1
|
Pan Y, Li Z, Miao Q, Shi H, Guo L, Feng L, Tian J. Phylogenitc analysis and immunogenicity comparision of porcine genotype G9 rotavirus in China from 2020-2023. Virol Sin 2025:S1995-820X(25)00007-0. [PMID: 39988297 DOI: 10.1016/j.virs.2025.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/30/2024] [Accepted: 02/12/2025] [Indexed: 02/25/2025] Open
Abstract
As an emerging genotype, the G9 genotype rotaviruses (RVs) are widespread among humans and pigs, and have been reported in many countries and regions in the recent years. Moreover, porcine G9 strains could cross the interspecies barrier to infect human. To investigate the epidemic trends of porcine G9 strains as well as the cross-immunoreactivity among different isolates, an epidemiological investigation about porcine G9 genotype RVs (PoRVs) was performed during the period 2020-2023 in multiple provinces of China. A total of nine representative strains were identified. The phylogenetic analysis based on viral VP7 gene showed that these strains mainly clustered with lineages III and VI, which revealed the predominant G9 PoRVs in China. Moreover, a new lineage, lineage VII, was identified, and strains of this lineage were found to be circulating in Guangdong and Taiwan. Except lineages I and IV, some isolates from other lineages could co-circulate in pigs and humans. Three G9 strains, namely 923H, 923E, and 923X, which belonged to the largest sub-lineage III, were isolated. Then, the significant cross-reactivity was observed among strains of the same or different lineages. This study is the first to systematically investigate the genetic and immunogenetic characteristics of porcine G9 genotype rotavirus in China, as well as the potential cross-species transmission between pigs and humans, providing a valuable direction for the effective prevention of porcine rotavirus.
Collapse
Affiliation(s)
- Yudi Pan
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150069, China
| | - Zixin Li
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150069, China
| | - Qian Miao
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150069, China
| | - Hongyan Shi
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150069, China
| | - Longjun Guo
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150069, China
| | - Li Feng
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150069, China.
| | - Jin Tian
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150069, China.
| |
Collapse
|
2
|
Peng R, Wang M, Shahar S, Xiong G, Zhang Q, Pang L, Wang H, Kong X, Li D, Duan Z. Epidemiological, molecular, and evolutionary characteristics of G1P[8] rotavirus in China on the eve of RotaTeq application. Front Cell Infect Microbiol 2024; 14:1453862. [PMID: 39717546 PMCID: PMC11666228 DOI: 10.3389/fcimb.2024.1453862] [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] [Academic Contribution Register] [Received: 06/24/2024] [Accepted: 11/05/2024] [Indexed: 12/25/2024] Open
Abstract
Introduction This study, conducted in China prior to RotaTeq's launch, examined the epidemiological, molecular, and evolutionary features of the G1P[8] genotype RVA in children admitted with diarrhea, to aid in evaluating its efficacy and impact on G1P[8] RVA in China. Methods Data from the Chinese viral diarrhea surveillance network were collected from January 2016 to December 2018. RVA strains identified as the G1P[8] genotype were subjected to whole-genome sequencing. Neutralizing epitope, amino acid selection pressure, and evolution dynamics analyses on VP7 and VP4 were performed using BioEdit v.7.0.9.0 and PyMOL v.2.5.2, four algorithms (MEME, SLAC, FEL, and FUBAR) in the Datamonkey online software, and the MCMC model in BEAST v. 1.10.4, respectively. Phylogenetic and identity features of 11 genes were assessed by DNAStar and MEGA v.7. Results Results showed that the detection rate of G1P[8] in China from 2016 to 2018 was generally low with significant seasonality. The whole genome of G1P[8] of four 2016 childhood diarrhea specimens was successfully sequenced. Phylogenetic and neutralizing epitope analysis showed that Rotavin-M1 might have better protection on G1P[8] prevalent in China than Rotarix and RotaTeq. Two conserved N-glycosylation sites on VP7 of Chinese G1P[8] might affect the protective effect of the vaccine. Evolution rate and selection pressure analysis identified the possibility of rapidly evolving and adapting to the new environment introduced by vaccines of G1P[8], whereas positive selection specific to VP4 indicated the potential tendency to select for dominant traits. Identity and phylogeny analysis showed that Chinese G1P[8] from before 2018 was generally stable with possible genetic recombination among local strains. Discussion These findings not only are of great significance for predicting the prevalence of G1P [8] in China, but also provide data reference for evaluating rotavirus vaccine efficacy.
Collapse
Affiliation(s)
- Rui Peng
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases (NITFID), National Health Commission Key Laboratory for Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
- Department of Biosciences, Faculty of Sciences, Universiti Teknologi Malaysia, Johor Bahru, Malaysia
| | - Mengxuan Wang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases (NITFID), National Health Commission Key Laboratory for Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Saleha Shahar
- Department of Biosciences, Faculty of Sciences, Universiti Teknologi Malaysia, Johor Bahru, Malaysia
| | - Guangping Xiong
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases (NITFID), National Health Commission Key Laboratory for Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Qing Zhang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases (NITFID), National Health Commission Key Laboratory for Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Lili Pang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases (NITFID), National Health Commission Key Laboratory for Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Hong Wang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases (NITFID), National Health Commission Key Laboratory for Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Xiangyu Kong
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases (NITFID), National Health Commission Key Laboratory for Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Dandi Li
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases (NITFID), National Health Commission Key Laboratory for Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Zhaojun Duan
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases (NITFID), National Health Commission Key Laboratory for Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| |
Collapse
|
3
|
Nagarathinam K, Scheck A, Labuhn M, Ströh LJ, Herold E, Veselkova B, Tune S, Cramer JT, Rosset S, Vollers SS, Bankwitz D, Ballmaier M, Böning H, Roth E, Khera T, Ahsendorf-Abidi HP, Dittrich-Breiholz O, Obleser J, Nassal M, Jäck HM, Pietschmann T, Correia BE, Krey T. Epitope-focused immunogens targeting the hepatitis C virus glycoproteins induce broadly neutralizing antibodies. SCIENCE ADVANCES 2024; 10:eado2600. [PMID: 39642219 PMCID: PMC11623273 DOI: 10.1126/sciadv.ado2600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 01/30/2024] [Accepted: 11/04/2024] [Indexed: 12/08/2024]
Abstract
Hepatitis C virus (HCV) infection causes ~290,000 annual human deaths despite the highly effective antiviral treatment available. Several viral immune evasion mechanisms have hampered the development of an effective vaccine against HCV, among them the remarkable conformational flexibility within neutralization epitopes in the HCV antigens. Here, we report the design of epitope-focused immunogens displaying two distinct HCV cross-neutralization epitopes. We show that these immunogens induce a pronounced, broadly neutralizing antibody response in laboratory and transgenic human antibody mice. Monoclonal human antibodies isolated from immunized human antibody mice specifically recognized the grafted epitopes and neutralized four diverse HCV strains. Our results highlight a promising strategy for developing HCV immunogens and provide an encouraging paradigm for targeting structurally flexible epitopes to improve the induction of neutralizing antibodies.
Collapse
Affiliation(s)
- Kumar Nagarathinam
- Institute of Biochemistry, Center of Structural and Cell Biology in Medicine, University of Lübeck, 23562 Lübeck, Germany
| | - Andreas Scheck
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, Lausanne CH-1015, Switzerland
- Swiss Institute of Bioinformatics (SIB), Lausanne CH-1015, Switzerland
| | - Maurice Labuhn
- Institute for Experimental Virology, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, 30625 Hannover, Germany
| | - Luisa J. Ströh
- Institute of Virology, Hannover Medical School, 30625 Hannover, Germany
| | - Elisabeth Herold
- Institute of Biochemistry, Center of Structural and Cell Biology in Medicine, University of Lübeck, 23562 Lübeck, Germany
| | - Barbora Veselkova
- Institute of Biochemistry, Center of Structural and Cell Biology in Medicine, University of Lübeck, 23562 Lübeck, Germany
| | - Sarah Tune
- Department of Psychology, University of Lübeck, 23562 Lübeck, Germany
- Center of Brain, Behavior, and Metabolism, University of Lübeck, 23562 Lübeck, Germany
| | | | - Stéphane Rosset
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, Lausanne CH-1015, Switzerland
- Swiss Institute of Bioinformatics (SIB), Lausanne CH-1015, Switzerland
| | - Sabrina S. Vollers
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, Lausanne CH-1015, Switzerland
- Swiss Institute of Bioinformatics (SIB), Lausanne CH-1015, Switzerland
| | - Dorothea Bankwitz
- Institute for Experimental Virology, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, 30625 Hannover, Germany
| | - Matthias Ballmaier
- Central Research Facility Cell Sorting, Hannover Medical School, 30625 Hannover, Germany
| | - Heike Böning
- Institute of Virology, Hannover Medical School, 30625 Hannover, Germany
| | - Edith Roth
- Division of Molecular Immunology, Department of Internal Medicine 3, Friedrich-Alexander University of Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Tanvi Khera
- Institute for Experimental Virology, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, 30625 Hannover, Germany
| | | | | | - Jonas Obleser
- Department of Psychology, University of Lübeck, 23562 Lübeck, Germany
- Center of Brain, Behavior, and Metabolism, University of Lübeck, 23562 Lübeck, Germany
| | - Michael Nassal
- Department of Internal Medicine 2/Molecular Biology, University Hospital Freiburg, 79106 Freiburg, Germany
| | - Hans-Martin Jäck
- Division of Molecular Immunology, Department of Internal Medicine 3, Friedrich-Alexander University of Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Thomas Pietschmann
- Institute for Experimental Virology, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, 30625 Hannover, Germany
- German Center for Infection Research (DZIF), partner site Hannover-Braunschweig, 30625 Hannover, Germany
- Excellence Cluster 2155 RESIST, Hannover Medical School, 30625 Hannover, Germany
| | - Bruno E. Correia
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, Lausanne CH-1015, Switzerland
- Swiss Institute of Bioinformatics (SIB), Lausanne CH-1015, Switzerland
| | - Thomas Krey
- Institute of Biochemistry, Center of Structural and Cell Biology in Medicine, University of Lübeck, 23562 Lübeck, Germany
- Institute of Virology, Hannover Medical School, 30625 Hannover, Germany
- Excellence Cluster 2155 RESIST, Hannover Medical School, 30625 Hannover, Germany
- German Center for Infection Research (DZIF), partner site Hamburg-Lübeck-Borstel-Riems, 38124 Braunschweig, Germany
- Centre for Structural Systems Biology (CSSB), 22607 Hamburg, Germany
| |
Collapse
|
4
|
Oliveira Matos AD, Araujo M, Paulino J, Franco FC, Luchs A, Sales-Campos H, Fiaccadori F, Souza M, Silva-Sales M. Mutations in the main antigenic sites of VP7 and VP8* from G3P[8] rotavirus a strains circulating in Brazil may impact immune evasion to rotavirus vaccination. Braz J Microbiol 2024:10.1007/s42770-024-01542-4. [PMID: 39505807 DOI: 10.1007/s42770-024-01542-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/27/2024] [Accepted: 10/03/2024] [Indexed: 11/08/2024] Open
Abstract
In the post-rotavirus (RVA) vaccination era, uncommon and zoonotic strains have emerged as causative agents of acute gastroenteritis in humans, including the equine-like G3P[8] strains. First identified in 2013, this strain has quickly spread worldwide, reaching the position of the most prevalent genotype in many countries, including Brazil. Here, we report full genotype characterization and phylogenetic analysis of two equine-like G3P[8] strains detected in Goiás, a state in the Cerrado biome of the Brazilian Midwestern region, during the year of 2019. The strains were detected in different socioeconomic and demographic contexts: GO-MR from an asymptomatic adult living in a rural traditional community and GO-H5 from a symptomatic child from the state capital, with access to safe drinking water and essential sanitation services. These strains also displayed different backbone constellations considering the NSP2 gene segment (G3-P[8]-I2-R2-C2-M2-A2-N2-T2-E2-H2 for GO-MR and G3-P[8]-I2-R2-C2-M2-A2-N1-T2-E2-H2 for GO-H5). Furthermore, significant mutations in the main epitope sites of the VP7 and VP8* proteins of the detected strains, and other Brazilian G3P[8] viruses, were found with the comparison to RV1 and RV5 vaccine proteins, indicating a potential ability of these viruses to evade vaccine protection, which may contribute to their prevalence both nationally and globally. In summary, this study corroborates the genetic diversity of equine-like G3P[8] DS-1-like strains circulating worldwide, highlights the epidemiological importance of adults as reservoirs of RVA and shows the substantial differences between these emerging strains and the currently used anti-RVA vaccines, which may partially explain their predominance due to potential evasion of vaccine-induced protection.
