed: 05/23/2025] Open
Abstract
Feline calicivirus (FCV) is a highly contagious pathogen responsible for respiratory infections, lingual ulceration, oral ulcers and systemic diseases in cats, posing a significant risk to feline family worldwide. Virus enters via nasal oral and conjunctival routes. Oropharynx is primary site of replication, induces epithelial necrosis. After recovery from acute disease most cats clear virus within 30 days. Some lifelong carriers via colonization of tonsillar and other tissues. Understanding the structural and nonstructural proteins of FCV is essential to know viral replication process, its pathogenesis and interaction with host immune system. This manuscript outlines the recent progress made on the characterization of FCV proteins with respect to their involvement in viral assembly, entry, immune evasion, and replication. Although structural proteins such as capsid have received most attention regarding viral attachment and host specificity, but nonstructural proteins are emerging as key players in influencing host cell activities and viral RNA synthesis. This review highlights the requirement for advanced structural research methods, large-scale antiviral screening, and thorough investigations into FCV-host interactions. These studies will not only enable us fully understand FCV, but also promote the progress of more universally applicable virological research and drug development.
Collapse
Affiliation(s)
- Sana Asif
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 518 Ziyue Road, Shanghai, 200241, P. R. China
| | - Deng Yingkun
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 518 Ziyue Road, Shanghai, 200241, P. R. China
| | - Chunchun Meng
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 518 Ziyue Road, Shanghai, 200241, P. R. China.
| |
Collapse
|
3
|
Sherman MB, Smith HQ, Cox F, Wobus CE, Lynch GC, Pettitt BM, Smith TJ. Murine norovirus allosteric escape mutants mimic gut activation. J Virol 2025:e0021925. [PMID: 40353669 DOI: 10.1128/jvi.00219-25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Accepted: 04/14/2025] [Indexed: 05/14/2025] Open
Abstract
Murine norovirus (MNV) undergoes large conformational changes in response to the environment. The T=3 icosahedral capsid is composed of 180 copies of ~58 kDa VP1 that has N-terminal (N), shell (S), and C-terminal protruding (P) domains. In phosphate-buffered saline, the P domains are loosely tethered to the shell and float ~15 Å above the surface. At conditions found in the gut (i.e., low pH with high metal ion and bile salt concentrations), the P domain rotates and drops onto the shell with intra P domain changes that enhance receptor interactions while blocking antibody binding. Two of our monoclonal antibodies (2D3 and 4F9) have broad strain recognition, and the only escape mutants, V339I and D348E, are located on the C'D' loop and ~20 Å from the epitope. Here, we determined the cryo-EM structures of V339I and D348E at neutral pH +/-metal ions and bile salts. These allosteric escape mutants have the activated conformation in the absence of gut triggers. Since this conformation is not recognized by antibodies, it explains how these mutants evade antibody recognition. Dynamic simulations of the P domain further suggest that movement of the C'D' loop may be the rate-limiting step in the conformational change and that V339I increases the motion of the A'B'/E'F' loops compared to the wild-type (WT), facilitating the transition to the activated state. These findings have important implications for norovirus vaccine design since they uncover a form of the viral capsid that should lend superior immune protection against subsequent challenge by wild-type virus.IMPORTANCEImmune protection from norovirus infection is notoriously transient in both humans and mice. Our results strongly suggest that this is likely because the "activated" form of the virus found in gut conditions is not recognized by antibodies created in the circulation. By reversibly presenting one structure in the gut and a completely different antigenic structure in circulation, the gut tissue can be infected in subsequent challenges, while extraintestinal organs are protected. We find here that allosteric escape mutants to the most broadly neutralizing antibodies thwart recognition by transitioning to the activated state without the need for gut triggers (i.e., bile, low pH, or metal ions). These findings are significant because it is now feasible to present the activated form of the virus to the immune system (for example, as a vaccine) to better protect the gut tissue for longer periods of time.
Collapse
Affiliation(s)
- Michael B Sherman
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch at Galveston, Galveston, Texas, USA
| | - Hong Q Smith
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch at Galveston, Galveston, Texas, USA
| | - Faith Cox
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch at Galveston, Galveston, Texas, USA
| | - Christiane E Wobus
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Gillian C Lynch
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch at Galveston, Galveston, Texas, USA
| | - B Montgomery Pettitt
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch at Galveston, Galveston, Texas, USA
| | - Thomas J Smith
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch at Galveston, Galveston, Texas, USA
| |
Collapse
|
4
|
Podadera A, Leuthold M, Martín-Alonso JM, Casais R, Álvarez AL, Lobo-Castañón MJ, Parra F, Dalton KP. Epitope mapping of a neutralizing antibody against rabbit hemorrhagic disease virus GI.2. Vet Res 2025; 56:74. [PMID: 40176146 PMCID: PMC11963670 DOI: 10.1186/s13567-025-01505-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 02/05/2025] [Indexed: 04/04/2025] Open
Abstract
In 2010, rabbit hemorrhagic disease virus (RHDV) GI.2 emerged, and unlike RHDV GI.1, it caused mortality in young rabbits, while existing vaccines were not fully protective. The GI.2-specific monoclonal antibody (mAb) 2D9 has been used as a tool to discriminate between these viruses in diagnostic tests. In this study, we mapped the binding epitope for 2D9 on the GI.2 The VP60 capsid protein demonstrated the neutralizing capacity of this mAb, which was able to prevent GI.2 infections in an experimental challenge. Our results suggest that external loops (1, 4 and 5) in the P2 subdomain of VP60 contribute to the discontinuous neutralizing epitope recognized by mAb 2D9. Moreover, analysis of naturally occurring RHDV GI.2 isolates revealed key residues involved in mAb 2D9 binding that are under selective pressure. The findings described in this work provide valuable information regarding our understanding of virus neutralization and immune escape, which may help in the development of novel antiviral compounds.
Collapse
Affiliation(s)
- Ana Podadera
- Instituto Universitario de Biotecnología de Asturias, Departamento de Bioquímica y Biología Molecular, Edificio Santiago Gascón, Universidad de Oviedo, Campus El Cristo, 33006, Oviedo, Spain
- Chemistry and Biochemistry Department, University of Windsor, 401 Sunset Avenue, Windsor, ON, N9B 3P4, Canada
| | - Mila Leuthold
- Medicinal Chemistry, Institute of Pharmacy and Molecular Biotechnology, University of Heidelberg, 69120, Heidelberg, Germany
| | - José Manuel Martín-Alonso
- Instituto Universitario de Biotecnología de Asturias, Departamento de Bioquímica y Biología Molecular, Edificio Santiago Gascón, Universidad de Oviedo, Campus El Cristo, 33006, Oviedo, Spain
| | - Rosa Casais
- SERIDA, Servicio Regional de Investigación y Desarrollo Agroalimentario, Centro de Biotecnología Animal, 33394, Gijón, Asturias, Spain
| | - Angel Luis Álvarez
- Instituto Universitario de Biotecnología de Asturias, Departamento de Bioquímica y Biología Molecular, Edificio Santiago Gascón, Universidad de Oviedo, Campus El Cristo, 33006, Oviedo, Spain
| | - M J Lobo-Castañón
- Departamento de Química Física y Analítica, Universidad de Oviedo, Av. Julián Clavería 8, 33006, Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias, Avenida de Roma, 33011, Oviedo, Spain
| | - Francisco Parra
- Instituto Universitario de Biotecnología de Asturias, Departamento de Bioquímica y Biología Molecular, Edificio Santiago Gascón, Universidad de Oviedo, Campus El Cristo, 33006, Oviedo, Spain
| | - Kevin Paul Dalton
- Instituto Universitario de Biotecnología de Asturias, Departamento de Bioquímica y Biología Molecular, Edificio Santiago Gascón, Universidad de Oviedo, Campus El Cristo, 33006, Oviedo, Spain.
| |
Collapse
|
5
|
Zhang SJ, Su D, Zhao SB, Xing JY, Zeng L, Wang J, Ming SL, Chu BB. Isolation, Identification, and Genetic Evolution Analysis of VP1 Gene of Feline Calicivirus Strain ZZ202306. Int J Mol Sci 2025; 26:2565. [PMID: 40141207 PMCID: PMC11942290 DOI: 10.3390/ijms26062565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Revised: 03/10/2025] [Accepted: 03/11/2025] [Indexed: 03/28/2025] Open
Abstract
This study investigated a suspected Feline calicivirus (FCV) outbreak at a veterinary facility in Zhengzhou, Henan Province, China. RT-PCR analysis confirmed the FCV presence, with subsequent CRFK cell culture propagation leading to the isolation and characterization of strain ZZ202306. Immunofluorescence and Western blot analyses validated the specificity of monoclonal antibodies targeting the FCV VP1 capsid protein. Transmission electron microscopy revealed non-enveloped virions of ~40 nm in diameter, exhibiting typical caliciviral architecture. Viral replication kinetics demonstrated exponential growth between 6 and 18 h post-inoculation, reaching a peak titer of 107.96 TCID50/0.1 mL. Genomic sequencing coupled with phylogenetic reconstruction of the VP1 gene revealed a close genetic relation to domestic Chinese strains and international variants, while maintaining distinct evolutionary divergence from other calicivirus genera.
Collapse
Affiliation(s)
- Shi-Jun Zhang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China; (S.-J.Z.); (D.S.); (S.-B.Z.); (J.-Y.X.); (L.Z.); (J.W.)
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs, Zhengzhou 450046, China
- Key Laboratory of Animal Growth and Development of Henan Province, Henan Agricultural University, Zhengzhou 450046, China
| | - Dan Su
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China; (S.-J.Z.); (D.S.); (S.-B.Z.); (J.-Y.X.); (L.Z.); (J.W.)
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs, Zhengzhou 450046, China
- Key Laboratory of Animal Growth and Development of Henan Province, Henan Agricultural University, Zhengzhou 450046, China
| | - Shi-Bo Zhao
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China; (S.-J.Z.); (D.S.); (S.-B.Z.); (J.-Y.X.); (L.Z.); (J.W.)
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs, Zhengzhou 450046, China
- Key Laboratory of Animal Growth and Development of Henan Province, Henan Agricultural University, Zhengzhou 450046, China
| | - Jia-You Xing
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China; (S.-J.Z.); (D.S.); (S.-B.Z.); (J.-Y.X.); (L.Z.); (J.W.)
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs, Zhengzhou 450046, China
- Key Laboratory of Animal Growth and Development of Henan Province, Henan Agricultural University, Zhengzhou 450046, China
| | - Lei Zeng
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China; (S.-J.Z.); (D.S.); (S.-B.Z.); (J.-Y.X.); (L.Z.); (J.W.)
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs, Zhengzhou 450046, China
- Key Laboratory of Animal Growth and Development of Henan Province, Henan Agricultural University, Zhengzhou 450046, China
| | - Jiang Wang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China; (S.-J.Z.); (D.S.); (S.-B.Z.); (J.-Y.X.); (L.Z.); (J.W.)
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs, Zhengzhou 450046, China
- Key Laboratory of Animal Growth and Development of Henan Province, Henan Agricultural University, Zhengzhou 450046, China
| | - Sheng-Li Ming
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China; (S.-J.Z.); (D.S.); (S.-B.Z.); (J.-Y.X.); (L.Z.); (J.W.)
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs, Zhengzhou 450046, China
- Key Laboratory of Animal Growth and Development of Henan Province, Henan Agricultural University, Zhengzhou 450046, China
| | - Bei-Bei Chu
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China; (S.-J.Z.); (D.S.); (S.-B.Z.); (J.-Y.X.); (L.Z.); (J.W.)
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs, Zhengzhou 450046, China
- Key Laboratory of Animal Growth and Development of Henan Province, Henan Agricultural University, Zhengzhou 450046, China
- International Joint Research Center of National Animal Immunology, Henan Agricultural University, Zhengzhou 450046, China
| |
Collapse
|
6
|
Lewis CB, Sherry L, Conley MJ, Nakashima M, Akbar S, Govindan N, Hosie MJ, Bhella D. Conformational Flexibility in Capsids Encoded by the Caliciviridae. Viruses 2024; 16:1835. [PMID: 39772145 PMCID: PMC11680396 DOI: 10.3390/v16121835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 11/06/2024] [Accepted: 11/22/2024] [Indexed: 01/11/2025] Open
Abstract
Caliciviruses are a diverse group of non-enveloped, positive-sense RNA viruses with a wide range of hosts and transmission routes. Norovirus is the most well-known member of the Caliciviridae; the acute gastroenteritis caused by human norovirus (HuNoV), for example, frequently results in closures of hospital wards and schools during the winter months. One area of calicivirus biology that has gained increasing attention over the past decade is the conformational flexibility exhibited by the protruding (P) domains of the major capsid protein VP1. This was observed in structure analyses of capsids encoded by many species and is often a consequence of environmental cues such as metal ions, changes to pH, or receptor/co-factor engagement. This review summarises the current understanding of P-domain flexibility, discussing the role this region plays in caliciviral infection and immune evasion, and highlighting potential avenues for further investigation.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - David Bhella
- MRC—University of Glasgow Centre for Virus Research, Garscube Campus, 464 Bearsden Road, Glasgow G61 1QH, UK; (C.B.L.); (L.S.); (M.J.C.); (M.N.); (S.A.); (M.J.H.)
| |
Collapse
|
7
|
Yang Y, Qi R, Chen M, Feng K, Liu Z, Kang H, Jiang Q, Qu L, Liu J. Screening and Immune Efficacy Evaluation of Antigens with Protection Against Feline Calicivirus. Vaccines (Basel) 2024; 12:1205. [PMID: 39591108 PMCID: PMC11598032 DOI: 10.3390/vaccines12111205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 10/17/2024] [Accepted: 10/21/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Feline calicivirus (FCV), a pathogen that causes upper respiratory tract diseases in felids, primarily leads to oral ulcers and various respiratory symptoms, which can be fatal in severe cases. Currently, FCV prevention and control rely primarily on vaccination; however, the existing vaccine types in China are mainly inactivated vaccines, leading to a single prevention and control method with suboptimal outcomes. METHODS AND RESULTS This study commences with a genetic evolution analysis of Chinese FCV isolates, confirming the presence of two major genotypes, GI and GII with GI emerging as the dominant form. We subsequently selected the broadly neutralizing vaccine candidate strain DL39 as the template for the truncation and expression of multiple recombinant proteins. Through serological assays, we successfully confirmed the optimal protective antigen region, which is designated CE39 (CDE). Further investigation revealed the location of the optimal protective antigen region within the CE region for both the GI and GII genotype strains. Capitalizing on this discovery, a bivalent recombinant protein, designated CE39-CEFB, was generated. Cat antisera generated against CE39 and CE39-CEFB proteins were used in cross-neutralization against various strains of different genotypes, yielding high neutralization titers ranging from 1:45 to 1:15 and from 1:48 to 1:29, respectively, which surpassed those induced by antisera from cats vaccinated with Mi-aosanduo (commercial vaccine, strain 255). Ultimately, in vivo challenge experiments were per-formed after immunizing cats with the CE39 and CE39-CEFB proteins, utilizing Miaosanduo as a control for comparison. The results demonstrated that immunization with both proteins effectively made cats less susceptible to FCV GI, GII, and VSD strains infection, resulting in superior immune efficacy compared with that in the Miaosanduo group. CONCLUSION These results indicate that this study successfully identified the antigen CE39, which has broad-spectrum antigenicity, through in vivo and in vitro experiments. These findings pre-liminarily demonstrate that the optimal protective antigen region of FCV strains is the CE region, laying a theoretical foundation for the development of novel broad-spectrum vaccines against FCV disease.
Collapse
Affiliation(s)
- Yupeng Yang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China
| | - Ruibin Qi
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China
| | - Mengru Chen
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Kexin Feng
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China
| | - Zhe Liu
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China
| | - Hongtao Kang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China
| | - Qian Jiang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China
| | - Liandong Qu
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China
| | - Jiasen Liu
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China
| |
Collapse
|
8
|
Sun W, Wang M, Shi Z, Wang P, Wang J, Du B, Wang S, Sun Z, Liu Z, Wei L, Yang D, He X, Wang J. VP2 mediates the release of the feline calicivirus RNA genome by puncturing the endosome membrane of infected cells. J Virol 2024; 98:e0035024. [PMID: 38591900 PMCID: PMC11092339 DOI: 10.1128/jvi.00350-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 03/19/2024] [Indexed: 04/10/2024] Open
Abstract
Feline calicivirus (FCV) is one of the few members of the Caliciviridae family that grows well in cell lines and, therefore, serves as a surrogate to study the biology of other viruses in the family. Conley et al. (14) demonstrated that upon the receptor engagement to the capsid, FCV VP2 forms a portal-like assembly, which might provide a channel for RNA release. However, the process of calicivirus RNA release is not yet fully understood. Our findings suggest that the separation of the FCV capsid from its genome RNA (gRNA) occurs rapidly in the early endosomes of infected cells. Using a liposome model decorated with the FCV cell receptor fJAM-A, we demonstrate that FCV releases its gRNA into the liposomes by penetrating membranes under low pH conditions. Furthermore, we found that VP2, which is rich in hydrophobic residues at its N-terminus, functions as the pore-forming protein. When we substituted the VP2 N-terminal hydrophobic residues, the gRNA release efficacy of the FCV mutants decreased. In conclusion, our results suggest that in the acidic environment of early endosomes, FCV VP2 functions as the pore-forming protein to mediate gRNA release into the cytoplasm of infected cells. This provides insight into the mechanism of calicivirus genome release.IMPORTANCEResearch on the biology and pathogenicity of certain caliciviruses, such as Norovirus and Sapovirus, is hindered by the lack of easy-to-use cell culture system. Feline calicivirus (FCV), which grows effectively in cell lines, is used as a substitute. At present, there is limited understanding of the genome release mechanism in caliciviruses. Our findings suggest that FCV uses VP2 to pierce the endosome membrane for genome release and provide new insights into the calicivirus gRNA release mechanism.
Collapse
Affiliation(s)
- Weiyao Sun
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Ming Wang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Zhibin Shi
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Pengfei Wang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Jinhui Wang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Bingchen Du
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Shida Wang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Zhenzhao Sun
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Zaisi Liu
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Lili Wei
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Decheng Yang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Xijun He
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Jingfei Wang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| |
Collapse
|
9
|
Cao L, Liu J, Li Y, Xie D, Yan Q, Li Q, Cao Y, Du W, Li J, Ye Z, Zhou D, Kang C, Cao S. Biological Characteristics of Feline Calicivirus Epidemic Strains in China and Screening of Broad-Spectrum Protective Vaccine Strains. Vaccines (Basel) 2023; 11:1858. [PMID: 38140261 PMCID: PMC10747013 DOI: 10.3390/vaccines11121858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 12/02/2023] [Accepted: 12/11/2023] [Indexed: 12/24/2023] Open
Abstract
Feline calicivirus (FCV) is one of the most important pathogens causing upper respiratory tract diseases in cats, posing a serious health threat to these animals. At present, FCV is mainly prevented through vaccination, but the protective efficacy of vaccines in China is limited. In this study, based on the differences in capsid proteins of isolates from different regions in China, as reported in our previous studies, seven representative FCV epidemic strains were selected and tested for their viral titers, virulence, immunogenicity, and extensive cross-protection. Subsequently, vaccine strains were selected to prepare inactivated vaccines. The whole-genome sequencing and analysis results showed that these seven representative FCV strains and 144 reference strains fell into five groups (A, B, C, D, and E). The strains isolated in China mainly fall into groups C and D, exhibiting regional characteristics. These Chinese isolates had a distant evolutionary relationship and low homology with the current FCV-255 vaccine strain. The screened FCV-HB7 and FCV-HB10 strains displayed desirable in vitro culture characteristics, with the highest virus proliferation titers (109.5 TCID50/mL) at 36 h post inoculation at a dose of 0.01 MOI. All five cats infected intranasally with FCV-HB7 or FCV-HB10 strains showed obvious clinical symptoms of FCV. The symptoms of cats infected with the FCV-HB7 strain were more severe than those infected with the FCV-HB10 strain. Both the single-strain inactivated immunization and combined bivalent inactivated vaccine immunization of FCV-HB7 and FCV-HB10 induced high neutralizing antibody titers in five cats immunized. Moreover, bivalent inactivated vaccine immunization protected cats from FCV-HB7 and FCV-HB10 strains. The cross-neutralizing antibody titer against seven representative FCV epidemic strains achieved by combined bivalent inactivated vaccine immunization was higher than that achieved by single-strain immunization, which was much higher than that achieved by commercial vaccine FCV-255 strain immunization. The above results suggest that the FCV-HB7 and FCV-HB10 strains screened in this study have great potential to become vaccine strains with broad-spectrum protective efficacy. However, their immune protective efficacy needs to be further verified by multiple methods before clinical application.
Collapse
Affiliation(s)
- Longlong Cao
- Wuhan Keqian Biology Co., Ltd., Wuhan 430070, China; (L.C.); (Y.L.); (Q.Y.); (Q.L.); (W.D.); (J.L.); (Z.Y.); (D.Z.)
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Jian Liu
- Shanghai Animal Disease Prevention and Control Center, Shanghai 201103, China;
| | - Yongfan Li
- Wuhan Keqian Biology Co., Ltd., Wuhan 430070, China; (L.C.); (Y.L.); (Q.Y.); (Q.L.); (W.D.); (J.L.); (Z.Y.); (D.Z.)
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Denglong Xie
- Zhejiang Hisun Animal Healthcare Products Co., Ltd., Hangzhou 311400, China;
| | - Quanhui Yan
- Wuhan Keqian Biology Co., Ltd., Wuhan 430070, China; (L.C.); (Y.L.); (Q.Y.); (Q.L.); (W.D.); (J.L.); (Z.Y.); (D.Z.)
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Qiuyan Li
- Wuhan Keqian Biology Co., Ltd., Wuhan 430070, China; (L.C.); (Y.L.); (Q.Y.); (Q.L.); (W.D.); (J.L.); (Z.Y.); (D.Z.)
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Yiran Cao
- Department of Life Science, Imperial College London, London SW7 2AZ, UK;
| | - Wenxin Du
- Wuhan Keqian Biology Co., Ltd., Wuhan 430070, China; (L.C.); (Y.L.); (Q.Y.); (Q.L.); (W.D.); (J.L.); (Z.Y.); (D.Z.)
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Jiakang Li
- Wuhan Keqian Biology Co., Ltd., Wuhan 430070, China; (L.C.); (Y.L.); (Q.Y.); (Q.L.); (W.D.); (J.L.); (Z.Y.); (D.Z.)
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Zijun Ye
- Wuhan Keqian Biology Co., Ltd., Wuhan 430070, China; (L.C.); (Y.L.); (Q.Y.); (Q.L.); (W.D.); (J.L.); (Z.Y.); (D.Z.)
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Dengyuan Zhou
- Wuhan Keqian Biology Co., Ltd., Wuhan 430070, China; (L.C.); (Y.L.); (Q.Y.); (Q.L.); (W.D.); (J.L.); (Z.Y.); (D.Z.)
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Chao Kang
- Wuhan Keqian Biology Co., Ltd., Wuhan 430070, China; (L.C.); (Y.L.); (Q.Y.); (Q.L.); (W.D.); (J.L.); (Z.Y.); (D.Z.)
| | - Shengbo Cao
- Wuhan Keqian Biology Co., Ltd., Wuhan 430070, China; (L.C.); (Y.L.); (Q.Y.); (Q.L.); (W.D.); (J.L.); (Z.Y.); (D.Z.)
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| |
Collapse
|
10
|
Salmen W, Hu L, Bok M, Chaimongkol N, Ettayebi K, Sosnovtsev SV, Soni K, Ayyar BV, Shanker S, Neill FH, Sankaran B, Atmar RL, Estes MK, Green KY, Parreño V, Prasad BVV. A single nanobody neutralizes multiple epochally evolving human noroviruses by modulating capsid plasticity. Nat Commun 2023; 14:6516. [PMID: 37845211 PMCID: PMC10579229 DOI: 10.1038/s41467-023-42146-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 09/28/2023] [Indexed: 10/18/2023] Open
Abstract
Acute gastroenteritis caused by human noroviruses (HuNoVs) is a significant global health and economic burden and is without licensed vaccines or antiviral drugs. The GII.4 HuNoV causes most epidemics worldwide. This virus undergoes epochal evolution with periodic emergence of variants with new antigenic profiles and altered specificity for histo-blood group antigens (HBGA), the determinants of cell attachment and susceptibility, hampering the development of immunotherapeutics. Here, we show that a llama-derived nanobody M4 neutralizes multiple GII.4 variants with high potency in human intestinal enteroids. The crystal structure of M4 complexed with the protruding domain of the GII.4 capsid protein VP1 revealed a conserved epitope, away from the HBGA binding site, fully accessible only when VP1 transitions to a "raised" conformation in the capsid. Together with dynamic light scattering and electron microscopy of the GII.4 VLPs, our studies suggest a mechanism in which M4 accesses the epitope by altering the conformational dynamics of the capsid and triggering its disassembly to neutralize GII.4 infection.
Collapse
Affiliation(s)
- Wilhelm Salmen
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, TX, USA
| | - Liya Hu
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, TX, USA
| | - Marina Bok
- Virology Institute and Technology Innovation, IVIT, CONICET-INTA, Hurlingham, Buenos Aires, Argentina
| | - Natthawan Chaimongkol
- Caliciviruses Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Khalil Ettayebi
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Stanislav V Sosnovtsev
- Caliciviruses Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Kaundal Soni
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, TX, USA
| | - B Vijayalakshmi Ayyar
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Sreejesh Shanker
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, TX, USA
| | - Frederick H Neill
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Banumathi Sankaran
- Berkeley Center for Structural Biology, Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley Laboratory, Berkeley, CA, USA
| | - Robert L Atmar
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Mary K Estes
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Kim Y Green
- Caliciviruses Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Viviana Parreño
- Virology Institute and Technology Innovation, IVIT, CONICET-INTA, Hurlingham, Buenos Aires, Argentina
| | - B V Venkataram Prasad
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, TX, USA.
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
11
|
Cao L, Li Q, Shi K, Wei L, Ouyang H, Ye Z, Du W, Ye J, Hui X, Li J, Cao S, Zhou D. Isolation and phylogenetic analysis of feline calicivirus strains from various region of China. ANIMAL DISEASES 2022. [DOI: 10.1186/s44149-022-00047-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
AbstractFeline calicivirus (FCV) is an important feline pathogen mainly causing upper respiratory tract disease, conjunctivitis, and stomatitis, and it is classified into genotype I and genotype II. To investigate the prevalence and molecular characteristics of FCV, this study collected 337 cat swab samples from animal hospitals in different regions of China from 2019 to 2021. The positive detection rate of FCV was 29.9% (101/337) by RT-PCR. Statistical analysis showed that FCV prevalence was significantly associated with living environment (p = 0.0004), age (p = 0.031) and clinical symptoms (p = 0.00), but not with sex (p = 0.092) and breed (p = 0.171). The 26 strains of FCV were isolated using F81 cells. Phylogenetic analysis showed that 10 isolates belonged to genotype I, and 16 isolates belonged to genotype II. These 26 isolates were highly genetically diverse, of which HB7 isolate had three same virulence-related amino acid loci with VSD strains. Potential loci distinguishing different genotypes were identified from 26 isolates, suggesting the genetic relationship between different genotypes. In addition, selection pressure analysis based on capsid protein of 26 isolates revealed that the protein is under diversifying selection. This study reveals the genetic diversity of FCV and provides a reference for the screening of vaccine candidate strains and the development of vaccines with better cross-protection effects.
Collapse
|
12
|
Fujita S, Koba R, Tohya Y. Identification of amino acid substitutions escaping from a broadly neutralizing monoclonal antibody of feline calicivirus. Virus Res 2022; 318:198848. [PMID: 35691421 DOI: 10.1016/j.virusres.2022.198848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 06/06/2022] [Accepted: 06/08/2022] [Indexed: 11/19/2022]
Abstract
Feline calicivirus (FCV) causes upper respiratory tract diseases in cats and has highly variable antigenicity for neutralization of each strain. Neutralizing epitopes of FCV are currently found in the hypervariable region (HVR) in the P2 domain of the major capsid protein VP1. Due to its unique ability to neutralize various FCV strains, 1D7 is a monoclonal antibody that may recognize a novel neutralizing epitope. While other neutralizing epitopes were characterized by producing neutralization-resistant variants, only 1D7-resistant variants could not be obtained, and its epitope has not been identified in the previous studies. In this study, we successfully generated these variants by multiple passaging of the FCV F4 strain in the presence of 1D7 and discovered that several amino acid substitutions (K638N, R662G, and T666I in the P1 domain of VP1) are involved in the decreased binding of 1D7. These substitution sites are also highly conserved among FCV strains compared with the substitution sites of other neutralization-resistant variants found in the HVR. Our results indicate that amino acid substitutions in the P1 domain, which are not responsible for direct interaction with the FCV receptor, are associated with neutralization escape. Since FCV can be conveniently cultured in vitro and the receptor required for infection is known, a detailed analysis of the 1D7 epitope could shed more light on the neutralization mechanism of the epitopes of viruses belonging to the Caliciviridae.