Collapse
Affiliation(s)
- Amanda de Oliveira Matos
- Laboratory of Virology and Cellular Culture (LABVICC), Institute of Tropical Pathology and Public Health, Federal University of Goiás (UFG), Goiânia, Brazil
- Laboratory of Mucosal Immunology and Immunoinformatics (LIMIM), Institute of Tropical Pathology and Public Health, Federal University of Goiás (UFG), Goiânia, Brazil
| | - Maísa Araujo
- Laboratory of Virology and Cellular Culture (LABVICC), Institute of Tropical Pathology and Public Health, Federal University of Goiás (UFG), Goiânia, Brazil
| | - Jordana Paulino
- Laboratory of Virology and Cellular Culture (LABVICC), Institute of Tropical Pathology and Public Health, Federal University of Goiás (UFG), Goiânia, Brazil
| | - Fernanda Craveiro Franco
- Laboratory of Virology and Cellular Culture (LABVICC), Institute of Tropical Pathology and Public Health, Federal University of Goiás (UFG), Goiânia, Brazil
| | - Adriana Luchs
- Enteric Diseases Laboratory, Virology Center, Adolfo Lutz Institute, São Paulo, Brazil
| | - Helioswilton Sales-Campos
- Laboratory of Mucosal Immunology and Immunoinformatics (LIMIM), Institute of Tropical Pathology and Public Health, Federal University of Goiás (UFG), Goiânia, Brazil
| | - Fabiola Fiaccadori
- Laboratory of Virology and Cellular Culture (LABVICC), Institute of Tropical Pathology and Public Health, Federal University of Goiás (UFG), Goiânia, Brazil
| | - Menira Souza
- Laboratory of Virology and Cellular Culture (LABVICC), Institute of Tropical Pathology and Public Health, Federal University of Goiás (UFG), Goiânia, Brazil
| | - Marcelle Silva-Sales
- Laboratory of Virology and Cellular Culture (LABVICC), Institute of Tropical Pathology and Public Health, Federal University of Goiás (UFG), Goiânia, Brazil.
| |
Collapse
|
5
|
Jampanil N, Kumthip K, Yodmeeklin A, Tacharoenmuang R, Akari Y, Komoto S, Okitsu S, Ushijima H, Maneekarn N, Khamrin P. Unusual G3P[10] bat-like rotavirus strains detected in children with acute gastroenteritis in Thailand. J Med Virol 2024; 96:e70014. [PMID: 39420695 DOI: 10.1002/jmv.70014] [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] [Academic Contribution Register] [Received: 06/05/2024] [Revised: 09/12/2024] [Accepted: 10/08/2024] [Indexed: 10/19/2024]
Abstract
Rotavirus A (RVA) is the main cause of acute gastroenteritis among children under the age of five globally. The unusual bat-like human RVA strains G3P[10] (RVA/Human-wt/THA/CMH079/05/2005/G3P[10] and RVA/Human-wt/THA/CMH-S015-19/2019/G3P[10]) were detected in children with acute gastroenteritis in 2005 and 2019, respectively, in the same geographical area of Northern Thailand. To elucidate the genetic backgrounds of these unusual or bat-like human RVA strains, the complete genome of these RVA strains was sequenced and phylogenetically analyzed. All eleven genome segments of these G3P[10] strains were genotyped as G3-P[10]-I8-R3-C3-M3-A9-N3-T3-E3-H6, which is closely related to bat G3P[10] RVA strain (RVA/Bat-tc/CHN/MYAS33/2013/G3P[10]) and bat-like human RVA strain (RVA/Human-wt/THA/MS2015-1-0001/2015/G3P[10]). The findings indicate that human G3P[10] RVA strains detected in this study (RVA/Human-wt/THA/CMH079/05/2005/G3P[10] and RVA/Human-wt/THA/CMH-S015-19/2019/G3P[10]) contained all eleven genome segments similar to those of bat RVA strains and appeared to be human RVA strains of bat origin. Phylogenetic analysis revealed that several genome segments of these two RVA strains were also closely related with those of other species in addition to bats and had a zoonotic transmission history. The results of this study supported the roles of interspecies transmission of RVA strains among bats and humans in the natural environment and provided convincing evidence that the evolution of human RVAs was closely interrelated with those of animal RVAs. Continuing surveillance of RVAs in humans and animals is imperative to gain a better understanding of the origin and the evolution of these viruses.
Collapse
Affiliation(s)
- Nutthawadee Jampanil
- Department of Microbiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Kattareeya Kumthip
- Department of Microbiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Emerging and Re-emerging Diarrheal Viruses Cluster, Chiang Mai University, Chiang Mai, Thailand
| | - Arpaporn Yodmeeklin
- Department of Microbiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Emerging and Re-emerging Diarrheal Viruses Cluster, Chiang Mai University, Chiang Mai, Thailand
| | - Ratana Tacharoenmuang
- Department of Virology and Parasitology, Fujita Health University School of Medicine, Toyoake, Aichi, Japan
- Department of Medical Sciences, National Institute of Health, Nonthaburi, Thailand
| | - Yuki Akari
- Department of Virology and Parasitology, Fujita Health University School of Medicine, Toyoake, Aichi, Japan
- Division of One Health, Research Center for GLOBAL and LOCAL Infectious Diseases (RCGLID), Oita University, Yufu, Oita, Japan
| | - Satoshi Komoto
- Department of Virology and Parasitology, Fujita Health University School of Medicine, Toyoake, Aichi, Japan
- Division of One Health, Research Center for GLOBAL and LOCAL Infectious Diseases (RCGLID), Oita University, Yufu, Oita, Japan
| | - Shoko Okitsu
- Department of Pathology and Microbiology, Division of Microbiology, Nihon University School of Medicine, Tokyo, Japan
| | - Hiroshi Ushijima
- Department of Pathology and Microbiology, Division of Microbiology, Nihon University School of Medicine, Tokyo, Japan
| | - Niwat Maneekarn
- Department of Microbiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Emerging and Re-emerging Diarrheal Viruses Cluster, Chiang Mai University, Chiang Mai, Thailand
| | - Pattara Khamrin
- Department of Microbiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Emerging and Re-emerging Diarrheal Viruses Cluster, Chiang Mai University, Chiang Mai, Thailand
| |
Collapse
|
6
|
Uprety T, Soni S, Sreenivasan C, Hause BM, Naveed A, Ni S, Graves AJ, Morrow JK, Meade N, Mellits KH, Adam E, Kennedy MA, Wang D, Li F. Genetic and antigenic characterization of two diarrhoeicdominant rotavirus A genotypes G3P[12] and G14P[12] circulating in the global equine population. J Gen Virol 2024; 105:002016. [PMID: 39163114 PMCID: PMC11335307 DOI: 10.1099/jgv.0.002016] [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] [Academic Contribution Register] [Received: 05/31/2024] [Accepted: 07/30/2024] [Indexed: 08/21/2024] Open
Abstract
Equine rotavirus species A (ERVA) G3P[12] and G14P[12] are two dominant genotypes that cause foal diarrhoea with a significant economic impact on the global equine industry. ERVA can also serve as a source of novel (equine-like) rotavirus species A (RVA) reassortants with zoonotic potential as those identified previously in 2013-2019 when equine G3-like RVA was responsible for worldwide outbreaks of severe gastroenteritis and hospitalizations in children. One hurdle to ERVA research is that the standard cell culture system optimized for human rotavirus replication is not efficient for isolating ERVA. Here, using an engineered cell line defective in antiviral innate immunity, we showed that both equine G3P[12] and G14P[12] strains can be rapidly isolated from diarrhoeic foals. The genome sequence analysis revealed that both G3P[12] and G14P[12] strains share the identical genotypic constellation except for VP7 and VP6 segments in which G3P[12] possessed VP7 of genotype G3 and VP6 of genotype I6 and G14P[12] had the combination of VP7 of genotype G14 and VP6 of genotype I2. Further characterization demonstrated that two ERVA genotypes have a limited cross-neutralization. The lack of an in vitro broad cross-protection between both genotypes supported the increased recent diarrhoea outbreaks due to equine G14P[12] in foals born to dams immunized with the inactivated monovalent equine G3P[12] vaccine. Finally, using the structural modelling approach, we provided the genetic basis of the antigenic divergence between ERVA G3P[12] and G14P[12] strains. The results of this study will provide a framework for further investigation of infection biology, pathogenesis and cross-protection of equine rotaviruses.