Collapse
Affiliation(s)
- Shigeru Fujita
- Laboratory of Veterinary Microbiology, Department of Veterinary Medicine, College of Bioresource Sciences, Nihon University, 1866 Kameino, Fujisawa, Kanagawa 252-0880, Japan
| | - Ryota Koba
- Laboratory of Veterinary Microbiology, Department of Veterinary Medicine, College of Bioresource Sciences, Nihon University, 1866 Kameino, Fujisawa, Kanagawa 252-0880, Japan
| | - Yukinobu Tohya
- Laboratory of Veterinary Microbiology, Department of Veterinary Medicine, College of Bioresource Sciences, Nihon University, 1866 Kameino, Fujisawa, Kanagawa 252-0880, Japan.
| |
Collapse
|
13
|
Huang J, Park GW, Jones RM, Fraser AM, Vinjé J, Jiang X. Efficacy of EPA-registered disinfectants against two human norovirus surrogates and Clostridioides difficile endospores. J Appl Microbiol 2022; 132:4289-4299. [PMID: 35279925 PMCID: PMC9119914 DOI: 10.1111/jam.15524] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 03/04/2022] [Accepted: 03/10/2022] [Indexed: 11/27/2022]
Abstract
AIMS To determine the efficacy of a panel of nine EPA-registered disinfectants against two human norovirus (HuNoV) surrogates (feline calicivirus [FCV] and Tulane virus [TuV]) and Clostridioides difficile endospores. METHODS AND RESULTS Nine EPA-registered products, five of which contained H2 O2 as active ingredient, were tested against infectious FCV, TuV and C. difficile endospores using two ASTM methods, a suspension and carrier test. Efficacy claims against FCV were confirmed for 8 of 9 products. The most efficacious product containing H2 O2 as ingredient achieved a >5.1 log reduction of FCV and >3.1 log reduction of TuV after 5 min, and >6.0 log reduction of C. difficile endospores after 10 min. Of the five products containing H2 O2 , no strong correlation (R2 = 0.25, p = 0.03) was observed between disinfection efficacy and H2 O2 concentration. Addition of 0.025% ferrous sulphate to 1% H2 O2 solution improved efficacy against FCV, TuV and C. difficile. CONCLUSION Disinfectants containing H2 O2 are the most efficacious disinfection products against FCV, TuV and C. difficile endospores. Product formulation, rather than the concentration of H2 O2 in a product, impacts the efficacy of a disinfection product. SIGNIFICANCE AND IMPACT OF STUDY H2 O2 -based disinfectants are efficacious against surrogate viruses for HuNoV and C. difficile endospores.
Collapse
Affiliation(s)
- Jinge Huang
- Department of Food, Nutrition, and Packaging Sciences, Clemson University, Clemson, South Carolina, USA
| | - Geun Woo Park
- Division of Viral Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Rachael M. Jones
- Department of Family and Preventive Medicine, University of Utah, Salt Lake City, Utah, USA
| | - Angela M. Fraser
- Department of Food, Nutrition, and Packaging Sciences, Clemson University, Clemson, South Carolina, USA
| | - Jan Vinjé
- Division of Viral Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Xiuping Jiang
- Department of Food, Nutrition, and Packaging Sciences, Clemson University, Clemson, South Carolina, USA
| |
Collapse
|
14
|
Qu Z, Kang H, Cui C, Meng K, Zhang X, Qu L, Zhang Y, Meng G. Purification-induced damage to calicivirus particles at near-atomic resolution. J Gen Virol 2022; 103. [PMID: 35579608 DOI: 10.1099/jgv.0.001742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The purification of virus particles is an essential process for the manufacture of vaccines. However, the application of different purification processes may affect the quality of the virus particles, such as structural integrity and homogeneity, which may further influence the infectivity and immunogenicity of the purified virus. In this study, we took Feline calicivirus (FCV), a common natural pathogen in cats belonging to Caliciviridae, as a research model. By using cryo-electron microscopy (cryo-EM), we incorporated the 3D classification process as a virus flexibility evaluation system. Cryo-EM images of virus particles resulting from different purification processes were compared at near-atomic resolution. The results indicated that molecular sieving purification will impact the stability of P-domains through increasing flexibility as determined by the evaluation system, which can be extended to assess the purification effect on the entire particle. This evaluation process can be further applied to all non-enveloped viruses.
Collapse
Affiliation(s)
- Zehui Qu
- College of Veterinary Medicine, China Agricultural University, Beijing, 100094, PR China.,The Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, PR China
| | - Hongtao Kang
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, PR China
| | - Chenxi Cui
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences (CAS), Beijing 100101, PR China.,University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Kaiwen Meng
- College of Veterinary Medicine, China Agricultural University, Beijing, 100094, PR China
| | - Xinzheng Zhang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences (CAS), Beijing 100101, PR China.,University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Liandong Qu
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, PR China
| | - Yueping Zhang
- College of Veterinary Medicine, China Agricultural University, Beijing, 100094, PR China
| | - Geng Meng
- College of Veterinary Medicine, China Agricultural University, Beijing, 100094, PR China
| |
Collapse
|
15
|
Atomic Structure of the Human Sapovirus Capsid Reveals a Unique Capsid Protein Conformation in Caliciviruses. J Virol 2022; 96:e0029822. [PMID: 35435722 PMCID: PMC9093105 DOI: 10.1128/jvi.00298-22] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Sapovirus (SaV) is a member of the Caliciviridae family, which causes acute gastroenteritis in humans and animals. Human sapoviruses (HuSaVs) are genetically and antigenically diverse, but the lack of a viral replication system and structural information has hampered the development of vaccines and therapeutics. Here, we successfully produced a self-assembled virus-like particle (VLP) from the HuSaV GI.6 VP1 protein, and the first atomic structure was determined using single-particle cryo-electron microscopy (cryo-EM) at a 2.9-Å resolution. The atomic model of the VP1 protein revealed a unique capsid protein conformation in caliciviruses. All N-terminal arms in the A, B, and C subunits interacted with adjacent shell domains after extending through their subunits. The roof of the arched VP1 dimer was formed between the P2 subdomains by the interconnected β strands and loops, and its buried surface was minimized compared to those of other caliciviruses. Four hypervariable regions that are potentially involved in the antigenic diversity of SaV formed extensive clusters on top of the P domain. Potential receptor binding regions implied by tissue culture mutants of porcine SaV were also located near these hypervariable clusters. Conserved sequence motifs of the VP1 protein, “PPG” and “GWS,” may stabilize the inner capsid shell and the outer protruding domain, respectively. These findings will provide the structural basis for the medical treatment of HuSaV infections and facilitate the development of vaccines, antivirals, and diagnostic systems. IMPORTANCE SaV and norovirus, belonging to the Caliciviridae family, are common causes of acute gastroenteritis in humans and animals. SaV and norovirus infections are public health problems in all age groups, which occur explosively and sporadically worldwide. HuSaV is genetically and antigenically diverse and is currently classified into 4 genogroups consisting of 18 genotypes based on the sequence similarity of the VP1 proteins. Despite these detailed genetic analyses, the lack of structural information on viral capsids has become a problem for the development of vaccines or antiviral drugs. The 2.9-Å atomic model of the HuSaV GI.6 VLP presented here not only revealed the location of the amino acid residues involved in immune responses and potential receptor binding sites but also provided essential information for the design of stable constructs needed for the development of vaccines and antivirals.
Collapse
|
16
|
Hu L, Salmen W, Chen R, Zhou Y, Neill F, Crowe JE, Atmar RL, Estes MK, Prasad BVV. Atomic structure of the predominant GII.4 human norovirus capsid reveals novel stability and plasticity. Nat Commun 2022; 13:1241. [PMID: 35273142 PMCID: PMC8913647 DOI: 10.1038/s41467-022-28757-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 02/08/2022] [Indexed: 12/30/2022] Open
Abstract
Human noroviruses (HuNoVs) cause sporadic and epidemic viral gastroenteritis worldwide. The GII.4 variants are responsible for most HuNoV infections, and GII.4 virus-like particles (VLPs) are being used in vaccine development. The atomic structure of the GII.4 capsid in the native T = 3 state has not been determined. Here we present the GII.4 VLP structure with T = 3 symmetry determined using X-ray crystallography and cryo-EM at 3.0 Å and 3.8 Å resolution, respectively, which reveals unanticipated novel features. A novel aspect in the crystal structure determined without imposing icosahedral symmetry is the remarkable adaptability of the capsid protein VP1 driven by the flexible hinge between the shell and the protruding domains. In both crystal and cryo-EM structures, VP1 adopts a stable conformation with the protruding domain resting on the shell domain, in contrast to the 'rising' conformation observed in recent cryo-EM structures of other GII.4 VLPs. Our studies further revealed that the resting state of VP1 dimer is stabilized by a divalent ion, and chelation using EDTA increases capsid diameter, exposing new hydrophobic and antigenic sites and suggesting a transition to the rising conformation. These novel insights into GII.4 capsid structure, stability, and antigen presentation may be useful for ongoing vaccine development.
Collapse
Affiliation(s)
- Liya Hu
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Wilhelm Salmen
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Rong Chen
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Yi Zhou
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Frederick Neill
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - James E Crowe
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN, USA.,Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Robert L Atmar
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA.,Section of Infectious Diseases, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Mary K Estes
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA.,Section of Infectious Diseases, Department of Medicine, Baylor College of Medicine, Houston, TX, USA.,Section of Gastroenterology and Hepatology, Department of Medicine, Baylor College of Medicine, Houston, TX, 77030, USA
| | - B V Venkataram Prasad
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX, USA. .,Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
17
|
Adams MC, Schiltz CJ, Heck ML, Chappie JS. Crystal structure of the potato leafroll virus coat protein and implications for viral assembly. J Struct Biol 2021; 214:107811. [PMID: 34813955 DOI: 10.1016/j.jsb.2021.107811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 11/04/2021] [Accepted: 11/13/2021] [Indexed: 10/19/2022]
Abstract
Luteoviruses, poleroviruses, and enamoviruses are insect-transmitted, agricultural pathogens that infect a wide array of plants, including staple food crops. Previous cryo-electron microscopy studies of virus-like particles show that luteovirid viral capsids are built from a structural coat protein that organizes with T = 3 icosahedral symmetry. Here, we present the crystal structure of a truncated version of the coat protein monomer from potato leafroll virus at 1.80-Å resolution. In the crystal lattice, monomers pack into flat sheets that preserve the two-fold and three-fold axes of icosahedral symmetry and show minimal structural deviations when compared to the full-length subunits of the assembled virus-like particle. These observations have important implications in viral assembly and maturation and suggest that the CP N-terminus and its interactions with RNA play an important role in generating capsid curvature.
Collapse
Affiliation(s)
- Myfanwy C Adams
- Department of Molecular Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Carl J Schiltz
- Department of Molecular Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Michelle L Heck
- Plant Pathology and Plant-Microbe Biology Section, School of Integrative Plant Science, Cornell University, Ithaca, NY 14853, USA; Boyce Thompson Institute, Ithaca, NY 14853, USA; Robert W. Holley Center for Agriculture and Health, Emerging Pests and Pathogens Research Unit, USDA Agricultural Research Service, Ithaca, NY 14853, USA
| | - Joshua S Chappie
- Department of Molecular Medicine, Cornell University, Ithaca, NY 14853, USA.
| |
Collapse
|
18
|
Kamiya R, Uchiyama J, Matsuzaki S, Murata K, Iwasaki K, Miyazaki N. Acid-stable capsid structure of Helicobacter pylori bacteriophage KHP30 by single-particle cryoelectron microscopy. Structure 2021; 30:300-312.e3. [PMID: 34597601 DOI: 10.1016/j.str.2021.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 07/04/2021] [Accepted: 09/09/2021] [Indexed: 10/20/2022]
Abstract
The acid-stable capsid structures of Helicobacter pylori phages KHP30 and KHP40 are solved at 2.7 and 3.0 Å resolutions by cryoelectron microscopy, respectively. The capsids have icosahedral T = 9 symmetry and consist of each 540 copies of 2 structural proteins, a major capsid protein, and a cement protein. The major capsid proteins form 12 pentagonal capsomeres occupying icosahedral vertexes and 80 hexagonal capsomeres located at icosahedral faces and edges. The major capsid protein has a unique protruding loop extending to the neighboring subunit that stabilizes hexagonal capsomeres. Furthermore, the capsid is decorated with trimeric cement proteins with a jelly roll motif. The cement protein trimer sits on the quasi-three-fold axis formed by three major capsid protein capsomeres, thereby enhancing the particle stability by connecting these capsomeres. Sequence and structure comparisons between the related Helicobacter pylori phages suggest a possible mechanism of phage adaptation to the human gastric environment.