Collapse
Affiliation(s)
- Tirth Uprety
- Department of Veterinary Science, Maxwell H. Gluck Equine Research Center, University of Kentucky, Lexington, Kentucky 40546, USA
| | - Shalini Soni
- Department of Veterinary Science, Maxwell H. Gluck Equine Research Center, University of Kentucky, Lexington, Kentucky 40546, USA
| | - Chithra Sreenivasan
- Department of Veterinary Science, Maxwell H. Gluck Equine Research Center, University of Kentucky, Lexington, Kentucky 40546, USA
| | - Ben M. Hause
- Department of Veterinary and Biomedical Sciences, Animal Disease Research and Diagnostic Laboratory, South Dakota State University, Brookings, South Dakota, 57007, USA
| | - Ahsan Naveed
- Department of Veterinary Science, Maxwell H. Gluck Equine Research Center, University of Kentucky, Lexington, Kentucky 40546, USA
| | - Shuisong Ni
- Department of Chemistry and Biochemistry, Miami University, Oxford, OH 45056, USA
| | - Amy J. Graves
- Equine Diagnostic Solutions, LLC, 1501 Bull Lea Rd, Suite 104, Lexington, Kentucky 40511, USA
| | - Jennifer K. Morrow
- Equine Diagnostic Solutions, LLC, 1501 Bull Lea Rd, Suite 104, Lexington, Kentucky 40511, USA
| | - Nathan Meade
- Division of Microbiology, Brewing, and Biotechnology, School of Biosciences, University of Nottingham, Sutton Bonington, UK
| | - Kenneth H. Mellits
- Division of Microbiology, Brewing, and Biotechnology, School of Biosciences, University of Nottingham, Sutton Bonington, UK
| | - Emma Adam
- Department of Veterinary Science, Maxwell H. Gluck Equine Research Center, University of Kentucky, Lexington, Kentucky 40546, USA
| | - Michael A. Kennedy
- Department of Veterinary and Biomedical Sciences, Animal Disease Research and Diagnostic Laboratory, South Dakota State University, Brookings, South Dakota, 57007, USA
| | - Dan Wang
- Department of Veterinary Science, Maxwell H. Gluck Equine Research Center, University of Kentucky, Lexington, Kentucky 40546, USA
| | - Feng Li
- Department of Veterinary Science, Maxwell H. Gluck Equine Research Center, University of Kentucky, Lexington, Kentucky 40546, USA
| |
Collapse
|
7
|
Sadiq A, Khan T, Bostan N, Yinda CK, Matthijnssens J. Antigenic epitope analysis of Pakistani G3 and G9 rotavirus strains compared to vaccine strains revealed multiple amino acid differences. Diagn Microbiol Infect Dis 2024; 109:116346. [PMID: 38759540 DOI: 10.1016/j.diagmicrobio.2024.116346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/27/2022] [Revised: 08/13/2023] [Accepted: 05/08/2024] [Indexed: 05/19/2024]
Abstract
Rotaviruses belong to genotype VP4-P[8] are a significant cause of severe loose diarrhea in infants and young children. In the present study, we characterised the complete genome of three of the Pakistani P[8]b RVA strains by Illumina HiSeq sequencing technology to determine the complete genotype constellation providing insight into the evolutionary dynamics of their genes using maximum likelihood analysis. The maximum genomic sequences of our study strains were similar to more recent human Wa-Like G1P[8]a, G3P[8]a, G4P[6], G4P[8], G9P[4], G9P[8]a, G11P[25],G12P[8]a and G12P[6] strains circulating around the world. Therefore, strains PAK274, PAK439 and PAK624 carry natively distinctive VP4 gene with universally common human Wa-Like genetic backbone. Comparing our study P[8]b strains with vaccines strains RotarixTM and RotaTeqTM, multiple amino acid differences were examined between vaccine virus antigenic epitopes and Pakistani isolates. Over time, these differences may result in the selection for strains that will escape the vaccine-induced RVA-neutralizing-antibody effect.
Collapse
Affiliation(s)
- Asma Sadiq
- Department of Microbiology, University of Jhang, Jhang, Pakistan
| | - Tariq Khan
- Department of Biosciences, COMSATS University (CUI), Park Road, Tarlai Kalan, Chak Shahzad, Islamabad,45550, Pakistan
| | - Nazish Bostan
- Department of Biosciences, COMSATS University (CUI), Park Road, Tarlai Kalan, Chak Shahzad, Islamabad,45550, Pakistan.
| | - Claude Kwe Yinda
- KU Leuven-University of Leuven, Department of Microbiology and Immunology, Rega Institute for Medical Research, Laboratory of Viral Metagenomics, Leuven, Belgium
| | - Jelle Matthijnssens
- KU Leuven-University of Leuven, Department of Microbiology and Immunology, Rega Institute for Medical Research, Laboratory of Viral Metagenomics, Leuven, Belgium
| |
Collapse
|
8
|
Wu L, Jing Z, Pan Y, Guo L, Li Z, Feng L, Tian J. Emergence of a novel pathogenic porcine G1P[7] rotavirus in China. Virology 2024; 598:110185. [PMID: 39096775 DOI: 10.1016/j.virol.2024.110185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/14/2024] [Revised: 07/07/2024] [Accepted: 07/20/2024] [Indexed: 08/05/2024]
Abstract
Among group A rotaviruses (RVAs), the G1 genotype is the main genotype causing diarrhea in children, but it has rarely been reported in pigs. During our epidemiological investigation, we detected G1P[7] rotavirus infection in piglets across several provinces in China and then isolated a porcine G1P[7] rotavirus strain (CN1P7). Sequencing revealed that the virus constellation was G1-P[7]-I5-R1-C1-M1-A8-N1-T1-E1-H1. Phylogenetic analyses revealed that CN1P7 most likely emerged due to genetic reassortment among porcine, human, giant panda and dog rotavirus strains. In vivo experiments were conducted on two-day-old piglets, which revealed that the CN1P7 strain was pathogenic to piglets. The virus was shed through the digestive tract and respiratory tract. In addition to the intestine, the CN1P7 strain displayed extraintestinal tropisms in piglets. Histopathological analysis revealed that the lung and small intestine were the targets of CN1P7. This study is the first to explore the molecular and pathogenic characterization of a pig-origin G1P[7] rotavirus.
Collapse
Affiliation(s)
- Ling Wu
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People's Republic of China
| | - Zhaoyang Jing
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People's Republic of China
| | - Yudi Pan
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People's Republic of China
| | - Longjun Guo
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People's Republic of China
| | - Zixin Li
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People's Republic of China
| | - Li Feng
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People's Republic of China.
| | - Jin Tian
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People's Republic of China.
| |
Collapse
|
9
|
Fukuda Y, Kondo K, Nakata S, Morita Y, Adachi N, Kogawa K, Ukae S, Kudou Y, Adachi S, Yamamoto M, Fukumura S, Tsugawa T. Whole-genome analysis of human group A rotaviruses in 1980s Japan and evolutionary assessment of global Wa-like strains across half a century. J Gen Virol 2024; 105. [PMID: 38836747 DOI: 10.1099/jgv.0.001998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 06/06/2024] Open
Abstract
Historically, the Wa-like strains of human group A rotavirus (RVA) have been major causes of gastroenteritis. However, since the 2010s, the circulation of non-Wa-like strains has been increasingly reported, indicating a shift in the molecular epidemiology of RVA. Although understanding RVA evolution requires the analysis of both current and historical strains, comprehensive pre-1980's sequencing data are scarce globally. We determined the whole-genome sequences of representative strains from six RVA gastroenteritis outbreaks observed at an infant home in Sapporo, Japan, between 1981 and 1989. These outbreaks were mainly caused by G1 or G3 Wa-like strains, resembling strains from the United States in the 1970s-1980s and from Malawi in the 1990s. Phylogenetic analysis of these infant home strains, together with Wa-like strains collected worldwide from the 1970s to 2020, revealed a notable trend: pre-2010 strains diverged into multiple lineages in many genomic segments, whereas post-2010 strains tended to converge into a single lineage. However, Bayesian skyline plot indicated near-constant effective population sizes from the 1970s to 2020, and selection pressure analysis identified positive selection only at amino acid 75 of NSP2. These results suggest that evidence supporting the influence of rotavirus vaccines, introduced globally since 2006, on Wa-like RVA molecular evolution is lacking at present, and phylogenetic analysis may simply reflect natural fluctuations in RVA molecular evolution. Evaluating the long-term impact of RV vaccines on the molecular evolution of RVA requires sustained surveillance.
Collapse
Affiliation(s)
- Yuya Fukuda
- Department of Pediatrics, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Kenji Kondo
- Department of Pediatrics, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Shuji Nakata
- Department of Pediatrics, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Yasuyuki Morita
- Department of Pediatrics, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Noriaki Adachi
- Department of Pediatrics, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Keiko Kogawa
- Department of Pediatrics, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Susumu Ukae
- Department of Pediatrics, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Yoshimasa Kudou
- Department of Pediatrics, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Shuhei Adachi
- Department of Pediatrics, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Masaki Yamamoto
- Department of Pediatrics, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Shinobu Fukumura
- Department of Pediatrics, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Takeshi Tsugawa
- Department of Pediatrics, Sapporo Medical University School of Medicine, Sapporo, Japan
| |
Collapse
|
10
|
Yarmohammadi H, Aghasadeghi M, Akhavan Sepahi A, Hamidi-fard M, Bahramali G. Designing the fusion protein of rotavirus VP8 and hepatitis A virus VP1 and evaluating the immunological response in BALB/c mice. IRANIAN JOURNAL OF MICROBIOLOGY 2024; 16:401-410. [PMID: 39005596 PMCID: PMC11245353 DOI: 10.18502/ijm.v16i3.15797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Academic Contribution Register] [Indexed: 07/16/2024]
Abstract
Background and Objectives Rotavirus and Hepatitis A virus are responsible for causing gastroenteritis and jaundice. The current vaccination approaches have proven insufficient, especially in low-income countries. In this study, we presented a novel dual-vaccine candidate that combines the rotavirus VP8 protein and the hepatitis A virus VP1. Materials and Methods The VP8*-rotavirus+AAY+HAV-VP1 fusion protein was produced using an Escherichia coli expression system. The recombinant protein had a molecular weight of approximately 45.5 kDa and was purified through affinity chromatography. BALB/c mice were injected subcutaneously with the recombinant protein, VP1, VP8 and vaccines for rotavirus and hepatitis A virus, both with and without ALUM and M720 adjuvants. ELISA assays were used to measure total IgG, IgG1, IgG2, and short-term and long-term IL-5 and IFN-γ responses. Results The fusion protein, when combined with adjuvants, elicited significantly higher total IgG, IgG1, and IgG2 responses compared to VP1 and VP8 alone, as well as the rotavirus and hepatitis A vaccines. Furthermore, it induced a higher short-term IL-5 and IFN-γ response while demonstrating a higher long-term IL-5 response compared to the rotavirus and hepatitis A vaccines. Conclusion This study demonstrates that the VP8*-rotavirus+AAY+HAV-VP1 fusion protein is a promising dual vaccine candidate for immunization against hepatitis A and rotaviruses.