Collapse
Affiliation(s)
- Ryosuke Kamiya
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8777, Japan
| | - Jumpei Uchiyama
- Laboratory of Veterinary Microbiology I, School of Veterinary Medicine, Azabu University, Kanagawa 252-5201, Japan; Department of Bacteriology, Graduate School of Medicine Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, Japan
| | - Shigenobu Matsuzaki
- Department of Clinical Laboratory Science, Faculty of Health Sciences, Kochi Gakuen University, Kochi 780-0955, Japan
| | - Kazuyoshi Murata
- National Institute for Physiological Sciences, Okazaki, Aichi 444-8585, Japan
| | - Kenji Iwasaki
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8777, Japan
| | - Naoyuki Miyazaki
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8777, Japan.
| |
Collapse
|
19
|
Alvarado G, Salmen W, Ettayebi K, Hu L, Sankaran B, Estes MK, Venkataram Prasad BV, Crowe JE. Broadly cross-reactive human antibodies that inhibit genogroup I and II noroviruses. Nat Commun 2021; 12:4320. [PMID: 34262046 PMCID: PMC8280134 DOI: 10.1038/s41467-021-24649-w] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 06/29/2021] [Indexed: 12/14/2022] Open
Abstract
The rational development of norovirus vaccine candidates requires a deep understanding of the antigenic diversity and mechanisms of neutralization of the virus. Here, we isolate and characterize a panel of broadly cross-reactive naturally occurring human monoclonal IgMs, IgAs and IgGs reactive with human norovirus (HuNoV) genogroup I or II (GI or GII). We note three binding patterns and identify monoclonal antibodies (mAbs) that neutralize at least one GI or GII HuNoV strain when using a histo-blood group antigen (HBGA) blocking assay. The HBGA blocking assay and a virus neutralization assay using human intestinal enteroids reveal that the GII-specific mAb NORO-320, mediates HBGA blocking and neutralization of multiple GII genotypes. The Fab form of NORO-320 neutralizes GII.4 infection more potently than the mAb, however, does not block HBGA binding. The crystal structure of NORO-320 Fab in complex with GII.4 P-domain shows that the antibody recognizes a highly conserved region in the P-domain distant from the HBGA binding site. Dynamic light scattering analysis of GII.4 virus-like particles with mAb NORO-320 shows severe aggregation, suggesting neutralization is by steric hindrance caused by multivalent cross-linking. Aggregation was not observed with the Fab form of NORO-320, suggesting that this clone also has additional inhibitory features.
Collapse
Affiliation(s)
- Gabriela Alvarado
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Wilhelm Salmen
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Khalil Ettayebi
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Liya Hu
- The Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Banumathi Sankaran
- Berkeley Center for Structural Biology, Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley Laboratory, Berkeley, CA, USA
| | - Mary K Estes
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
- Department of Medicine-Gastroenterology and Hepatology, Baylor College of Medicine, Houston, TX, USA
| | - B V Venkataram Prasad
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA.
- The Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX, USA.
| | - James E Crowe
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA.
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN, USA.
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
20
|
The Cryo-EM Structure of Vesivirus 2117 Highlights Functional Variations in Entry Pathways for Viruses in Different Clades of the Vesivirus Genus. J Virol 2021; 95:e0028221. [PMID: 33853966 DOI: 10.1128/jvi.00282-21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Vesivirus 2117 is an adventitious agent that has been responsible for lost productivity in biopharmaceutical production following contamination of Chinese hamster ovary cell cultures in commercial bioreactors. A member of the Caliciviridae, 2117 is classified within the Vesivirus genus in a clade that includes canine and mink caliciviruses but is distinct from the vesicular exanthema of swine virus (VESV) clade, which includes the extensively studied feline calicivirus (FCV). We have used cryogenic electron microscopy (cryo-EM) to determine the structure of the capsid of this small, icosahedral, positive-sense-RNA-containing virus. We show that the outer face of the dimeric capsomeres, which contains the receptor binding site and major immunodominant epitopes in all caliciviruses studied thus far, is quite different from that of FCV. This is a consequence of a 22-amino-acid insertion in the sequence of the FCV major capsid protein that forms a "cantilevered arm" that both plays an important role in receptor engagement and undergoes structural rearrangements thought to be important for genome delivery to the cytosol. Our data highlight a potentially important difference in the attachment and entry pathways employed by the different clades of the Vesivirus genus. IMPORTANCE Vesivirus 2117 has caused significant losses in manufacturing of biopharmaceutical products following contamination of cell cultures used in their production. We report the structure of the vesivirus 2117 capsid, the shell that encloses the virus's genome. Comparison of this structure with that of a related vesivirus, feline calicivirus (FCV), highlighted potentially important differences related to virus attachment and entry. Our findings suggest that these two viruses may bind differently to receptors at the host cell surface. We also show that a region of the capsid protein of FCV that rearranges following receptor engagement is not present in vesivirus 2117. These structural changes in the FCV capsid have been shown to allow the assembly of a portal-like structure that is hypothesized to deliver the viral genome to the cell's interior. Our data suggest that the 2117 portal assembly may employ a different means of anchoring to the outer face of the capsid.
Collapse
|
21
|
Anam ZE, Joshi N, Gupta S, Yadav P, Chaurasiya A, Kahlon AK, Kaushik S, Munde M, Ranganathan A, Singh S. A De novo Peptide from a High Throughput Peptide Library Blocks Myosin A -MTIP Complex Formation in Plasmodium falciparum. Int J Mol Sci 2020; 21:ijms21176158. [PMID: 32859024 PMCID: PMC7503848 DOI: 10.3390/ijms21176158] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 05/09/2020] [Accepted: 05/15/2020] [Indexed: 01/09/2023] Open
Abstract
Apicomplexan parasites, through their motor machinery, produce the required propulsive force critical for host cell-entry. The conserved components of this so-called glideosome machinery are myosin A and myosin A Tail Interacting Protein (MTIP). MTIP tethers myosin A to the inner membrane complex of the parasite through 20 amino acid-long C-terminal end of myosin A that makes direct contacts with MTIP, allowing the invasion of Plasmodium falciparum in erythrocytes. Here, we discovered through screening a peptide library, a de-novo peptide ZA1 that binds the myosin A tail domain. We demonstrated that ZA1 bound strongly to myosin A tail and was able to disrupt the native myosin A tail MTIP complex both in vitro and in vivo. We then showed that a shortened peptide derived from ZA1, named ZA1S, was able to bind myosin A and block parasite invasion. Overall, our study identified a novel anti-malarial peptide that could be used in combination with other antimalarials for blocking the invasion of Plasmodium falciparum.
Collapse
Affiliation(s)
- Zill e Anam
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi 110067, India; (Z.e.A.); (P.Y.); (A.C.); (A.K.K.); (S.K.)
| | - Nishant Joshi
- Department of Life Sciences, School of Natural Sciences, Shiv Nadar University, Greater Noida, Uttar Pradesh 201304, India;
| | - Sakshi Gupta
- School of Physical Sciences, Jawaharlal Nehru University, New Delhi 110067, India; (S.G.); (M.M.)
| | - Preeti Yadav
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi 110067, India; (Z.e.A.); (P.Y.); (A.C.); (A.K.K.); (S.K.)
| | - Ayushi Chaurasiya
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi 110067, India; (Z.e.A.); (P.Y.); (A.C.); (A.K.K.); (S.K.)
| | - Amandeep Kaur Kahlon
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi 110067, India; (Z.e.A.); (P.Y.); (A.C.); (A.K.K.); (S.K.)
| | - Shikha Kaushik
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi 110067, India; (Z.e.A.); (P.Y.); (A.C.); (A.K.K.); (S.K.)
| | - Manoj Munde
- School of Physical Sciences, Jawaharlal Nehru University, New Delhi 110067, India; (S.G.); (M.M.)
| | - Anand Ranganathan
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi 110067, India; (Z.e.A.); (P.Y.); (A.C.); (A.K.K.); (S.K.)
- Correspondence: (A.R.); (S.S.)
| | - Shailja Singh
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi 110067, India; (Z.e.A.); (P.Y.); (A.C.); (A.K.K.); (S.K.)
- Correspondence: (A.R.); (S.S.)
| |
Collapse
|
22
|
Galkin S, Rozina A, Zalevsky A, Gottikh M, Anisenko A. A Fluorescent Assay to Search for Inhibitors of HIV-1 Integrase Interactions with Human Ku70 Protein, and Its Application for Characterization of Oligonucleotide Inhibitors. Biomolecules 2020; 10:E1236. [PMID: 32854330 PMCID: PMC7563236 DOI: 10.3390/biom10091236] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 08/21/2020] [Accepted: 08/24/2020] [Indexed: 12/15/2022] Open
Abstract
The search for compounds that can inhibit the interaction of certain viral proteins with their cellular partners is a promising trend in the development of antiviral drugs. We have previously shown that binding of HIV-1 integrase with human Ku70 protein is essential for viral replication. Here, we present a novel, cheap, and fast assay to search for inhibitors of these proteins' binding based on the usage of genetically encoded fluorescent tags linked to both integrase and Ku70. Using this approach, we have elucidated structure-activity relationships for a set of oligonucleotide conjugates with eosin and shown that their inhibitory activity is primarily achieved through interactions between the conjugate nucleic bases and integrase. Molecular modeling of HIV-1 integrase in complex with the conjugates suggests that they can shield E212/L213 residues in integrase, which are crucial for its efficient binding to Ku70, in a length-dependent manner. Using the developed system, we have found the 11-mer phosphorothioate bearing 3'-end eosin-Y to be the most efficient inhibitor among the tested conjugates.
Collapse
Affiliation(s)
- Simon Galkin
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119992 Moscow, Russia; (S.G.); (A.R.); (A.Z.)
| | - Anna Rozina
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119992 Moscow, Russia; (S.G.); (A.R.); (A.Z.)
| | - Arthur Zalevsky
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119992 Moscow, Russia; (S.G.); (A.R.); (A.Z.)
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997 Moscow, Russia
| | - Marina Gottikh
- Chemistry Department, Lomonosov Moscow State University, 119992 Moscow, Russia;
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia
| | - Andrey Anisenko
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119992 Moscow, Russia; (S.G.); (A.R.); (A.Z.)
- Chemistry Department, Lomonosov Moscow State University, 119992 Moscow, Russia;
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia
| |
Collapse
|
23
|
Song C, Takai-Todaka R, Miki M, Haga K, Fujimoto A, Ishiyama R, Oikawa K, Yokoyama M, Miyazaki N, Iwasaki K, Murakami K, Katayama K, Murata K. Dynamic rotation of the protruding domain enhances the infectivity of norovirus. PLoS Pathog 2020; 16:e1008619. [PMID: 32614892 PMCID: PMC7331980 DOI: 10.1371/journal.ppat.1008619] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Accepted: 05/11/2020] [Indexed: 12/16/2022] Open
Abstract
Norovirus is the major cause of epidemic nonbacterial gastroenteritis worldwide. Lack of structural information on infection and replication mechanisms hampers the development of effective vaccines and remedies. Here, using cryo-electron microscopy, we show that the capsid structure of murine noroviruses changes in response to aqueous conditions. By twisting the flexible hinge connecting two domains, the protruding (P) domain reversibly rises off the shell (S) domain in solutions of higher pH, but rests on the S domain in solutions of lower pH. Metal ions help to stabilize the resting conformation in this process. Furthermore, in the resting conformation, the cellular receptor CD300lf is readily accessible, and thus infection efficiency is significantly enhanced. Two similar P domain conformations were also found simultaneously in the human norovirus GII.3 capsid, although the mechanism of the conformational change is not yet clear. These results provide new insights into the mechanisms of non-enveloped norovirus transmission that invades host cells, replicates, and sometimes escapes the hosts immune system, through dramatic environmental changes in the gastrointestinal tract.
Collapse
Affiliation(s)
- Chihong Song
- National Institute for Physiological Sciences, Okazaki, Japan
| | - Reiko Takai-Todaka
- Laboratory of Viral Infection I, Department of Infection Control and Immunology, Ōmura Satoshi Memorial Institute & Graduate School of Infection Control Sciences, Kitasato University, Tokyo, Japan
| | | | - Kei Haga
- Laboratory of Viral Infection I, Department of Infection Control and Immunology, Ōmura Satoshi Memorial Institute & Graduate School of Infection Control Sciences, Kitasato University, Tokyo, Japan
| | - Akira Fujimoto
- Laboratory of Viral Infection I, Department of Infection Control and Immunology, Ōmura Satoshi Memorial Institute & Graduate School of Infection Control Sciences, Kitasato University, Tokyo, Japan
| | - Ryoka Ishiyama
- Laboratory of Viral Infection I, Department of Infection Control and Immunology, Ōmura Satoshi Memorial Institute & Graduate School of Infection Control Sciences, Kitasato University, Tokyo, Japan
| | - Kazuki Oikawa
- Laboratory of Viral Infection I, Department of Infection Control and Immunology, Ōmura Satoshi Memorial Institute & Graduate School of Infection Control Sciences, Kitasato University, Tokyo, Japan
| | | | - Naoyuki Miyazaki
- Institute for Protein Research, Osaka University, Suita, Japan
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance, University of Tsukuba, Tsukuba, Japan
| | - Kenji Iwasaki
- Institute for Protein Research, Osaka University, Suita, Japan
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance, University of Tsukuba, Tsukuba, Japan
| | | | - Kazuhiko Katayama
- Laboratory of Viral Infection I, Department of Infection Control and Immunology, Ōmura Satoshi Memorial Institute & Graduate School of Infection Control Sciences, Kitasato University, Tokyo, Japan
- National Institute of Infectious Diseases, Tokyo, Japan
- * E-mail: (KK); (KM)
| | - Kazuyoshi Murata
- National Institute for Physiological Sciences, Okazaki, Japan
- * E-mail: (KK); (KM)
| |
Collapse
|
24
|
Cubillos-Zapata C, Angulo I, Almanza H, Borrego B, Zamora-Ceballos M, Castón JR, Mena I, Blanco E, Bárcena J. Precise location of linear epitopes on the capsid surface of feline calicivirus recognized by neutralizing and non-neutralizing monoclonal antibodies. Vet Res 2020; 51:59. [PMID: 32357948 PMCID: PMC7195702 DOI: 10.1186/s13567-020-00785-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 04/21/2020] [Indexed: 12/16/2022] Open
Abstract
We report the generation, characterization and epitope mapping of a panel of 26 monoclonal antibodies (MAbs) against the VP1 capsid protein of feline calicivirus (FCV). Two close but distinct linear epitopes were identified at the capsid outermost surface (P2 subdomain) of VP1, within the E5′HVR antigenic hypervariable region: one spanning amino acids 431-435 (PAGDY), highly conserved and recognized by non-neutralizing MAbs; and a second epitope spanning amino acids 445-451 (ITTANQY), highly variable and recognized by neutralizing MAbs. These antibodies might be valuable for diagnostic applications, as well as for further research in different aspects of the biology of FCV.
Collapse
Affiliation(s)
- Carolina Cubillos-Zapata
- Centro de Investigación en Sanidad Animal, INIA-CISA, Valdeolmos, Madrid, Spain.,Innate Immunity Group, IdiPAZ Institute for Health Research, La Paz Hospital, 28046, Madrid, Spain
| | - Iván Angulo
- Centro de Investigación en Sanidad Animal, INIA-CISA, Valdeolmos, Madrid, Spain
| | - Horacio Almanza
- Centro de Investigación en Sanidad Animal, INIA-CISA, Valdeolmos, Madrid, Spain.,Facultad de Medicina y Psicología de la Universidad Autónoma de Baja California, Tijuana, Mexico
| | - Belén Borrego
- Centro de Investigación en Sanidad Animal, INIA-CISA, Valdeolmos, Madrid, Spain
| | | | - José R Castón
- Department of Structure of Macromolecules, Centro Nacional de Biotecnología/CSIC, Cantoblanco, Madrid, Spain
| | - Ignacio Mena
- Centro de Investigación en Sanidad Animal, INIA-CISA, Valdeolmos, Madrid, Spain.,Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Esther Blanco
- Centro de Investigación en Sanidad Animal, INIA-CISA, Valdeolmos, Madrid, Spain
| | - Juan Bárcena
- Centro de Investigación en Sanidad Animal, INIA-CISA, Valdeolmos, Madrid, Spain.
| |
Collapse
|
25
|
Aboubakr HA, Sampedro Parra F, Collins J, Bruggeman P, Goyal SM. Ìn situ inactivation of human norovirus GII.4 by cold plasma: Ethidium monoazide (EMA)-coupled RT-qPCR underestimates virus reduction and fecal material suppresses inactivation. Food Microbiol 2020; 85:103307. [PMID: 31500711 DOI: 10.1016/j.fm.2019.103307] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 08/19/2019] [Accepted: 08/20/2019] [Indexed: 12/14/2022]
Abstract
Cold atmospheric-gaseous plasma (CAP) is an emerging non-thermal technology for decontamination of foodborne bacterial and viral pathogens. We obtained a >5 log10 reduction in the titer (TCID50) of feline calicivirus (FCV) on stainless steel discs and Romaine lettuce leaves after 3 min wet exposure to air plasma generated by a two-dimensional array of integrated coaxial-microhollow dielectric barrier discharge (2D-AICM-DBD). However, when human norovirus (HuNoV GII.4) was treated for 5 min under the same conditions, ~2.6 log10 (>99.5%) reduction in genome copy number was observed as measured by ethidium monoazide-coupled RT-qPCR (EMA-RT-qPCR). To assess this discrepancy, we studied CAP's effect on FCV by the cell culture method and by the EMA-coupled RT-qPCR method. It was found that the molecular titration method (EMA-RT-qPCR) underestimates the level of virus reduction by CAP. Additionally, the fecal matter present in HuNoV samples partially suppressed virucidal activity of CAP. Assuming that the lower virus reduction measured by EMA-RT-qPCR method compared to cell culture method for FCV is the same as for HuNoV, we can conclude that FCV may be used as a surrogate for HuNoV to assess the virucidal effect of CAP. CAP is able to inactivate 3.5 Log10 units of HuNoV at low titers after 2 min of exposure.
Collapse
Affiliation(s)
- Hamada A Aboubakr
- Department of Veterinary Population Medicine and Veterinary Diagnostic Laboratory, College of Veterinary Medicine, University of Minnesota, 1333 Gortner Ave,St. Paul, MN, 55108, USA.
| | - Fernando Sampedro Parra
- Center for Animal Health and Food Safety, University of Minnesota, St. Paul, MN, 55018, USA.
| | - James Collins
- Department of Veterinary Population Medicine and Veterinary Diagnostic Laboratory, College of Veterinary Medicine, University of Minnesota, 1333 Gortner Ave,St. Paul, MN, 55108, USA.
| | - Peter Bruggeman
- Department of Mechanical Engineering, College of Science and Engineering, University of Minnesota, 111 Church Street SE, Minneapolis, MN, 55455, USA.
| | - Sagar M Goyal
- Department of Veterinary Population Medicine and Veterinary Diagnostic Laboratory, College of Veterinary Medicine, University of Minnesota, 1333 Gortner Ave,St. Paul, MN, 55108, USA.
| |
Collapse
|
26
|
Heffron J, McDermid B, Maher E, McNamara PJ, Mayer BK. Mechanisms of virus mitigation and suitability of bacteriophages as surrogates in drinking water treatment by iron electrocoagulation. WATER RESEARCH 2019; 163:114877. [PMID: 31349091 DOI: 10.1016/j.watres.2019.114877] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 07/14/2019] [Accepted: 07/15/2019] [Indexed: 05/03/2023]
Abstract
Emerging water treatment technologies using ferrous and zero-valent iron show promising virus mitigation by both inactivation and adsorption. In this study, iron electrocoagulation was investigated for virus mitigation in drinking water via bench-scale batch experiments. Relative contributions of physical removal and inactivation, as determined by recovery via pH 9.5 beef broth elution, were investigated for three mammalian viruses (adenovirus, echovirus, and feline calicivirus) and four bacteriophage surrogates (fr, MS2, P22, and ΦX174). Though no one bacteriophage exactly represented mitigation of the mammalian viruses in all water matrices, bacteriophage ΦX174 was the only surrogate that showed overall removal comparable to that of the mammalian viruses. Bacteriophages fr, MS2, and P22 were all more susceptible to inactivation than the three mammalian viruses, raising concerns about the suitability of these common surrogates as indicators of virus mitigation. To determine why some bacteriophages were particularly susceptible to inactivation, mechanisms of bacteriophage mitigation due to electrocoagulation were investigated. Physical removal was primarily due to inclusion in flocs, while inactivation was primarily due to ferrous iron oxidation. Greater electrostatic attraction, virus aggregation, and capsid durability were proposed as reasons for virus susceptibility to ferrous-based inactivation. Results suggest that overall treatment claims based on bacteriophage mitigation for any iron-based technology should be critically considered due to higher susceptibility of bacteriophages to inactivation via ferrous oxidation.
Collapse
Affiliation(s)
- Joe Heffron
- Department of Civil, Construction and Environmental Engineering, Marquette University, 1637 W. Wisconsin Ave., Milwaukee, WI, 53233, USA
| | - Brad McDermid
- Department of Civil, Construction and Environmental Engineering, Marquette University, 1637 W. Wisconsin Ave., Milwaukee, WI, 53233, USA
| | - Emily Maher
- Department of Civil, Construction and Environmental Engineering, Marquette University, 1637 W. Wisconsin Ave., Milwaukee, WI, 53233, USA
| | - Patrick J McNamara
- Department of Civil, Construction and Environmental Engineering, Marquette University, 1637 W. Wisconsin Ave., Milwaukee, WI, 53233, USA
| | - Brooke K Mayer
- Department of Civil, Construction and Environmental Engineering, Marquette University, 1637 W. Wisconsin Ave., Milwaukee, WI, 53233, USA.
| |
Collapse
|
27
|
Graziano VR, Wei J, Wilen CB. Norovirus Attachment and Entry. Viruses 2019; 11:E495. [PMID: 31151248 PMCID: PMC6630345 DOI: 10.3390/v11060495] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 05/24/2019] [Accepted: 05/25/2019] [Indexed: 12/19/2022] Open
Abstract
Human norovirus is a major human pathogen causing the majority of cases of viral gastroenteritis globally. Viral entry is the first step of the viral life cycle and is a significant determinant of cell tropism, host range, immune interactions, and pathogenesis. Bile salts and histo-blood group antigens are key mediators of norovirus entry; however, the molecular mechanisms by which these molecules promote infection and the identity of a potential human norovirus receptor remain unknown. Recently, there have been several important advances in norovirus entry biology including the identification of CD300lf as the receptor for murine norovirus and of the role of the minor capsid protein VP2 in viral genome release. Here, we will review the current understanding about norovirus attachment and entry and highlight important future directions.
Collapse
Affiliation(s)
- Vincent R Graziano
- Departments of Laboratory Medicine and Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA.
| | - Jin Wei
- Departments of Laboratory Medicine and Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA.
| | - Craig B Wilen
- Departments of Laboratory Medicine and Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA.
| |
Collapse
|
28
|
Conley MJ, McElwee M, Azmi L, Gabrielsen M, Byron O, Goodfellow IG, Bhella D. Calicivirus VP2 forms a portal-like assembly following receptor engagement. Nature 2019; 565:377-381. [PMID: 30626974 DOI: 10.1038/s41586-018-0852-1] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 12/10/2018] [Indexed: 11/08/2022]
Abstract
To initiate infection, many viruses enter their host cells by triggering endocytosis following receptor engagement. However, the mechanisms by which non-enveloped viruses escape the endosome are poorly understood. Here we present near-atomic-resolution cryo-electron microscopy structures for feline calicivirus both undecorated and labelled with a soluble fragment of its cellular receptor, feline junctional adhesion molecule A. We show that VP2, a minor capsid protein encoded by all caliciviruses1,2, forms a large portal-like assembly at a unique three-fold axis of symmetry, following receptor engagement. This assembly-which was not detected in undecorated virions-is formed of twelve copies of VP2, arranged with their hydrophobic N termini pointing away from the virion surface. Local rearrangement at the portal site leads to the opening of a pore in the capsid shell. We hypothesize that the portal-like assembly functions as a channel for the delivery of the calicivirus genome, through the endosomal membrane, into the cytoplasm of a host cell, thereby initiating infection. VP2 was previously known to be critical for the production of infectious virus3; our findings provide insights into its structure and function that advance our understanding of the Caliciviridae.
Collapse
Affiliation(s)
- Michaela J Conley
- Medical Research Council University of Glasgow Centre for Virus Research, Glasgow, UK
| | - Marion McElwee
- Medical Research Council University of Glasgow Centre for Virus Research, Glasgow, UK
| | - Liyana Azmi
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | | | - Olwyn Byron
- School of Life Sciences, University of Glasgow, Glasgow, UK
| | - Ian G Goodfellow
- Division of Virology, Department of Pathology, University of Cambridge, Cambridge, UK
| | - David Bhella
- Medical Research Council University of Glasgow Centre for Virus Research, Glasgow, UK.
| |
Collapse
|
29
|
Abstract
Genetic economy is a key feature in all aspects of viral replication. Some viruses are able to function as independently evolving entities with as few as two genes, while satellite viruses have been described that encode a single gene product. To accommodate the need for genetic economy, the viral infectious entity - the virion, generally assembles in a highly-symmetrical manner. Viral structural proteins are multifunctional, accomplishing several tasks beyond their primary role of forming protective shells. These include mediating attachment and entry, avoiding and regulating host responses to infection and sometimes mediating gene expression and genome replication. Here we introduce some of the basic principles of virus assembly with examples to show how recurring motifs are seen in spherical viruses that infect diverse host species, how some viruses use helical assemblies to encapsidate their genomes and how viral envelope glycoproteins accomplish membrane fusion.
Collapse
|
30
|
Bat Caliciviruses and Human Noroviruses Are Antigenically Similar and Have Overlapping Histo-Blood Group Antigen Binding Profiles. mBio 2018; 9:mBio.00869-18. [PMID: 29789360 PMCID: PMC5964351 DOI: 10.1128/mbio.00869-18] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Emerging zoonotic viral diseases remain a challenge to global public health. Recent surveillance studies have implicated bats as potential reservoirs for a number of viral pathogens, including coronaviruses and Ebola viruses. Caliciviridae represent a major viral family contributing to emerging diseases in both human and animal populations and have been recently identified in bats. In this study, we blended metagenomics, phylogenetics, homology modeling, and in vitro assays to characterize two novel bat calicivirus (BtCalV) capsid sequences, corresponding to strain BtCalV/A10/USA/2009, identified in Perimyotis subflavus near Little Orleans, MD, and bat norovirus. We observed that bat norovirus formed virus-like particles and had epitopes and receptor-binding patterns similar to those of human noroviruses. To determine whether these observations stretch across multiple bat caliciviruses, we characterized a novel bat calicivirus, BtCalV/A10/USA/2009. Phylogenetic analysis revealed that BtCalV/A10/USA/2009 likely represents a novel Caliciviridae genus and is most closely related to "recoviruses." Homology modeling revealed that the capsid sequences of BtCalV/A10/USA/2009 and bat norovirus resembled human norovirus capsid sequences and retained host ligand binding within the receptor-binding domains similar to that seen with human noroviruses. Both caliciviruses bound histo-blood group antigens in patterns that overlapped those seen with human and animal noroviruses. Taken together, our results indicate the potential for bat caliciviruses to bind histo-blood group antigens and overcome a significant barrier to cross-species transmission. Additionally, we have shown that bat norovirus maintains antigenic epitopes similar to those seen with human noroviruses, providing further evidence of evolutionary descent. Our results reiterate the importance of surveillance of wild-animal populations, especially of bats, for novel viral pathogens.IMPORTANCE Caliciviruses are rapidly evolving viruses that cause pandemic outbreaks associated with significant morbidity and mortality globally. The animal reservoirs for human caliciviruses are unknown; bats represent critical reservoir species for several emerging and zoonotic diseases. Recent reports have identified several bat caliciviruses but have not characterized biological functions associated with disease risk, including their potential emergence in other mammalian populations. In this report, we identified a novel bat calicivirus that is most closely related to nonhuman primate caliciviruses. Using this new bat calicivirus and a second norovirus-like bat calicivirus capsid gene sequence, we generated virus-like particles that have host carbohydrate ligand binding patterns similar to those of human and animal noroviruses and that share antigens with human noroviruses. The similarities to human noroviruses with respect to binding patterns and antigenic epitopes illustrate the potential for bat caliciviruses to emerge in other species and the importance of pathogen surveillance in wild-animal populations.