Collapse
Affiliation(s)
- Hassan Yarmohammadi
- Department of Microbiology, North Tehran Branch, Islamic Azad University, Tehran, Iran
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran
| | | | - Abbas Akhavan Sepahi
- Department of Microbiology, North Tehran Branch, Islamic Azad University, Tehran, Iran
| | | | - Golnaz Bahramali
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
11
|
Vetter J, Lee M, Eichwald C. The Role of the Host Cytoskeleton in the Formation and Dynamics of Rotavirus Viroplasms. Viruses 2024; 16:668. [PMID: 38793550 PMCID: PMC11125917 DOI: 10.3390/v16050668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/24/2024] [Revised: 04/22/2024] [Accepted: 04/23/2024] [Indexed: 05/26/2024] Open
Abstract
Rotavirus (RV) replicates within viroplasms, membraneless electron-dense globular cytosolic inclusions with liquid-liquid phase properties. In these structures occur the virus transcription, replication, and packaging of the virus genome in newly assembled double-layered particles. The viroplasms are composed of virus proteins (NSP2, NSP5, NSP4, VP1, VP2, VP3, and VP6), single- and double-stranded virus RNAs, and host components such as microtubules, perilipin-1, and chaperonins. The formation, coalescence, maintenance, and perinuclear localization of viroplasms rely on their association with the cytoskeleton. A stabilized microtubule network involving microtubules and kinesin Eg5 and dynein molecular motors is associated with NSP5, NSP2, and VP2, facilitating dynamic processes such as viroplasm coalescence and perinuclear localization. Key post-translation modifications, particularly phosphorylation events of RV proteins NSP5 and NSP2, play pivotal roles in orchestrating these interactions. Actin filaments also contribute, triggering the formation of the viroplasms through the association of soluble cytosolic VP4 with actin and the molecular motor myosin. This review explores the evolving understanding of RV replication, emphasizing the host requirements essential for viroplasm formation and highlighting their dynamic interplay within the host cell.
Collapse
Affiliation(s)
| | | | - Catherine Eichwald
- Institute of Virology, University of Zurich, 8057 Zurich, Switzerland; (J.V.); (M.L.)
| |
Collapse
|
12
|
de Sautu M, Herrmann T, Scanavachi G, Jenni S, Harrison SC. The rotavirus VP5*/VP8* conformational transition permeabilizes membranes to Ca2. PLoS Pathog 2024; 20:e1011750. [PMID: 38574119 PMCID: PMC11020617 DOI: 10.1371/journal.ppat.1011750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/16/2023] [Revised: 04/16/2024] [Accepted: 03/04/2024] [Indexed: 04/06/2024] Open
Abstract
Rotaviruses infect cells by delivering into the cytosol a transcriptionally active inner capsid particle (a "double-layer particle": DLP). Delivery is the function of a third, outer layer, which drives uptake from the cell surface into small vesicles from which the DLPs escape. In published work, we followed stages of rhesus rotavirus (RRV) entry by live-cell imaging and correlated them with structures from cryogenic electron microscopy and tomography (cryo-EM and cryo-ET). The virus appears to wrap itself in membrane, leading to complete engulfment and loss of Ca2+ from the vesicle produced by the wrapping. One of the outer-layer proteins, VP7, is a Ca2+-stabilized trimer; loss of Ca2+ releases both VP7 and the other outer-layer protein, VP4, from the particle. VP4, activated by cleavage into VP8* and VP5*, is a trimer that undergoes a large-scale conformational rearrangement, reminiscent of the transition that viral fusion proteins undergo to penetrate a membrane. The rearrangement of VP5* thrusts a 250-residue, C-terminal segment of each of the three subunits outward, while allowing the protein to remain attached to the virus particle and to the cell being infected. We proposed that this segment inserts into the membrane of the target cell, enabling Ca2+ to cross. In the work reported here, we show the validity of key aspects of this proposed sequence. By cryo-EM studies of liposome-attached virions ("triple-layer particles": TLPs) and single-particle fluorescence imaging of liposome-attached TLPs, we confirm insertion of the VP4 C-terminal segment into the membrane and ensuing generation of a Ca2+ "leak". The results allow us to formulate a molecular description of early events in entry. We also discuss our observations in the context of other work on double-strand RNA virus entry.
Collapse
Affiliation(s)
- Marilina de Sautu
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts, United States of America
- Laboratory of Molecular Medicine, Boston Children’s Hospital, Boston, Massachusetts, United States of America
| | - Tobias Herrmann
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Gustavo Scanavachi
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts, United States of America
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, Massachusetts, United States of America
| | - Simon Jenni
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Stephen C. Harrison
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts, United States of America
- Laboratory of Molecular Medicine, Boston Children’s Hospital, Boston, Massachusetts, United States of America
- Howard Hughes Medical Institute, Harvard Medical School, Boston, Massachusetts, United States of America
| |
Collapse
|
13
|
He Y, Zhou J, Gao H, Liu C, Zhan P, Liu X. Broad-spectrum antiviral strategy: Host-targeting antivirals against emerging and re-emerging viruses. Eur J Med Chem 2024; 265:116069. [PMID: 38160620 DOI: 10.1016/j.ejmech.2023.116069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/03/2023] [Revised: 12/06/2023] [Accepted: 12/16/2023] [Indexed: 01/03/2024]
Abstract
Viral infections are amongst the most prevalent diseases that pose a significant threat to human health. Targeting viral proteins or host factors represents two primary strategies for the development of antiviral drugs. In contrast to virus-targeting antivirals (VTAs), host-targeting antivirals (HTAs) offer advantages in terms of overcoming drug resistance and effectively combating a wide range of viruses, including newly emerging ones. Therefore, targeting host factors emerges as an extremely promising strategy with the potential to address critical challenges faced by VTAs. In recent years, extensive research has been conducted on the discovery and development of HTAs, leading to the approval of maraviroc, a chemokine receptor type 5 (CCR5) antagonist used for the treatment of HIV-1 infected individuals, with several other potential treatments in various stages of development for different viral infections. This review systematically summarizes advancements made in medicinal chemistry regarding various host targets and classifies them into four distinct catagories based on their involvement in the viral life cycle: virus attachment and entry, biosynthesis, nuclear import and export, and viral release.
Collapse
Affiliation(s)
- Yong He
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology, Ministry of Education, School of Pharmaceutical Sciences, Shandong University, Ji'nan, 250012, Shandong Province, PR China
| | - Jiahui Zhou
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology, Ministry of Education, School of Pharmaceutical Sciences, Shandong University, Ji'nan, 250012, Shandong Province, PR China
| | - Huizhan Gao
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology, Ministry of Education, School of Pharmaceutical Sciences, Shandong University, Ji'nan, 250012, Shandong Province, PR China
| | - Chuanfeng Liu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology, Ministry of Education, School of Pharmaceutical Sciences, Shandong University, Ji'nan, 250012, Shandong Province, PR China
| | - Peng Zhan
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology, Ministry of Education, School of Pharmaceutical Sciences, Shandong University, Ji'nan, 250012, Shandong Province, PR China.
| | - Xinyong Liu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology, Ministry of Education, School of Pharmaceutical Sciences, Shandong University, Ji'nan, 250012, Shandong Province, PR China.
| |
Collapse
|
14
|
Carossino M, Vissani MA, Barrandeguy ME, Balasuriya UBR, Parreño V. Equine Rotavirus A under the One Health Lens: Potential Impacts on Public Health. Viruses 2024; 16:130. [PMID: 38257830 PMCID: PMC10819593 DOI: 10.3390/v16010130] [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] [Academic Contribution Register] [Received: 12/15/2023] [Revised: 12/29/2023] [Accepted: 01/12/2024] [Indexed: 01/24/2024] Open
Abstract
Group A rotaviruses are a well-known cause of viral gastroenteritis in infants and children, as well as in many mammalian species and birds, affecting them at a young age. This group of viruses has a double-stranded, segmented RNA genome with high genetic diversity linked to point mutations, recombination, and, importantly, reassortment. While initial molecular investigations undertaken in the 1900s suggested host range restriction among group A rotaviruses based on the fact that different gene segments were distributed among different animal species, recent molecular surveillance and genome constellation genotyping studies conducted by the Rotavirus Classification Working Group (RCWG) have shown that animal rotaviruses serve as a source of diversification of human rotavirus A, highlighting their zoonotic potential. Rotaviruses occurring in various animal species have been linked with contributing genetic material to human rotaviruses, including horses, with the most recent identification of equine-like G3 rotavirus A infecting children. The goal of this article is to review relevant information related to rotavirus structure/genomic organization, epidemiology (with a focus on human and equine rotavirus A), evolution, inter-species transmission, and the potential zoonotic role of equine and other animal rotaviruses. Diagnostics, surveillance and the current status of human and livestock vaccines against RVA are also reviewed.
Collapse
Affiliation(s)
- Mariano Carossino
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA;
- Louisiana Animal Disease Diagnostic Laboratory, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Maria Aldana Vissani
- Escuela de Veterinaria, Facultad de Ciencias Agrarias y Veterinarias, Universidad del Salvador, Pilar, Buenos Aires B1630AHU, Argentina; (M.A.V.); (M.E.B.)
- Instituto de Virología, CICVyA, Instituto Nacional de Tecnología Agropecuaria (INTA), Buenos Aires B1686LQF, Argentina;
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires C1033AAJ, Argentina
| | - Maria E. Barrandeguy
- Escuela de Veterinaria, Facultad de Ciencias Agrarias y Veterinarias, Universidad del Salvador, Pilar, Buenos Aires B1630AHU, Argentina; (M.A.V.); (M.E.B.)