Collapse
|
31
|
Kim Y, Chang KO. Fexaramine as an entry blocker for feline caliciviruses. Antiviral Res 2018; 152:76-83. [PMID: 29454892 PMCID: PMC7125566 DOI: 10.1016/j.antiviral.2018.02.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 02/06/2018] [Accepted: 02/12/2018] [Indexed: 12/21/2022]
Abstract
Feline calicivirus (FCV) is a small non-enveloped virus containing a single-stranded, positive-sense RNA genome of approximately 7.7 kb. FCV is a highly infectious pathogen of cats and typically causes moderate, self-limiting acute oral and upper respiratory tract diseases or chronic oral diseases. In addition, in recent years, virulent, systemic FCV (vs-FCV) strains causing severe systemic diseases with a high mortality rate of up to 67% have been reported in cats. Although FCV vaccines are commercially available, their efficacy is limited due to antigenic diversity of FCV strains and short duration of immunity. In this study, we identified fexaramine as a potent inhibitor of FCV including vs-FCV strains in cell culture and demonstrated that fexaramine is a entry blocker for FCV by using various experiments including time-of-addition studies, generation of resistant viruses in cell culture and the reverse genetics system. A fexaramine resistant FCV mutant has a single amino acid change in the P2 domain of VP1 (the major capsid), and the importance of this mutation for conferring resistance was confirmed using the reverse genetics system. A comparative analysis of viral resistance was also performed using a peptidyl inhibitor (NPI52) targeting FCV 3C-like protease. Finally, the effects of combination treatment of fexaramine and NPI52 against FCV replication and emergence of resistant viruses were investigated in cell culture.
Collapse
Affiliation(s)
- Yunjeong Kim
- Department of Diagnostic Medicine and Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA.
| | - Kyeong-Ok Chang
- Department of Diagnostic Medicine and Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA.
| |
Collapse
|
32
|
Conserved Surface Residues on the Feline Calicivirus Capsid Are Essential for Interaction with Its Receptor Feline Junctional Adhesion Molecule A (fJAM-A). J Virol 2018; 92:JVI.00035-18. [PMID: 29386293 DOI: 10.1128/jvi.00035-18] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2018] [Accepted: 01/24/2018] [Indexed: 12/11/2022] Open
Abstract
Host cell surface receptors are required for attachment, binding, entry, and infection by nonenveloped viruses. Receptor binding can induce conformational changes in the viral capsid and/or the receptor that couple binding with downstream events in the virus life cycle (intracellular signaling, endocytosis and trafficking, and membrane penetration). Virus-receptor interactions also influence viral spread and pathogenicity. The interaction between feline calicivirus (FCV) and its receptor, feline junctional adhesion molecule A (fJAM-A), on host cells is required for infection and induces irreversible, inactivating conformational changes in the capsid of some viral strains. Cryoelectron microscopy (cryo-EM) structures of FCV bound to fJAM-A showed several possible virus-receptor interactions. However, the specific residues on the viral capsid required for binding are not known. Capsid residues that may be involved in postbinding events have been implicated by isolation of soluble receptor-resistant (srr) mutants in which changes in the capsid protein sequence change the capacity of such srr mutants to be inactivated upon incubation with soluble fJAM-A. To clarify which residues on the surface of FCV are required for its interaction with fJAM-A and to potentially identify residues required for postreceptor binding events, we used the existing atomic-resolution structures of FCV and the FCV-fJAM-A cryo-EM structures to select 14 capsid residues for mutation and preparation of recombinant viral capsids. Using this approach, we identified residues on the FCV capsid that are required for fJAM-A binding and other residues that are not required for binding but are required for infection that are likely important for subsequent postbinding events.IMPORTANCE Feline calicivirus (FCV) is a common cause of mild upper respiratory disease in cats. Some FCV isolates can cause virulent systemic disease. The genetic determinants of virulence for FCV are unknown. We previously found that virulent FCV isolates have faster in vitro growth kinetics than less virulent isolates. Differences in viral growth in vitro may correlate with differences in virulence. Here, we investigated the roles of specific FCV capsid residues on the receptor-virus interaction and viral growth in vitro We show that the capsid protein genes of the virulent FCV-5 isolate determine its faster in vitro growth kinetics compared to those of the nonvirulent FCV-Urbana infectious clone. We also identified residues on the capsid VP1 protein that are important for receptor binding or for steps subsequent to receptor binding. Our data provide further insight into the specific molecular interactions between fJAM-A and the FCV capsid that regulate binding and infectious entry.
Collapse
|
33
|
Aboubakr HA, Mor SK, Higgins L, Armien A, Youssef MM, Bruggeman PJ, Goyal SM. Cold argon-oxygen plasma species oxidize and disintegrate capsid protein of feline calicivirus. PLoS One 2018; 13:e0194618. [PMID: 29566061 PMCID: PMC5864060 DOI: 10.1371/journal.pone.0194618] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 03/06/2018] [Indexed: 11/18/2022] Open
Abstract
Possible mechanisms that lead to inactivation of feline calicivirus (FCV) by cold atmospheric-pressure plasma (CAP) generated in 99% argon-1% O2 admixture were studied. We evaluated the impact of CAP exposure on the FCV viral capsid protein and RNA employing several cultural, molecular, proteomic and morphologic characteristics techniques. In the case of long exposure (2 min) to CAP, the reactive species of CAP strongly oxidized the major domains of the viral capsid protein (VP1) leading to disintegration of a majority of viral capsids. In the case of short exposure (15 s), some of the virus particles retained their capsid structure undamaged but failed to infect the host cells in vitro. In the latter virus particles, CAP exposure led to the oxidation of specific amino acids located in functional peptide residues in the P2 subdomain of the protrusion (P) domain, the dimeric interface region of VP1 dimers, and the movable hinge region linking the S and P domains. These regions of the capsid are known to play an essential role in the attachment and entry of the virus to the host cell. These observations suggest that the oxidative effect of CAP species inactivates the virus by hindering virus attachment and entry into the host cell. Furthermore, we found that the oxidative impact of plasma species led to oxidation and damage of viral RNA once it becomes unpacked due to capsid destruction. The latter effect most likely plays a secondary role in virus inactivation since the intact FCV genome is infectious even after damage to the capsid.
Collapse
Affiliation(s)
- Hamada A. Aboubakr
- Department of Veterinary Population Medicine and Veterinary Diagnostic Laboratory, College of Veterinary Medicine, University of Minnesota, St. Paul, MN, United States of America
- Food Science and Technology Department, Faculty of Agriculture, Alexandria University, Aflaton St, El-Shatby, Alexandria, Egypt
| | - Sunil K. Mor
- Department of Veterinary Population Medicine and Veterinary Diagnostic Laboratory, College of Veterinary Medicine, University of Minnesota, St. Paul, MN, United States of America
| | - LeeAnn Higgins
- Department of Biochemistry, Molecular Biology, and Biophysics & Proteomics Center for Mass Spectrometry, University of Minnesota, St. Paul, MN, United States of America
| | - Anibal Armien
- Department of Veterinary Population Medicine and Veterinary Diagnostic Laboratory, College of Veterinary Medicine, University of Minnesota, St. Paul, MN, United States of America
| | - Mohammed M. Youssef
- Food Science and Technology Department, Faculty of Agriculture, Alexandria University, Aflaton St, El-Shatby, Alexandria, Egypt
| | - Peter J. Bruggeman
- Department of Mechanical Engineering, College of Science and Engineering, University of Minnesota, Minneapolis, MN, United States of America
| | - Sagar M. Goyal
- Department of Veterinary Population Medicine and Veterinary Diagnostic Laboratory, College of Veterinary Medicine, University of Minnesota, St. Paul, MN, United States of America
- * E-mail:
| |
Collapse
|
34
|
Orchard RC, Wilen CB, Doench JG, Baldridge MT, McCune BT, Lee YCJ, Lee S, Pruett-Miller SM, Nelson CA, Fremont DH, Virgin HW. Discovery of a proteinaceous cellular receptor for a norovirus. Science 2016; 353:933-6. [PMID: 27540007 DOI: 10.1126/science.aaf1220] [Citation(s) in RCA: 224] [Impact Index Per Article: 24.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2015] [Accepted: 07/29/2016] [Indexed: 12/26/2022]
Abstract
Noroviruses (NoVs) are a leading cause of gastroenteritis globally, yet the host factors required for NoV infection are poorly understood. We identified host molecules that are essential for murine NoV (MNoV)-induced cell death, including CD300lf as a proteinaceous receptor. We found that CD300lf is essential for MNoV binding and replication in cell lines and primary cells. Additionally, Cd300lf(-/-) mice are resistant to MNoV infection. Expression of CD300lf in human cells breaks the species barrier that would otherwise restrict MNoV replication. The crystal structure of the CD300lf ectodomain reveals a potential ligand-binding cleft composed of residues that are critical for MNoV infection. Therefore, the presence of a proteinaceous receptor is the primary determinant of MNoV species tropism, whereas other components of cellular machinery required for NoV replication are conserved between humans and mice.
Collapse
Affiliation(s)
- Robert C Orchard
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Craig B Wilen
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - John G Doench
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Megan T Baldridge
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Broc T McCune
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Ying-Chiang J Lee
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Sanghyun Lee
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Shondra M Pruett-Miller
- Genome Engineering and iPSC Center, Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Christopher A Nelson
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Daved H Fremont
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Herbert W Virgin
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
35
|
Kemenesi G, Gellért Á, Dallos B, Görföl T, Boldogh S, Estók P, Marton S, Oldal M, Martella V, Bányai K, Jakab F. Sequencing and molecular modeling identifies candidate members of Caliciviridae family in bats. INFECTION GENETICS AND EVOLUTION 2016; 41:227-232. [PMID: 27085289 PMCID: PMC7172268 DOI: 10.1016/j.meegid.2016.04.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 04/05/2016] [Accepted: 04/06/2016] [Indexed: 12/20/2022]
Abstract
Emerging viral diseases represent an ongoing challenge for globalized world and bats constitute an immense, partially explored, reservoir of potentially zoonotic viruses. Caliciviruses are important human and animal pathogens and, as observed for human noroviruses, they may impact on human health on a global scale. By screening fecal samples of bats in Hungary, calicivirus RNA was identified in the samples of Myotis daubentonii and Eptesicus serotinus bats. In order to characterize more in detail the bat caliciviruses, large portions of the genome sequence of the viruses were determined. Phylogenetic analyses and molecular modeling identified firmly the two viruses as candidate members within the Caliciviridae family, with one calicivirus strain resembling members of the Sapovirus genus and the other bat calicivirus being more related to porcine caliciviruses of the proposed genus Valovirus. This data serves the effort for detecting reservoir hosts for potential emerging viruses and recognize important evolutionary relationships. Two novel bat caliciviruses were genetically characterized. Mature viral capsids were molecularly modeled. Bat caliciviruses are highly heterogeneous genetically. The two novel viruses are genetically related to valoviruses and sapoviruses. New sequences were most closely related to Chinese sequences.
Collapse
Affiliation(s)
- Gábor Kemenesi
- Virological Research Group, János Szentágothai Research Center, University of Pécs, Pécs, Hungary; Institute of Biology, Faculty of Sciences, University of Pécs, Pécs, Hungary
| | - Ákos Gellért
- Department of Applied Genomics, Agricultural Institute, Centre for Agricultural Research, Hungarian Academy of Sciences, Martonvásár, Hungary
| | - Bianka Dallos
- Virological Research Group, János Szentágothai Research Center, University of Pécs, Pécs, Hungary; Institute of Biology, Faculty of Sciences, University of Pécs, Pécs, Hungary
| | - Tamás Görföl
- Department of Zoology, Hungarian Natural History Museum, Budapest, Hungary; Institute for Veterinary Medical Research, Centre for Agricultural Research, Hungarian Academy of Sciences, Budapest, Hungary
| | | | - Péter Estók
- Department of Zoology, Eszterházy Károly College, Eger, Hungary
| | - Szilvia Marton
- Institute for Veterinary Medical Research, Centre for Agricultural Research, Hungarian Academy of Sciences, Budapest, Hungary
| | - Miklós Oldal
- Virological Research Group, János Szentágothai Research Center, University of Pécs, Pécs, Hungary; Institute of Biology, Faculty of Sciences, University of Pécs, Pécs, Hungary
| | - Vito Martella
- Dipartimento di Medicina Veterinaria, Universitá Aldo Moro di Bari, Valenzano, Italy
| | - Krisztián Bányai
- Institute for Veterinary Medical Research, Centre for Agricultural Research, Hungarian Academy of Sciences, Budapest, Hungary
| | - Ferenc Jakab
- Virological Research Group, János Szentágothai Research Center, University of Pécs, Pécs, Hungary; Institute of Biology, Faculty of Sciences, University of Pécs, Pécs, Hungary.
| |
Collapse
|
36
|
Structural Biology of Noroviruses. VIRAL GASTROENTERITIS 2016. [PMCID: PMC7149786 DOI: 10.1016/b978-0-12-802241-2.00016-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
Abstract
Noroviruses constitute a major genus in the family Caliciviridae, which contains icosahedral viruses with positive-sense single-stranded RNA genome. In humans, these constantly evolving viruses are the cause of sporadic and epidemic gastroenteritis. Despite a lack of a reproducible cell culture system or a small animal model, remarkable progress has been made in our understanding of the molecular biology, immunology, structural biology, and evolution of human noroviruses. This understanding is further enhanced by studies of nonhuman noroviruses and animal caliciviruses that are cultivatable. The main focus of this chapter is to review our current understanding of the structural biology of noroviruses in particular and of caliciviruses in general, with an emphasis on the unique modular organization of the capsid that allows for strain-dependent variations in glycan recognition and antigenicity to facilitate sustained virus evolution. Finally, structures of the proteins are reviewed that are critical for virus replication and that can be targeted in the design of small molecule drugs for use as effective antivirals.