- Instituto de Virología, CICVyA, Instituto Nacional de Tecnología Agropecuaria (INTA), Buenos Aires B1686LQF, Argentina;
| | - Udeni B. R. Balasuriya
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA;
- Louisiana Animal Disease Diagnostic Laboratory, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Viviana Parreño
- Instituto de Virología, CICVyA, Instituto Nacional de Tecnología Agropecuaria (INTA), Buenos Aires B1686LQF, Argentina;
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires C1033AAJ, Argentina
| |
Collapse
|
15
|
Potgieter RL, Mwangi PN, Mogotsi MT, Uwimana J, Mutesa L, Muganga N, Murenzi D, Tusiyenge L, Seheri ML, Steele AD, Mwenda JM, Nyaga MM. Genomic Analysis of Rwandan G9P[8] Rotavirus Strains Pre- and Post-RotaTeq ® Vaccine Reveals Significant Distinct Sub-Clustering in a Post-Vaccination Cohort. Viruses 2023; 15:2321. [PMID: 38140562 PMCID: PMC10747556 DOI: 10.3390/v15122321] [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] [Academic Contribution Register] [Received: 10/19/2023] [Revised: 11/16/2023] [Accepted: 11/23/2023] [Indexed: 12/24/2023] Open
Abstract
Although the introduction of rotavirus vaccines has substantially contributed to the reduction in rotavirus morbidity and mortality, concerns persist about the re-emergence of variant strains that might alter vaccine effectiveness in the long term. The G9 strains re-emerged in Africa during the mid-1990s and have more recently become predominant in some countries, such as Ghana and Zambia. In Rwanda, during the 2011 to 2015 routine surveillance period, G9P[8] persisted during both the pre- and post-vaccine periods. The pre-vaccination cohort was based on the surveillance period of 2011 to 2012, and the post-vaccination cohort was based on the period of 2013 to 2015, excluding 2014. The RotaTeq® vaccine that was first introduced in Rwanda in 2012 is genotypically heterologous to Viral Protein 7 (VP7) G9. This study elucidated the whole genome of Rwandan G9P[8] rotavirus strains pre- and post-RotaTeq® vaccine introduction. Fecal samples from Rwandan children under the age of five years (pre-vaccine n = 23; post-vaccine n = 7), conventionally genotyped and identified as G9P[8], were included. Whole-genome sequencing was then performed using the Illumina® MiSeq platform. Phylogenetic analysis and pair-wise sequence analysis were performed using MEGA6 software. Distinct clustering of three post-vaccination study strains was observed in all 11 gene segments, compared to the other Rwandan G9P[8] study strains. Specific amino acid differences were identified across the gene segments of these three 2015 post-vaccine strains. Important amino acid differences were identified at position N242S in the VP7 genome segment of the three post-vaccine G9 strains compared to the other G9 strains. This substitution occurs at a neutralization epitope site and may slightly affect protein interaction at that position. These findings indicate that the Rwandan G9P[8] strains revealed a distinct sub-clustering pattern among post-vaccination study strains circulating in Rwanda, with changes at neutralization epitopes, which may play a role in neutralization escape from vaccine candidates. This emphasizes the need for continuous whole-genome surveillance to better understand the evolution and epidemiology of the G9P[8] strains post-vaccination.
Collapse
Affiliation(s)
- Robyn-Lee Potgieter
- Next Generation Sequencing Unit and Division of Virology, Faculty of Health Sciences, University of the Free State, Bloemfontein 9300, South Africa; (R.-L.P.); (P.N.M.); (M.T.M.)
| | - Peter N. Mwangi
- Next Generation Sequencing Unit and Division of Virology, Faculty of Health Sciences, University of the Free State, Bloemfontein 9300, South Africa; (R.-L.P.); (P.N.M.); (M.T.M.)
| | - Milton T. Mogotsi
- Next Generation Sequencing Unit and Division of Virology, Faculty of Health Sciences, University of the Free State, Bloemfontein 9300, South Africa; (R.-L.P.); (P.N.M.); (M.T.M.)
| | - Jeannine Uwimana
- Department of Pediatrics, Kigali University Teaching Hospital, College of Medicine and Health Sciences, University of Rwanda, Kigali P.O. Box 4285, Rwanda; (J.U.); (L.M.); (N.M.); (D.M.); (L.T.)
| | - Leon Mutesa
- Department of Pediatrics, Kigali University Teaching Hospital, College of Medicine and Health Sciences, University of Rwanda, Kigali P.O. Box 4285, Rwanda; (J.U.); (L.M.); (N.M.); (D.M.); (L.T.)
- Centre for Human Genetics, College of Medicine and Health Sciences, University of Rwanda, Kigali P.O. Box 4285, Rwanda
| | - Narcisse Muganga
- Department of Pediatrics, Kigali University Teaching Hospital, College of Medicine and Health Sciences, University of Rwanda, Kigali P.O. Box 4285, Rwanda; (J.U.); (L.M.); (N.M.); (D.M.); (L.T.)
| | - Didier Murenzi
- Department of Pediatrics, Kigali University Teaching Hospital, College of Medicine and Health Sciences, University of Rwanda, Kigali P.O. Box 4285, Rwanda; (J.U.); (L.M.); (N.M.); (D.M.); (L.T.)
| | - Lisine Tusiyenge
- Department of Pediatrics, Kigali University Teaching Hospital, College of Medicine and Health Sciences, University of Rwanda, Kigali P.O. Box 4285, Rwanda; (J.U.); (L.M.); (N.M.); (D.M.); (L.T.)
| | - Mapaseka L. Seheri
- Diarrheal Pathogens Research Unit, Sefako Makgatho Health Sciences University, Medunsa, Pretoria 0204, South Africa; (M.L.S.); (A.D.S.)
| | - A. Duncan Steele
- Diarrheal Pathogens Research Unit, Sefako Makgatho Health Sciences University, Medunsa, Pretoria 0204, South Africa; (M.L.S.); (A.D.S.)
| | - Jason M. Mwenda
- World Health Organization, Regional Office for Africa, Brazzaville P.O. Box 06, Congo;
| | - Martin M. Nyaga
- Next Generation Sequencing Unit and Division of Virology, Faculty of Health Sciences, University of the Free State, Bloemfontein 9300, South Africa; (R.-L.P.); (P.N.M.); (M.T.M.)
| |
Collapse
|
16
|
Latifi T, Jalilvand S, Golsaz-Shirazi F, Arashkia A, Kachooei A, Afchangi A, Zafarian S, Roohvand F, Shoja Z. Characterization and immunogenicity of a novel chimeric hepatitis B core-virus like particles (cVLPs) carrying rotavirus VP8*protein in mice model. Virology 2023; 588:109903. [PMID: 37832344 DOI: 10.1016/j.virol.2023.109903] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/12/2023] [Revised: 09/23/2023] [Accepted: 10/02/2023] [Indexed: 10/15/2023]
Abstract
Given the efficacy and safety issues of the WHO for approved/prequalified live attenuated rotavirus (RV) vaccines, studies on alternative non-replicating modals and proper RV antigens are actively undertaken. Herein, we report the novel chimeric hepatitis B core-virus like particles (VLPs) carrying RV VP8*26-231 protein of a P [8] strain (cVLPVP8*), as a parenteral VLP RV vaccine candidate. SDS-PAGE and Western blotting analyses indicated the expected size of the E. coli-derived HBc-VP8* protein that self-assembled to cVLPVP8* particles. Immunization in mice indicated development of higher levels of IgG and IgA as well as higher IgG1/IgG2a ratios by cVLPVP8* vaccination compared to the VP8* alone. Assessment of neutralizing antibodies (nAbs) indicated development of heterotypic nAbs with cross-reactivity to a heterotypic RV strain by cVLPVP8* immunization compared to VP8* alone. The observed anti-VP8* cross-reactivity might indicate the possibility of developing a Pan-genomic RVA vaccine based on the cVLPVP8* formulation that deserves further challenge studies.
Collapse
Affiliation(s)
- Tayebeh Latifi
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran; Department of Virology, Pasteur Institute of Iran, Tehran, Iran
| | - Somayeh Jalilvand
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Forough Golsaz-Shirazi
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Arash Arashkia
- Department of Virology, Pasteur Institute of Iran, Tehran, Iran; Research Center for Emerging and Reemerging Infectious Diseases, Pasteur Institute of Iran, Tehran, Iran
| | - Atefeh Kachooei
- Department of Virology, Pasteur Institute of Iran, Tehran, Iran; Department of Virology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Atefeh Afchangi
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran; Department of Virology, Pasteur Institute of Iran, Tehran, Iran
| | - Saman Zafarian
- Department of Virology, Pasteur Institute of Iran, Tehran, Iran; Department of Microbial Biotechnology, College of Science, University of Tehran, Tehran, Iran
| | - Farzin Roohvand
- Department of Virology, Pasteur Institute of Iran, Tehran, Iran
| | - Zabihollah Shoja
- Department of Virology, Pasteur Institute of Iran, Tehran, Iran; Research Center for Emerging and Reemerging Infectious Diseases, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
17
|
Asensio-Cob D, Rodríguez JM, Luque D. Rotavirus Particle Disassembly and Assembly In Vivo and In Vitro. Viruses 2023; 15:1750. [PMID: 37632092 PMCID: PMC10458742 DOI: 10.3390/v15081750] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/30/2023] [Revised: 08/11/2023] [Accepted: 08/14/2023] [Indexed: 08/27/2023] Open
Abstract
Rotaviruses (RVs) are non-enveloped multilayered dsRNA viruses that are major etiologic agents of diarrheal disease in humans and in the young in a large number of animal species. The viral particle is composed of three different protein layers that enclose the segmented dsRNA genome and the transcriptional complexes. Each layer defines a unique subparticle that is associated with a different phase of the replication cycle. Thus, while single- and double-layered particles are associated with the intracellular processes of selective packaging, genome replication, and transcription, the viral machinery necessary for entry is located in the third layer. This modular nature of its particle allows rotaviruses to control its replication cycle by the disassembly and assembly of its structural proteins. In this review, we examine the significant advances in structural, molecular, and cellular RV biology that have contributed during the last few years to illuminating the intricate details of the RV particle disassembly and assembly processes.