Collapse
|
37
|
Mechanism of Cell Culture Adaptation of an Enteric Calicivirus, the Porcine Sapovirus Cowden Strain. J Virol 2015; 90:1345-58. [PMID: 26581980 DOI: 10.1128/jvi.02197-15] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Accepted: 11/08/2015] [Indexed: 01/21/2023] Open
Abstract
UNLABELLED The porcine sapovirus (SaV) (PoSaV) Cowden strain is one of only a few culturable enteric caliciviruses. Compared to the wild-type (WT) PoSaV Cowden strain, tissue culture-adapted (TC) PoSaV has two conserved amino acid substitutions in the RNA-dependent RNA polymerase (RdRp) and six in the capsid protein (VP1). By using the reverse-genetics system, we identified that 4 amino acid substitutions in VP1 (residues 178, 289, 324, and 328), but not the substitutions in the RdRp region, were critical for the cell culture adaptation of the PoSaV Cowden strain. The other two substitutions in VP1 (residues 291 and 295) reduced virus replication in vitro. Three-dimensional (3D) structural analysis of VP1 showed that residue 178 was located near the dimer-dimer interface, which may affect VP1 assembly and oligomerization; residues 289, 291, 324, and 328 were located at protruding subdomain 2 (P2) of VP1, which may influence virus binding to cellular receptors; and residue 295 was located at the interface of two monomeric VP1 proteins, which may influence VP1 dimerization. Although reversion of the mutation at residue 291 or 295 from that of the TC strain to that of the WT reduced virus replication in vitro, it enhanced virus replication in vivo, and the revertants induced higher-level serum and mucosal antibody responses than those induced by the TC PoSaV Cowden strain. Our findings reveal the molecular basis for PoSaV adaptation to cell culture. These findings may provide new, critical information for the cell culture adaptation of other PoSaV strains and human SaVs or noroviruses. IMPORTANCE The tissue culture-adapted porcine sapovirus Cowden strain is one of only a few culturable enteric caliciviruses. We discovered that 4 amino acid substitutions in VP1 (residues 178, 289, 324, and 328) were critical for its adaptation to LLC-PK cells. Two substitutions in VP1 (residues 291 and 295) reduced virus replication in vitro but enhanced virus replication and induced higher-level serum and mucosal antibody responses in gnotobiotic pigs than those induced by the tissue culture-adapted strain. Structural modeling analysis of VP1 suggested that residue 178 may affect VP1 assembly and oligomerization; residues 289, 291, 324, and 328 may influence virus binding to cellular receptors; and residue 295 may influence VP1 dimerization. Our findings will provide new information for the cell culture adaptation of other sapoviruses and possibly noroviruses.
Collapse
|
38
|
Burmeister WP, Buisson M, Estrozi LF, Schoehn G, Billet O, Hannas Z, Sigoillot C, Poulet H. Structure determination of feline calicivirus virus-like particles in the context of a pseudo-octahedral arrangement. PLoS One 2015; 10:e0119289. [PMID: 25794153 PMCID: PMC4368116 DOI: 10.1371/journal.pone.0119289] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Accepted: 01/12/2015] [Indexed: 12/21/2022] Open
Abstract
The vesivirus feline calicivirus (FCV) is a positive strand RNA virus encapsidated by an icosahedral T=3 shell formed by the viral VP1 protein. Upon its expression in the insect cell - baculovirus system in the context of vaccine development, two types of virus-like particles (VLPs) were formed, a majority built of 60 subunits (T=1) and a minority probably built of 180 subunits (T=3). The structure of the small particles was determined by x-ray crystallography at 0.8 nm resolution helped by cryo-electron microscopy in order to understand their formation. Cubic crystals belonged to space group P213. Their self-rotation function showed the presence of an octahedral pseudo-symmetry similar to the one described previously by Agerbandje and co-workers for human parvovirus VLPs. The crystal structure could be solved starting from the published VP1 structure in the context of the T=3 viral capsid. In contrast to viral capsids, where the capsomers are interlocked by the exchange of the N-terminal arm (NTA) domain, this domain is disordered in the T=1 capsid of the VLPs. Furthermore it is prone to proteolytic cleavage. The relative orientation of P (protrusion) and S (shell) domains is alerted so as to fit VP1 to the smaller T=1 particle whereas the intermolecular contacts around 2-fold, 3-fold and 5-fold axes are conserved. By consequence the surface of the VLP is very similar compared to the viral capsid and suggests a similar antigenicity. The knowledge of the structure of the VLPs will help to improve their stability, in respect to a use for vaccination.
Collapse
Affiliation(s)
- Wim P. Burmeister
- Unit of Virus Host Cell Interactions, Université Grenoble Alpes, Grenoble, France
- Unit of Virus Host Cell Interactions, Unité Mixte Internationale 3265, Centre National de Recherche Scientifique, Grenoble, France
- * E-mail:
| | - Marlyse Buisson
- Unit of Virus Host Cell Interactions, Université Grenoble Alpes, Grenoble, France
- Unit of Virus Host Cell Interactions, Unité Mixte Internationale 3265, Centre National de Recherche Scientifique, Grenoble, France
- Laboratoire de Virologie, Centre Hospitalo-Universitaire de Grenoble, Grenoble, France
| | - Leandro F. Estrozi
- Institut de Biologie Structurale Jean-Pierre Ebel, Commissariat d’Energie Atomique, Grenoble, France
- Institut de Biologie Structurale Jean-Pierre Ebel, Centre National de Recherche Scientifique, Grenoble, France
- Institut de Biologie Structurale Jean-Pierre Ebel, Université Grenoble Alpes, Grenoble, France
| | - Guy Schoehn
- Institut de Biologie Structurale Jean-Pierre Ebel, Commissariat d’Energie Atomique, Grenoble, France
- Institut de Biologie Structurale Jean-Pierre Ebel, Centre National de Recherche Scientifique, Grenoble, France
- Institut de Biologie Structurale Jean-Pierre Ebel, Université Grenoble Alpes, Grenoble, France
| | | | - Zahia Hannas
- Lyon Gerland Laboratory, Merial R&D, Lyon, France
| | | | - Hervé Poulet
- Lyon Gerland Laboratory, Merial R&D, Lyon, France
| |
Collapse
|
39
|
Host ICAMs play a role in cell invasion by Mycobacterium tuberculosis and Plasmodium falciparum. Nat Commun 2015; 6:6049. [PMID: 25586702 DOI: 10.1038/ncomms7049] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Accepted: 12/05/2014] [Indexed: 12/19/2022] Open
Abstract
Intercellular adhesion molecules (ICAMs) belong to the immunoglobulin superfamily and participate in diverse cellular processes including host-pathogen interactions. ICAM-1 is expressed on various cell types including macrophages, whereas ICAM-4 is restricted to red blood cells. Here we report the identification of an 11-kDa synthetic protein, M5, that binds to human ICAM-1 and ICAM-4, as shown by in vitro interaction studies, surface plasmon resonance and immunolocalization. M5 greatly inhibits the invasion of macrophages and erythrocytes by Mycobacterium tuberculosis and Plasmodium falciparum, respectively. Pharmacological and siRNA-mediated inhibition of ICAM-1 expression also results in reduced M. tuberculosis invasion of macrophages. ICAM-4 binds to P. falciparum merozoites, and the addition of recombinant ICAM-4 to parasite cultures blocks invasion of erythrocytes by newly released merozoites. Our results indicate that ICAM-1 and ICAM-4 play roles in host cell invasion by M. tuberculosis and P. falciparum, respectively, either as receptors or as crucial accessory molecules.
Collapse
|
40
|
Abstract
UNLABELLED Flavivirus replication is mediated by a membrane-associated replication complex where viral membrane proteins NS2A, NS2B, NS4A, and NS4B serve as the scaffold for the replication complex formation. Here, we used dengue virus serotype 2 (DENV-2) as a model to characterize viral NS4A-NS4B interaction. NS4A interacts with NS4B in virus-infected cells and in cells transiently expressing NS4A and NS4B in the absence of other viral proteins. Recombinant NS4A and NS4B proteins directly bind to each other with an estimated Kd (dissociation constant) of 50 nM. Amino acids 40 to 76 (spanning the first transmembrane domain, consisting of amino acids 50 to 73) of NS4A and amino acids 84 to 146 (also spanning the first transmembrane domain, consisting of amino acids 101 to 129) of NS4B are the determinants for NS4A-NS4B interaction. Nuclear magnetic resonance (NMR) analysis suggests that NS4A residues 17 to 80 form two amphipathic helices (helix α1, comprised of residues 17 to 32, and helix α2, comprised of residues 40 to 47) that associate with the cytosolic side of endoplasmic reticulum (ER) membrane and helix α3 (residues 52 to 75) that transverses the ER membrane. In addition, NMR analysis identified NS4A residues that may participate in the NS4A-NS4B interaction. Amino acid substitution of these NS4A residues exhibited distinct effects on viral replication. Three of the four NS4A mutations (L48A, T54A, and L60A) that affected the NS4A-NS4B interaction abolished or severely reduced viral replication; in contrast, two NS4A mutations (F71A and G75A) that did not affect NS4A-NS4B interaction had marginal effects on viral replication, demonstrating the biological relevance of the NS4A-NS4B interaction to DENV-2 replication. Taken together, the study has provided experimental evidence to argue that blocking the NS4A-NS4B interaction could be a potential antiviral approach. IMPORTANCE Flavivirus NS4A and NS4B proteins are essential components of the ER membrane-associated replication complex. The current study systematically characterizes the interaction between flavivirus NS4A and NS4B. Using DENV-2 as a model, we show that NS4A interacts with NS4B in virus-infected cells, in cells transiently expressing NS4A and NS4B proteins, or in vitro with recombinant NS4A and NS4B proteins. We mapped the minimal regions required for the NS4A-NS4B interaction to be amino acids 40 to 76 of NS4A and amino acids 84 to 146 of NS4B. NMR analysis revealed the secondary structure of amino acids 17 to 80 of NS4A and the NS4A amino acids that may participate in the NS4A-NS4B interaction. Functional analysis showed a correlation between viral replication and NS4A-NS4B interaction, demonstrating the biological importance of the NS4A-NS4B interaction. The study has advanced our knowledge of the molecular function of flavivirus NS4A and NS4B proteins. The results also suggest that inhibitors of the NS4A-NS4B interaction could be pursued for flavivirus antiviral development.
Collapse
|
41
|
Farkas T. Rhesus enteric calicivirus surrogate model for human norovirus gastroenteritis. J Gen Virol 2014; 96:1504-14. [PMID: 25502652 DOI: 10.1099/jgv.0.000020] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Human noroviruses are one of the major causes of acute gastroenteritis worldwide. Due to the lack of an efficient human norovirus cell culture system coupled with an animal model, human norovirus research mainly relies on human volunteer studies and surrogate models. Current models either utilize human norovirus-infected animals including the gnotobiotic pig or calf and the chimpanzee models, or employ other members of the family Caliciviridae including cell culture propagable surrogate caliciviruses such as the feline calicivirus, murine norovirus and most recently the Tulane virus. One of the major features of human noroviruses is their extreme biological diversity, including genetic, antigenic and histo-blood group antigen binding diversity, and possible differences of virulence and environmental stability. This extreme biological diversity and its effect on intervention/prevention strategies cannot be modelled by uniform groups of surrogates, much less by single isolates. Tulane virus, the prototype recovirus strain, was discovered in 2008. Since then, several other novel recoviruses have been described and cell culture adapted. Recent studies indicate that the epidemiology, the biological features and diversity of recoviruses and the course of infection and clinical disease in recovirus-infected macaques more closely reflect those properties of human noroviruses than any of the current surrogates. This review aims to summarize what is currently known about recoviruses, highlight their biological similarities to human noroviruses and discuss applications of the model in addressing questions relevant for human norovirus research.
Collapse
Affiliation(s)
- Tibor Farkas
- 1Division of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA 2University of Cincinnati College of Medicine, Cincinnati, OH, USA
| |
Collapse
|
42
|
Shionoiri N, Nogariya O, Tanaka M, Matsunaga T, Tanaka T. Capsid protein oxidation in feline calicivirus using an electrochemical inactivation treatment. JOURNAL OF HAZARDOUS MATERIALS 2014; 283:410-415. [PMID: 25310599 DOI: 10.1016/j.jhazmat.2014.09.049] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2014] [Revised: 09/22/2014] [Accepted: 09/23/2014] [Indexed: 06/04/2023]
Abstract
Pathogenic viral infections are an international public health concern, and viral disinfection has received increasing attention. Electrochemical treatment has been used for treatment of water contaminated by bacteria for several decades, and although in recent years several reports have investigated viral inactivation kinetics, the mode of action of viral inactivation by electrochemical treatment remains unclear. Here, we demonstrated the inactivation of feline calicivirus (FCV), a surrogate for human noroviruses, by electrochemical treatment in a developed flow-cell equipped with a screen-printed electrode. The viral infectivity titer was reduced by over 5 orders of magnitude after 15 min of treatment at 0.9V vs. Ag/AgCl. Proteomic study of electrochemically inactivated virus revealed oxidation of peptides located in the viral particles; oxidation was not observed in the non-treated sample. Furthermore, transmission electron microscopy revealed that viral particles in the treated sample had irregular structures. These results suggest that electrochemical treatment inactivates FCV via oxidation of peptides in the structural region, causing structural deformation of virus particles. This first report of viral protein damage through electrochemical treatment will contribute to broadening the understanding of viral inactivation mechanisms.
Collapse
Affiliation(s)
- Nozomi Shionoiri
- Division of Biotechnology and Life Science, Institute of Engineering, Tokyo University of Agriculture and Technology, 2-24-16 Naka-cho, Koganei, Tokyo 184-8588, Japan
| | - Osamu Nogariya
- Division of Biotechnology and Life Science, Institute of Engineering, Tokyo University of Agriculture and Technology, 2-24-16 Naka-cho, Koganei, Tokyo 184-8588, Japan
| | - Masayoshi Tanaka
- Division of Biotechnology and Life Science, Institute of Engineering, Tokyo University of Agriculture and Technology, 2-24-16 Naka-cho, Koganei, Tokyo 184-8588, Japan
| | - Tadashi Matsunaga
- Division of Biotechnology and Life Science, Institute of Engineering, Tokyo University of Agriculture and Technology, 2-24-16 Naka-cho, Koganei, Tokyo 184-8588, Japan
| | - Tsuyoshi Tanaka
- Division of Biotechnology and Life Science, Institute of Engineering, Tokyo University of Agriculture and Technology, 2-24-16 Naka-cho, Koganei, Tokyo 184-8588, Japan.
| |
Collapse
|
43
|
Kim DS, Hosmillo M, Alfajaro MM, Kim JY, Park JG, Son KY, Ryu EH, Sorgeloos F, Kwon HJ, Park SJ, Lee WS, Cho D, Kwon J, Choi JS, Kang MI, Goodfellow I, Cho KO. Both α2,3- and α2,6-linked sialic acids on O-linked glycoproteins act as functional receptors for porcine Sapovirus. PLoS Pathog 2014; 10:e1004172. [PMID: 24901849 PMCID: PMC4047124 DOI: 10.1371/journal.ppat.1004172] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Accepted: 04/21/2014] [Indexed: 12/23/2022] Open
Abstract
Sapovirus, a member of the Caliciviridae family, is an important cause of acute gastroenteritis in humans and pigs. Currently, the porcine sapovirus (PSaV) Cowden strain remains the only cultivable member of the Sapovirus genus. While some caliciviruses are known to utilize carbohydrate receptors for entry and infection, a functional receptor for sapovirus is unknown. To characterize the functional receptor of the Cowden strain of PSaV, we undertook a comprehensive series of protein-ligand biochemical assays in mock and PSaV-infected cell culture and/or piglet intestinal tissue sections. PSaV revealed neither hemagglutination activity with red blood cells from any species nor binding activity to synthetic histo-blood group antigens, indicating that PSaV does not use histo-blood group antigens as receptors. Attachment and infection of PSaV were markedly blocked by sialic acid and Vibrio cholerae neuraminidase (NA), suggesting a role for α2,3-linked, α2,6-linked or α2,8-linked sialic acid in virus attachment. However, viral attachment and infection were only partially inhibited by treatment of cells with sialidase S (SS) or Maackia amurensis lectin (MAL), both specific for α2,3-linked sialic acid, or Sambucus nigra lectin (SNL), specific for α2,6-linked sialic acid. These results indicated that PSaV recognizes both α2,3- and α2,6-linked sialic acids for viral attachment and infection. Treatment of cells with proteases or with benzyl 4-O-β-D-galactopyranosyl-β-D-glucopyranoside (benzylGalNAc), which inhibits O-linked glycosylation, also reduced virus binding and infection, whereas inhibition of glycolipd synthesis or N-linked glycosylation had no such effect on virus binding or infection. These data suggest PSaV binds to cellular receptors that consist of α2,3- and α2,6-linked sialic acids on glycoproteins attached via O-linked glycosylation.