Collapse
Affiliation(s)
- Dunia Asensio-Cob
- Department of Molecular Medicine, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, 686 Bay Street, Toronto, ON M5G0A4, Canada;
| | - Javier M. Rodríguez
- Department of Structure of Macromolecules, Centro Nacional de Biotecnología/CSIC, Cantoblanco, 28049 Madrid, Spain
| | - Daniel Luque
- Electron Microscopy Unit UCCT/ISCIII, 28220 Majadahonda, Spain
- School of Biomedical Sciences, The University of New South Wales, Sydney, NSW 2052, Australia
- Electron Microscope Unit, Mark Wainwright Analytical Centre, The University of New South Wales, Sydney, NSW 2052, Australia
| |
Collapse
|
18
|
Tahar AS, Ong EJ, Rahardja A, Mamora D, Lim KT, Ahmed K, Kulai D, Tan CS. Emergence of equine-like G3 and porcine-like G9 rotavirus strains in Sarawak, Malaysia: 2019-2021. J Med Virol 2023; 95:e28987. [PMID: 37501648 DOI: 10.1002/jmv.28987] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/04/2023] [Revised: 07/10/2023] [Accepted: 07/13/2023] [Indexed: 07/29/2023]
Abstract
Rotavirus is the leading causative viral agent of pediatric acute gastroenteritis globally, infecting mostly children 5 years old and below. Data on rotavirus prevalence in Malaysia is scarce, despite the WHO's recommendation for continuous rotavirus surveillance, and has underestimated the need for national rotavirus vaccination. Characteristics of the current rotavirus strains in Malaysia have to be determined to understand the rotavirus epidemiology and vaccine compatibility. This study sought to determine the genetic relatedness of Sarawak rotavirus strains with global strains and to determine the antigenic coverage and epitope compatibility of Rotarix and RotaTeq vaccines with the Sarawak rotavirus strains via in silico analysis. A total of 89 stool samples were collected from pediatric patients (<5 years old) with acute gastroenteritis at private hospitals in Kuching, Sarawak. Rotavirus was detected using reverse transcription-polymerase chain reaction. Positive amplicons were analyzed using nucleotide sequencing before phylogenetic analyses and assessment of epitope compatibility. Genotyping revealed G1P[8] (1/13; 7.7%), G3P[8] (3/13; 23%), G9P[4] (1/13; 7.7%), and G9P[8] (3/13; 23%), G9P[X] (1/13; 7.7%), GXP[4] (1/13; 7.7%), and GXP[8] (3/13; 23%) in samples. All wild-type Sarawak rotavirus strains, with the exception of G1, showed variations in their phylogenetic and antigenic epitope characteristics.
Collapse
Affiliation(s)
- Ahmad Syatir Tahar
- Centre for Tropical and Emerging Diseases, Faculty of Medicine and Health Sciences, Universiti Malaysia Sarawak, Kota Samarahan, Sarawak, Malaysia
| | - Eng Joe Ong
- Borneo Medical Centre, Kuching, Sarawak, Malaysia
| | | | - Dewi Mamora
- Borneo Medical Centre, Kuching, Sarawak, Malaysia
| | | | - Kamruddin Ahmed
- Department of Pathobiology and Medical Diagnostics, Faculty of Medicine and Health Sciences, Universiti Malaysia Sabah, Kota Kinabalu, Sabah, Malaysia
- Borneo Medical and Health Research Centre, Faculty of Medicine and Health Sciences, Universiti Malaysia Sabah, Kota Kinabalu, Sabah, Malaysia
| | - Dorothy Kulai
- Universiti Teknologi Mara Sarawak, Kota Samarahan, Sarawak, Malaysia
| | - Cheng Siang Tan
- Centre for Tropical and Emerging Diseases, Faculty of Medicine and Health Sciences, Universiti Malaysia Sarawak, Kota Samarahan, Sarawak, Malaysia
| |
Collapse
|
19
|
Peng R, Li D, Wang J, Xiong G, Wang M, Liu D, Wei Y, Pang L, Sun X, Li H, Kong X, Shahar S, Duan Z. Reassortment and genomic analysis of a G9P[8]-E2 rotavirus isolated in China. Virol J 2023; 20:135. [PMID: 37349792 PMCID: PMC10286334 DOI: 10.1186/s12985-023-02064-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/30/2023] [Accepted: 05/07/2023] [Indexed: 06/24/2023] Open
Abstract
OBJECTIVE To isolate a prevalent G9P[8] group A rotavirus (RVA) (N4006) in China and investigate its genomic and evolutionary characteristics, with the goal of facilitating the development of a new rotavirus vaccine. METHODS The RVA G9P[8] genotype from a diarrhea sample was passaged in MA104 cells. The virus was evaluated by TEM, polyacrylamide gel electrophoresis, and indirect immunofluorescence assay. The complete genome of virus was obtained by RT-PCR and sequencing. The genomic and evolutionary characteristics of the virus were evaluated by nucleic acid sequence analysis with MEGA ver. 5.0.5 and DNASTAR software. The neutralizing epitopes of VP7 and VP4 (VP5* and VP8*) were analyzed using BioEdit ver. 7.0.9.0 and PyMOL ver. 2.5.2. RESULTS The RVA N4006 (G9P[8] genotype) was adapted in MA104 cells with a high titer (105.5 PFU/mL). Whole-genome sequence analysis showed N4006 to be a reassortant rotavirus of Wa-like G9P[8] RVA and the NSP4 gene of DS-1-like G2P[4] RVA, with the genotype constellation G9-P[8]-I1-R1-C1-M1-A1-N1-T1-E2-H1 (G9P[8]-E2). Phylogenetic analysis indicated that N4006 had a common ancestor with Japanese G9P[8]-E2 rotavirus. Neutralizing epitope analysis showed that VP7, VP5*, and VP8* of N4006 had low homology with vaccine viruses of the same genotype and marked differences with vaccine viruses of other genotypes. CONCLUSION The RVA G9P[8] genotype with the G9-P[8]-I1-R1-C1-M1-A1-N1-T1-E2-H1 (G9P[8]-E2) constellation predominates in China and may originate from reassortment between Japanese G9P[8] with Japanese DS-1-like G2P[4] rotaviruses. The antigenic variation of N4006 with the vaccine virus necessitates an evaluation of the effect of the rotavirus vaccine on G9P[8]-E2 genotype rotavirus.
Collapse
Affiliation(s)
- Rui Peng
- Department of Biosciences, Faculty of Sciences, Universiti Teknologi Malaysia, Johor Bahru, 81310 Malaysia
- NHC Key Laboratory of Medical Viruses and Viral Diseases, Institute of Viral Disease Prevention and Control, National Health Commission, Chinese Centre for Disease Control and Prevention, Beijing, 102206 China
- College of Life Science, Hengshui University, Hengshui, 053000 China
| | - Dandi Li
- NHC Key Laboratory of Medical Viruses and Viral Diseases, Institute of Viral Disease Prevention and Control, National Health Commission, Chinese Centre for Disease Control and Prevention, Beijing, 102206 China
| | - Jindong Wang
- Department of Pathogenic Biology, Weifang Medical University, Weifang, 261053 China
| | - Guangping Xiong
- College of Public Health, Gansu University of Chinese Medicine, Lanzhou, 730000 China
| | - Mengxuan Wang
- NHC Key Laboratory of Medical Viruses and Viral Diseases, Institute of Viral Disease Prevention and Control, National Health Commission, Chinese Centre for Disease Control and Prevention, Beijing, 102206 China
| | - Dan Liu
- NHC Key Laboratory of Medical Viruses and Viral Diseases, Institute of Viral Disease Prevention and Control, National Health Commission, Chinese Centre for Disease Control and Prevention, Beijing, 102206 China
| | - Yuhang Wei
- College of Public Health, Gansu University of Chinese Medicine, Lanzhou, 730000 China
| | - Lili Pang
- NHC Key Laboratory of Medical Viruses and Viral Diseases, Institute of Viral Disease Prevention and Control, National Health Commission, Chinese Centre for Disease Control and Prevention, Beijing, 102206 China
| | - Xiaoman Sun
- NHC Key Laboratory of Medical Viruses and Viral Diseases, Institute of Viral Disease Prevention and Control, National Health Commission, Chinese Centre for Disease Control and Prevention, Beijing, 102206 China
| | - Huiying Li
- NHC Key Laboratory of Medical Viruses and Viral Diseases, Institute of Viral Disease Prevention and Control, National Health Commission, Chinese Centre for Disease Control and Prevention, Beijing, 102206 China
| | - Xiangyu Kong
- NHC Key Laboratory of Medical Viruses and Viral Diseases, Institute of Viral Disease Prevention and Control, National Health Commission, Chinese Centre for Disease Control and Prevention, Beijing, 102206 China
| | - Saleha Shahar
- Department of Biosciences, Faculty of Sciences, Universiti Teknologi Malaysia, Johor Bahru, 81310 Malaysia
| | - Zhaojun Duan
- NHC Key Laboratory of Medical Viruses and Viral Diseases, Institute of Viral Disease Prevention and Control, National Health Commission, Chinese Centre for Disease Control and Prevention, Beijing, 102206 China
| |
Collapse
|
20
|
Manjate F, João ED, Mwangi P, Chirinda P, Mogotsi M, Messa A, Garrine M, Vubil D, Nobela N, Nhampossa T, Acácio S, Tate JE, Parashar U, Weldegebriel G, Mwenda JM, Alonso PL, Cunha C, Nyaga M, Mandomando I. Genomic characterization of the rotavirus G3P[8] strain in vaccinated children, reveals possible reassortment events between human and animal strains in Manhiça District, Mozambique. Front Microbiol 2023; 14:1193094. [PMID: 37342557 PMCID: PMC10277737 DOI: 10.3389/fmicb.2023.1193094] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/24/2023] [Accepted: 04/20/2023] [Indexed: 06/23/2023] Open
Abstract
Mozambique introduced the rotavirus vaccine (Rotarix®; GlaxoSmithKline Biologicals, Rixensart, Belgium) in 2015, and since then, the Centro de Investigação em Saúde de Manhiça has been monitoring its impact on rotavirus-associated diarrhea and the trend of circulating strains, where G3P[8] was reported as the predominant strain after the vaccine introduction. Genotype G3 is among the most commonly detected Rotavirus strains in humans and animals, and herein, we report on the whole genome constellation of G3P[8] detected in two children (aged 18 months old) hospitalized with moderate-to-severe diarrhea at the Manhiça District Hospital. The two strains had a typical Wa-like genome constellation (I1-R1-C1-M1-A1-N1-T1-E1-H1) and shared 100% nucleotide (nt) and amino acid (aa) identities in 10 gene segments, except for VP6. Phylogenetic analysis demonstrated that genome segments encoding VP7, VP6, VP1, NSP3, and NSP4 of the two strains clustered most closely with porcine, bovine, and equine strains with identities ranging from 86.9-99.9% nt and 97.2-100% aa. Moreover, they consistently formed distinct clusters with some G1P[8], G3P[8], G9P[8], G12P[6], and G12P[8] strains circulating from 2012 to 2019 in Africa (Mozambique, Kenya, Rwanda, and Malawi) and Asia (Japan, China, and India) in genome segments encoding six proteins (VP2, VP3, NSP1-NSP2, NSP5/6). The identification of segments exhibiting the closest relationships with animal strains shows significant diversity of rotavirus and suggests the possible occurrence of reassortment events between human and animal strains. This demonstrates the importance of applying next-generation sequencing to monitor and understand the evolutionary changes of strains and evaluate the impact of vaccines on strain diversity.
Collapse
Affiliation(s)
- Filomena Manjate
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
- Global Health and Tropical Medicine (GHTM), Instituto de Higiene e Medicina Tropical (IHMT), Universidade Nova de Lisboa (UNL), Lisbon, Portugal
| | - Eva D. João
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
| | - Peter Mwangi
- Next Generation Sequencing Unit and Division of Virology, Faculty of Health Sciences, University of the Free State, Bloemfontein, South Africa
| | - Percina Chirinda
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
| | - Milton Mogotsi
- Next Generation Sequencing Unit and Division of Virology, Faculty of Health Sciences, University of the Free State, Bloemfontein, South Africa
| | - Augusto Messa
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
| | - Marcelino Garrine
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
- Global Health and Tropical Medicine (GHTM), Instituto de Higiene e Medicina Tropical (IHMT), Universidade Nova de Lisboa (UNL), Lisbon, Portugal
| | - Delfino Vubil
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
| | - Nélio Nobela
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
| | - Tacilta Nhampossa
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
- Instituto Nacional de Saúde, Ministério da Saúde, Marracuene, Mozambique
| | - Sozinho Acácio
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
- Instituto Nacional de Saúde, Ministério da Saúde, Marracuene, Mozambique
| | - Jacqueline E. Tate
- Centers for Disease Control and Prevention (CDC), Atlanta, GA, United States
| | - Umesh Parashar
- Centers for Disease Control and Prevention (CDC), Atlanta, GA, United States
| | - Goitom Weldegebriel
- African Rotavirus Surveillance Network, Immunization, Vaccines, and Development Program, Regional Office for Africa, World Health Organization, Brazzaville, Democratic Republic of Congo
| | - Jason M. Mwenda
- African Rotavirus Surveillance Network, Immunization, Vaccines, and Development Program, Regional Office for Africa, World Health Organization, Brazzaville, Democratic Republic of Congo
| | - Pedro L. Alonso
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
| | - Celso Cunha
- Global Health and Tropical Medicine (GHTM), Instituto de Higiene e Medicina Tropical (IHMT), Universidade Nova de Lisboa (UNL), Lisbon, Portugal
| | - Martin Nyaga
- Next Generation Sequencing Unit and Division of Virology, Faculty of Health Sciences, University of the Free State, Bloemfontein, South Africa
| | - Inácio Mandomando
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
- Instituto Nacional de Saúde, Ministério da Saúde, Marracuene, Mozambique
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
21
|
Mwangi PN, Potgieter RL, Uwimana J, Mutesa L, Muganga N, Murenzi D, Tusiyenge L, Mwenda JM, Mogotsi MT, Rakau K, Esona MD, Steele AD, Seheri ML, Nyaga MM. The Evolution of Post-Vaccine G8P[4] Group a Rotavirus Strains in Rwanda; Notable Variance at the Neutralization Epitope Sites. Pathogens 2023; 12:658. [PMID: 37242329 PMCID: PMC10223037 DOI: 10.3390/pathogens12050658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/05/2023] [Revised: 04/24/2023] [Accepted: 04/26/2023] [Indexed: 05/28/2023] Open
Abstract
Africa has a high level of genetic diversity of rotavirus strains, which is suggested to be a possible reason contributing to the suboptimal effectiveness of rotavirus vaccines in this region. One strain that contributes to this rotavirus diversity in Africa is the G8P[4]. This study aimed to elucidate the entire genome and evolution of Rwandan G8P[4] strains. Illumina sequencing was performed for twenty-one Rwandan G8P[4] rotavirus strains. Twenty of the Rwandan G8P[4] strains had a pure DS-1-like genotype constellation, and one strain had a reassortant genotype constellation. Notable radical amino acid differences were observed at the neutralization sites when compared with cognate regions in vaccine strains potentially playing a role in neutralization escape. Phylogenetic analysis revealed that the closest relationship was with East African human group A rotavirus (RVA) strains for five of the genome segments. Two genome sequences of the NSP4 genome segment were closely related to bovine members of the DS-1-like family. Fourteen VP1 and eleven VP3 sequences had the closest relationships with the RotaTeq™ vaccine WC3 bovine genes. These findings suggest that the evolution of VP1 and VP3 might have resulted from reassortment events with RotaTeq™ vaccine WC3 bovine genes. The close phylogenetic relationship with East African G8P[4] strains from Kenya and Uganda suggests co-circulation in these countries. These findings highlight the need for continued whole-genomic surveillance to elucidate the evolution of G8P[4] strains, especially after the introduction of rotavirus vaccination.
Collapse
Affiliation(s)
- Peter N. Mwangi
- Next Generation Sequencing Unit, Division of Virology, Faculty of Health Sciences, University of the Free State, Bloemfontein 9300, South Africa
| | - Robyn-Lee Potgieter
- Next Generation Sequencing Unit, Division of Virology, Faculty of Health Sciences, University of the Free State, Bloemfontein 9300, South Africa
| | - Jeannine Uwimana
- Kigali University Teaching Hospital, College of Medicine and Health Sciences, University of Rwanda, Kigali P.O. Box 4285, Rwanda
| | - Leon Mutesa
- Kigali University Teaching Hospital, College of Medicine and Health Sciences, University of Rwanda, Kigali P.O. Box 4285, Rwanda
- Centre for Human Genetics, College of Medicine and Health Sciences, University of Rwanda, Kigali P.O. Box 4285, Rwanda
| | - Narcisse Muganga
- Kigali University Teaching Hospital, College of Medicine and Health Sciences, University of Rwanda, Kigali P.O. Box 4285, Rwanda
| | - Didier Murenzi
- Kigali University Teaching Hospital, College of Medicine and Health Sciences, University of Rwanda, Kigali P.O. Box 4285, Rwanda
| | - Lisine Tusiyenge
- Kigali University Teaching Hospital, College of Medicine and Health Sciences, University of Rwanda, Kigali P.O. Box 4285, Rwanda
| | - Jason M. Mwenda
- World Health Organization, Regional Office for Africa, Brazzaville P.O. Box 06, Congo
| | - Milton T. Mogotsi
- Next Generation Sequencing Unit, Division of Virology, Faculty of Health Sciences, University of the Free State, Bloemfontein 9300, South Africa
| | - Kebareng Rakau
- Diarrhoeal Pathogens Research Unit, Sefako Makgatho Health Sciences University (MEDUNSA), Pretoria 0204, South Africa
| | - Mathew D. Esona
- Diarrhoeal Pathogens Research Unit, Sefako Makgatho Health Sciences University (MEDUNSA), Pretoria 0204, South Africa
| | - A. Duncan Steele
- Diarrhoeal Pathogens Research Unit, Sefako Makgatho Health Sciences University (MEDUNSA), Pretoria 0204, South Africa
| | - Mapaseka L. Seheri
- Diarrhoeal Pathogens Research Unit, Sefako Makgatho Health Sciences University (MEDUNSA), Pretoria 0204, South Africa
| | - Martin M. Nyaga
- Next Generation Sequencing Unit, Division of Virology, Faculty of Health Sciences, University of the Free State, Bloemfontein 9300, South Africa
| |
Collapse
|
22
|
Makori TO, Bargul JL, Lambisia AW, Mwanga MJ, Murunga N, de Laurent ZR, Lewa CS, Mutunga M, Kellam P, Cotten M, Nokes DJ, Phan M, Agoti CN. Genomic epidemiology of the rotavirus G2P[4] strains in coastal Kenya pre- and post-rotavirus vaccine introduction, 2012-8. Virus Evol 2023; 9:vead025. [PMID: 37207000 PMCID: PMC10190042 DOI: 10.1093/ve/vead025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/24/2022] [Revised: 03/07/2023] [Accepted: 04/14/2023] [Indexed: 05/21/2023] Open
Abstract
The introduction of rotavirus vaccines into the national immunization programme in many countries has led to a decline in childhood diarrhoea disease burden. Coincidentally, the incidence of some rotavirus group A (RVA) genotypes has increased, which may result from non-vaccine-type replacement. Here, we investigate the evolutionary genomics of rotavirus G2P[4] which has shown an increase in countries that introduced the monovalent Rotarix® vaccine. We examined sixty-three RVA G2P[4] strains sampled from children (aged below 13 years) admitted to Kilifi County Hospital, coastal Kenya, pre- (2012 to June 2014) and post-(July 2014 to 2018) rotavirus vaccine introduction. All the sixty-three genome sequences showed a typical DS-1-like genome constellation (G2-P[4]-I2-R2-C2-M2-A2-N2-T2-E2-H2). Pre-vaccine G2 sequences predominantly classified as sub-lineage IVa-3 and co-circulated with low numbers of sub-lineage IVa-1 strains, whereas post-vaccine G2 sequences mainly classified into sub-lineage IVa-3. In addition, in the pre-vaccine period, P[4] sub-lineage IVa strains co-circulated with low numbers of P[4] lineage II strains, but P[4] sub-lineage IVa strains predominated in the post-vaccine period. On the global phylogeny, the Kenyan pre- and post-vaccine G2P[4] strains clustered separately, suggesting that different virus populations circulated in the two periods. However, the strains from both periods exhibited conserved amino acid changes in the known antigenic epitopes, suggesting that replacement of the predominant G2P[4] cluster was unlikely a result of immune escape. Our findings demonstrate that the pre- and post-vaccine G2P[4] strains circulating in Kilifi, coastal Kenya, differed genetically but likely were antigenically similar. This information informs the discussion on the consequences of rotavirus vaccination on rotavirus diversity.