Collapse
Affiliation(s)
- Deok-Song Kim
- Laboratory of Veterinary Pathology, College of Veterinary Medicine, Chonnam National University, Gwangju, Republic of Korea
| | - Myra Hosmillo
- Laboratory of Veterinary Pathology, College of Veterinary Medicine, Chonnam National University, Gwangju, Republic of Korea
| | - Mia Madel Alfajaro
- Laboratory of Veterinary Pathology, College of Veterinary Medicine, Chonnam National University, Gwangju, Republic of Korea
| | - Ji-Yun Kim
- Laboratory of Veterinary Pathology, College of Veterinary Medicine, Chonnam National University, Gwangju, Republic of Korea
| | - Jun-Gyu Park
- Laboratory of Veterinary Pathology, College of Veterinary Medicine, Chonnam National University, Gwangju, Republic of Korea
| | - Kyu-Yeol Son
- Laboratory of Veterinary Pathology, College of Veterinary Medicine, Chonnam National University, Gwangju, Republic of Korea
| | - Eun-Hye Ryu
- Laboratory of Veterinary Pathology, College of Veterinary Medicine, Chonnam National University, Gwangju, Republic of Korea
| | - Frederic Sorgeloos
- Division of Virology, Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | - Hyung-Jun Kwon
- Bioindustry Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup, Republic of Korea
| | - Su-Jin Park
- Bioindustry Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup, Republic of Korea
| | - Woo Song Lee
- Bioindustry Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup, Republic of Korea
| | - Duck Cho
- Department of Laboratory Medicine, Chonnam National University Hwasun Hospital, Jeollanam-do, Republic of Korea
| | - Joseph Kwon
- Division of Life Science, Korea Basic Science Institute, Daejeon, Republic of Korea
| | - Jong-Soon Choi
- Division of Life Science, Korea Basic Science Institute, Daejeon, Republic of Korea
| | - Mun-Il Kang
- Laboratory of Veterinary Pathology, College of Veterinary Medicine, Chonnam National University, Gwangju, Republic of Korea
| | - Ian Goodfellow
- Division of Virology, Department of Pathology, University of Cambridge, Cambridge, United Kingdom
- * E-mail: (IG); (KOC)
| | - Kyoung-Oh Cho
- Laboratory of Veterinary Pathology, College of Veterinary Medicine, Chonnam National University, Gwangju, Republic of Korea
- * E-mail: (IG); (KOC)
| |
Collapse
|
44
|
Lee JY, Choi DY, Cho MY, Park KE, Lee SH, Hun Cho S, Hong KS, Lim YT. Targeted theranostic nanoparticles: receptor-mediated entry into cells, pH-induced signal generation and cytosolic delivery. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2014; 10:901-906. [PMID: 24106164 DOI: 10.1002/smll.201302136] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2013] [Revised: 08/27/2013] [Indexed: 06/02/2023]
Abstract
Virus-like theranostic nanoparticles: virus-like poly(amino acid) nanoparticles are synthesized that can be internalized via receptor-mediated endocytosis, resulting in encapsulated pH-activatable fluorescence probes that can be turned on in acidic environments but otherwise remain undetectable. The encapsulated anticancer drugs are also released into cytosol by endosome disruption.
Collapse
Affiliation(s)
- Ji Yeon Lee
- Graduate School and Department of Analytical Science and Technology, Chungnam National University, Daejeon, 305-764, Republic of Korea
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Kniel KE. The makings of a good human norovirus surrogate. Curr Opin Virol 2014; 4:85-90. [PMID: 24492067 DOI: 10.1016/j.coviro.2014.01.002] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Revised: 01/03/2014] [Accepted: 01/08/2014] [Indexed: 11/18/2022]
Abstract
Norovirus is undoubtedly a leading cause of acute gastroenteritis. A large limitation to the study of human norovirus is the lack of consensus research using norovirus surrogates. Over two decades of research have included vast comparisons of norovirus surrogates within the Calicivirus family. A discussion on the continued use of norovirus surrogates includes use of surrogates to adequately assess environmental persistence and food preservation technologies. Choice of proper surrogate may be influenced by a myriad of issues, including ease of propagation, genetic similarities, and binding properties. While it remains impossible to routinely culture human norovirus in vitro the continued use of a variety of norovirus surrogates remains crucial to facilitate an understanding of norovirus in order to reduce the public health impact of the disease.
Collapse
Affiliation(s)
- Kalmia E Kniel
- Department of Animal and Food Sciences, University of Delaware, 044 Townsend Hall, 531 South College Ave, Newark, DE 19716, United States.
| |
Collapse
|
46
|
Genetic characterization of feline calicivirus strains associated with varying disease manifestations during an outbreak season in Missouri (1995–1996). Virus Genes 2013; 48:96-110. [DOI: 10.1007/s11262-013-1005-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2013] [Accepted: 10/24/2013] [Indexed: 11/26/2022]
|
47
|
Tresset G, Decouche V, Bryche JF, Charpilienne A, Le Cœur C, Barbier C, Squires G, Zeghal M, Poncet D, Bressanelli S. Unusual self-assembly properties of Norovirus Newbury2 virus-like particles. Arch Biochem Biophys 2013; 537:144-52. [DOI: 10.1016/j.abb.2013.07.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2013] [Revised: 06/12/2013] [Accepted: 07/03/2013] [Indexed: 10/26/2022]
|
48
|
Vongpunsawad S, Venkataram Prasad BV, Estes MK. Norwalk Virus Minor Capsid Protein VP2 Associates within the VP1 Shell Domain. J Virol 2013; 87:4818-25. [PMID: 23408637 PMCID: PMC3624303 DOI: 10.1128/jvi.03508-12] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Accepted: 02/08/2013] [Indexed: 11/20/2022] Open
Abstract
The major capsid protein of norovirus VP1 assembles to form an icosahedral viral particle. Despite evidence that the Norwalk virus (NV) minor structural protein VP2 is present in infectious virions, the available crystallographic and electron cryomicroscopy structures of NV have not revealed the location of VP2. In this study, we determined that VP1 associates with VP2 at the interior surface of the capsid, specifically with the shell (S) domain of VP1. We mapped the interaction site to amino acid 52 of VP1, an isoleucine located within a sequence motif IDPWI in the S domain that is highly conserved across norovirus genogroups. Mutation of this isoleucine abrogated VP2 incorporation into virus-like particles without affecting the ability for VP1 to dimerize and form particles. The highly basic nature of VP2 and its location interior to the viral particle are consistent with its potential role in assisting capsid assembly and genome encapsidation.
Collapse
Affiliation(s)
- Sompong Vongpunsawad
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | | | | |
Collapse
|
49
|
Cryo-EM structure of a novel calicivirus, Tulane virus. PLoS One 2013; 8:e59817. [PMID: 23533651 PMCID: PMC3606144 DOI: 10.1371/journal.pone.0059817] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2012] [Accepted: 02/19/2013] [Indexed: 12/18/2022] Open
Abstract
Tulane virus (TV) is a newly isolated cultivatable calicivirus that infects juvenile rhesus macaques. Here we report a 6.3 Å resolution cryo-electron microscopy structure of the TV virion. The TV virion is about 400 Å in diameter and consists of a T = 3 icosahedral protein capsid enclosing the RNA genome. 180 copies of the major capsid protein VP1 (∼57 KDa) are organized into two types of dimers A/B and C/C and form a thin, smooth shell studded with 90 dimeric protrusions. The overall capsid organization and the capsid protein fold of TV closely resemble that of other caliciviruses, especially of human Norwalk virus, the prototype human norovirus. These close structural similarities support TV as an attractive surrogate for the non-cultivatable human noroviruses. The most distinctive feature of TV is that its C/C dimers are in a highly flexible conformation with significantly reduced interactions between the shell (S) domain and the protruding (P) domain of VP1. A comparative structural analysis indicated that the P domains of TV C/C dimers were much more flexible than those of other caliciviruses. These observations, combined with previous studies on other caliciviruses, led us to hypothesize that the enhanced flexibility of C/C dimer P domains are likely required for efficient calicivirus-host cell interactions and the consequent uncoating and genome release. Residues in the S-P1 hinge between the S and P domain may play a critical role in the flexibility of P domains of C/C dimers.
Collapse
|
50
|
Wang X, Xu F, Liu J, Gao B, Liu Y, Zhai Y, Ma J, Zhang K, Baker TS, Schulten K, Zheng D, Pang H, Sun F. Atomic model of rabbit hemorrhagic disease virus by cryo-electron microscopy and crystallography. PLoS Pathog 2013; 9:e1003132. [PMID: 23341770 PMCID: PMC3547835 DOI: 10.1371/journal.ppat.1003132] [Citation(s) in RCA: 88] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2012] [Accepted: 11/29/2012] [Indexed: 02/03/2023] Open
Abstract
Rabbit hemorrhagic disease, first described in China in 1984, causes hemorrhagic necrosis of the liver. Its etiological agent, rabbit hemorrhagic disease virus (RHDV), belongs to the Lagovirus genus in the family Caliciviridae. The detailed molecular structure of any lagovirus capsid has yet to be determined. Here, we report a cryo-electron microscopic (cryoEM) reconstruction of wild-type RHDV at 6.5 Å resolution and the crystal structures of the shell (S) and protruding (P) domains of its major capsid protein, VP60, each at 2.0 Å resolution. From these data we built a complete atomic model of the RHDV capsid. VP60 has a conserved S domain and a specific P2 sub-domain that differs from those found in other caliciviruses. As seen in the shell portion of the RHDV cryoEM map, which was resolved to ∼5.5 Å, the N-terminal arm domain of VP60 folds back onto its cognate S domain. Sequence alignments of VP60 from six groups of RHDV isolates revealed seven regions of high variation that could be mapped onto the surface of the P2 sub-domain and suggested three putative pockets might be responsible for binding to histo-blood group antigens. A flexible loop in one of these regions was shown to interact with rabbit tissue cells and contains an important epitope for anti-RHDV antibody production. Our study provides a reliable, pseudo-atomic model of a Lagovirus and suggests a new candidate for an efficient vaccine that can be used to protect rabbits from RHDV infection. Rabbit hemorrhagic disease (RHD), first described in China in 1984, causes hemorrhagic necrosis of the liver within three days after infection and with a mortality rate that exceeds 90%. RHD has spread to large parts of the world and threatens the rabbit industry and related ecology. Its etiological agent, rabbit hemorrhagic disease virus (RHDV), belongs to the Lagovirus genus in the family Caliciviridae. Currently, the absence of a high-resolution model of any lagovirus impedes our understanding of its molecular interactions with hosts and successful design of an efficient anti-RHDV vaccine. Here, we use hybrid structural approaches to construct a pseudo-atomic model of RHDV that reveals significant differences in the P2 sub-domain of the major capsid protein compared to that seen in other caliciviruses. We identified seven regions of high sequence variation in this sub-domain that dictate the binding specificities of histo-blood group antigens. In one of these regions, we identified an antigenic peptide that interacts with rabbit tissue cells and elicits a significant immune response in rabbits and, hence, protects them from RHDV infection. Our pseudo-atomic model provides a structural framework for developing new anti-RHDV vaccines and will also help guide use of the RHDV capsid as a vehicle to display human tumor antigens as part of anti-tumor therapy.
Collapse
Affiliation(s)
- Xue Wang
- National Laboratory of Biomacromolecules, Institute of Biophysics (IBP), Chinese Academy of Sciences (CAS), Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Fengting Xu
- Department of Biochemistry and Molecular Biology, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Jiasen Liu
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Science, Harbin, China
| | - Bingquan Gao
- National Laboratory of Biomacromolecules, Institute of Biophysics (IBP), Chinese Academy of Sciences (CAS), Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yanxin Liu
- Beckman Institute and Department of Physics, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Yujia Zhai
- National Laboratory of Biomacromolecules, Institute of Biophysics (IBP), Chinese Academy of Sciences (CAS), Beijing, China
| | - Jun Ma
- National Laboratory of Biomacromolecules, Institute of Biophysics (IBP), Chinese Academy of Sciences (CAS), Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Kai Zhang
- National Laboratory of Biomacromolecules, Institute of Biophysics (IBP), Chinese Academy of Sciences (CAS), Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Timothy S. Baker
- Department of Chemistry and Biochemistry and Division of Biological Sciences, University of California-San Diego, La Jolla, California, United States of America
| | - Klaus Schulten
- Beckman Institute and Department of Physics, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Dong Zheng
- Analytical and Testing Center, Beijing Normal University, Beijing, China
- * E-mail: (DZ); (HP); (FS)
| | - Hai Pang
- School of Medicine, Tsinghua University, Beijing, China
- * E-mail: (DZ); (HP); (FS)
| | - Fei Sun
- National Laboratory of Biomacromolecules, Institute of Biophysics (IBP), Chinese Academy of Sciences (CAS), Beijing, China
- * E-mail: (DZ); (HP); (FS)
| |
Collapse
|