Collapse
Affiliation(s)
- Timothy O Makori
- Epidemiology and Demography Department Kenya Medical Research Institute (KEMRI)-Wellcome Trust Research Programme, Off Hospital Road, P.O BOX 230-80108, Kilifi, Kenya
- Department of Biochemistry, Jomo Kenyatta University of Agriculture and Technology, Kalimoni, PO Box 62000-00200, Juja, Kenya
| | - Joel L Bargul
- Department of Biochemistry, Jomo Kenyatta University of Agriculture and Technology, Kalimoni, PO Box 62000-00200, Juja, Kenya
- International Centre of Insect Physiology and Ecology, Animal Health Theme, ICIPE Road Kasarani, P.O BOX 30772-00100, Nairobi, Kenya
| | - Arnold W Lambisia
- Epidemiology and Demography Department Kenya Medical Research Institute (KEMRI)-Wellcome Trust Research Programme, Off Hospital Road, P.O BOX 230-80108, Kilifi, Kenya
| | - Mike J Mwanga
- Epidemiology and Demography Department Kenya Medical Research Institute (KEMRI)-Wellcome Trust Research Programme, Off Hospital Road, P.O BOX 230-80108, Kilifi, Kenya
| | - Nickson Murunga
- Epidemiology and Demography Department Kenya Medical Research Institute (KEMRI)-Wellcome Trust Research Programme, Off Hospital Road, P.O BOX 230-80108, Kilifi, Kenya
| | - Zaydah R de Laurent
- Epidemiology and Demography Department Kenya Medical Research Institute (KEMRI)-Wellcome Trust Research Programme, Off Hospital Road, P.O BOX 230-80108, Kilifi, Kenya
| | - Clement S Lewa
- Epidemiology and Demography Department Kenya Medical Research Institute (KEMRI)-Wellcome Trust Research Programme, Off Hospital Road, P.O BOX 230-80108, Kilifi, Kenya
| | - Martin Mutunga
- Epidemiology and Demography Department Kenya Medical Research Institute (KEMRI)-Wellcome Trust Research Programme, Off Hospital Road, P.O BOX 230-80108, Kilifi, Kenya
| | - Paul Kellam
- Department of Infectious Diseases, Faculty of Medicine, Imperial College London, Exhibition Road, London SW7 2AZ, UK
- Kymab Ltd, The Bennet Building (B930), Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Matthew Cotten
- Medical Research Centre (MRC)/Uganda Virus Research Institute, Plot No: 51-59 Nakiwogo Road, P.O.Box 49, Entebbe, Uganda
- MRC-University of Glasgow, Centre for Virus Research Glasgow, 464 Bearsden Road, Glasgow G61 1QH UK
| | - D James Nokes
- Epidemiology and Demography Department Kenya Medical Research Institute (KEMRI)-Wellcome Trust Research Programme, Off Hospital Road, P.O BOX 230-80108, Kilifi, Kenya
- School of Life Sciences and Zeeman Institute (SBIDER), The University of Warwick, Gibbet Hill Campus, Coventry CV4 7AL, UK
| | - My Phan
- Medical Research Centre (MRC)/Uganda Virus Research Institute, Plot No: 51-59 Nakiwogo Road, P.O.Box 49, Entebbe, Uganda
- MRC-University of Glasgow, Centre for Virus Research Glasgow, 464 Bearsden Road, Glasgow G61 1QH UK
| | - Charles N Agoti
- Epidemiology and Demography Department Kenya Medical Research Institute (KEMRI)-Wellcome Trust Research Programme, Off Hospital Road, P.O BOX 230-80108, Kilifi, Kenya
- School of Health and Human Sciences, Pwani University, Kilifi-Malindi Road, P.O BOX 195-80108, Kilifi, Kenya
| |
Collapse
|
23
|
Characterization of Sialic Acid-Independent Simian Rotavirus Mutants in Viral Infection and Pathogenesis. J Virol 2023; 97:e0139722. [PMID: 36602365 PMCID: PMC9888295 DOI: 10.1128/jvi.01397-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 01/06/2023] Open
Abstract
Rotaviruses (RVs) are nonenveloped viruses that cause gastroenteritis in infants and young children. Sialic acid is an initial receptor, especially for animal RVs, including rhesus RV. Sialic acid binds to the VP8* subunit, a part of the outer capsid protein VP4 of RV. Although interactions between virus and glycan receptors influence tissue and host tropism and viral pathogenicity, research has long been limited to biochemical and structural studies due to the unavailability of an RV reverse genetics system. Here, we examined the importance of sialic acid in RV infections using recombinant RVs harboring mutations in sialic acid-binding sites in VP4 via a simian RV strain SA11-based reverse genetics system. RV VP4 mutants that could not bind to sialic acid had replicated to decreased viral titer in MA104 cells. Wild-type virus infectivity was reduced, while that of VP4 mutants was not affected in sialic acid-deficient cells. Unexpectedly, in vivo experiments demonstrated that VP4 mutants suppressed mouse pups' weight gain and exacerbated diarrhea symptoms compared to wild-type viruses. Intestinal contents enhanced VP4 mutants' infectivity. Thus, possibly via interactions with other unknown receptors and/or intestinal contents, VP4 mutants are more likely than wild-type viruses to proliferate in the murine intestine, causing diarrhea and weight loss. These results suggest that RVs binding sialic acid notably affect viral infection in vitro and viral pathogenesis in vivo. IMPORTANCE Various studies have been conducted on the binding of VP8* and glycans, and the direct interaction between purified VP8* and glycans has been investigated by crystalline structure analyses. Here, we used a reverse genetics system to generate rotaviruses (RVs) with various VP4 mutants. The generated mutant strains clarified the importance of glycan binding in vitro and in vivo. Moreover, even when VP4 mutants could not bind to sialic acid, they were able to bind to an unknown receptor. As RVs evolve, pathogenicity can also be modified by easily altering the glycans to which VP4 binds.
Collapse
|
24
|
Ates O, Yesilbag K. Characterization of bovine rotavirus isolates from diarrheic calves in Türkiye. Mol Biol Rep 2023; 50:3063-3071. [PMID: 36689052 PMCID: PMC9870195 DOI: 10.1007/s11033-022-08169-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/22/2022] [Accepted: 12/01/2022] [Indexed: 01/24/2023]
Abstract
BACKGROUND Neonatal calf diarrhea, which is the most common cause in calf deaths, leads to significant economic losses in dairy farming around the world. Diarrhea develops due to infectious and non-infectious reasons. Group A Rotaviruses (RVA) are the leading and predisposing factor for acute neonatal gastroenteritis. METHODS AND RESULTS In this study, 20 diarrheic fecal samples were collected from one farm in Balıkesir province of Turkey. During virus isolation, a total of 2 stool samples were detected to produce cytopathogenic effects in MA-104 cell line. The two samples (RV-36, RV-38) were tested positive with antigen ELISA kits detecting RVA antigens. In order to detect the presence of rotavirus viral nucleic acid in cell supernatants, VP6 gene region-specific RT-PCR test was performed and the samples RV-36 and RV-38 were positive for RVA viral nucleic acid. By RT-PCR using genotype specific primers, both the isolates RV-36 and RV-38 formed amplicons compatible with G10 and P[11] genotypes of RVA. RVA nucleic acids segments were also visualized by poliacrilamide gel electrophoresis (PAGE) method. The phylogenetic tree constructed according to the VP6 gene region showed that these isolates were in the Rotavirus A group and in the I2 cluster same as other bovine and some human RVA isolates. CONCLUSION Succesful isolation of RVA G10P[11] was echieved in the cattle farm. As rotaviruses play the most important role in the etiology of diarrhea in newborn calves respected genotype G10P[11] should be considered in selection of the vaccines applied to the dams. Those isolates can be further evaluated as vaccine candidate.
Collapse
Affiliation(s)
- Ozer Ates
- Department of Virology, Faculty of Veterinary Medicine, Bursa Uludag University, 16059 Bursa, Turkey ,Department of Laboratory Animals Science, Faculty of Veterinary Medicine, Afyon Kocatepe University, 03204 Afyonkarahisar, Turkey
| | - Kadir Yesilbag
- Department of Virology, Faculty of Veterinary Medicine, Bursa Uludag University, 16059 Bursa, Turkey
| |
Collapse
|
25
|
Raev S, Amimo J, Saif L, Vlasova A. Intestinal mucin-type O-glycans: the major players in the host-bacteria-rotavirus interactions. Gut Microbes 2023; 15:2197833. [PMID: 37020288 PMCID: PMC10078158 DOI: 10.1080/19490976.2023.2197833] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 12/07/2022] [Accepted: 03/28/2023] [Indexed: 04/07/2023] Open
Abstract
Rotavirus (RV) causes severe diarrhea in young children and animals worldwide. Several glycans terminating in sialic acids (SAs) and histo-blood group antigens (HBGAs) on intestinal epithelial cell (IEC) surface have been recognized to act as attachment sites for RV. IECs are protected by the double layer of mucus of which O-glycans (including HBGAs and SAs) are a major organic component. Luminal mucins, as well as bacterial glycans, can act as decoy molecules removing RV particles from the gut. The composition of the intestinal mucus is regulated by complex O-glycan-specific interactions among the gut microbiota, RV and the host. In this review, we highlight O-glycan-mediated interactions within the intestinal lumen prior to RV attachment to IECs. A better understanding of the role of mucus is essential for the development of alternative therapeutic tools including the use of pre- and probiotics to control RV infection.
Collapse
Affiliation(s)
- S.A. Raev
- Center for Food Animal Health, Department of Animal Sciences, College of Food, Agricultural and Environmental Sciences, The Ohio State University, Wooster, OH, USA
| | - J.O. Amimo
- Center for Food Animal Health, Department of Animal Sciences, College of Food, Agricultural and Environmental Sciences, The Ohio State University, Wooster, OH, USA
- Department of Animal Production, Faculty of Veterinary Medicine, University of Nairobi, Nairobi, Kenya
| | - L.J. Saif
- Center for Food Animal Health, Department of Animal Sciences, College of Food, Agricultural and Environmental Sciences, The Ohio State University, Wooster, OH, USA
| | - A.N. Vlasova
- Center for Food Animal Health, Department of Animal Sciences, College of Food, Agricultural and Environmental Sciences, The Ohio State University, Wooster, OH, USA
| |
Collapse
|
26
|
Maass T, Ssebyatika G, Brückner M, Breckwoldt L, Krey T, Mallagaray A, Peters T, Frank M, Creutznacher R. Binding of Glycans to the SARS CoV-2 Spike Protein, an Open Question: NMR Data on Binding Site Localization, Affinity, and Selectivity. Chemistry 2022; 28:e202202614. [PMID: 36161798 PMCID: PMC9537997 DOI: 10.1002/chem.202202614] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/22/2022] [Revised: 09/26/2022] [Accepted: 09/26/2022] [Indexed: 11/25/2022]
Abstract
We have used NMR experiments to explore the binding of selected glycans and glycomimetics to the SARS CoV-2 spike glycoprotein (S-protein) and to its receptor binding domain (RBD). STD NMR experiments confirm the binding of sialoglycans to the S-protein of the prototypic Wuhan strain virus and yield dissociation constants in the millimolar range. The absence of STD effects for sialoglycans in the presence of the Omicron/BA.1 S-protein reflects a loss of binding as a result of S-protein evolution. Likewise, no STD effects are observed for the deletion mutant Δ143-145 of the Wuhan S-protein, thus supporting localization of the binding site in the N-terminal domain (NTD). The glycomimetics Oseltamivir and Zanamivir bind weakly to the S-protein of both virus strains. Binding of blood group antigens to the Wuhan S-protein cannot be confirmed by STD NMR. Using 1 H,15 N TROSY HSQC-based chemical shift perturbation (CSP) experiments, we excluded binding of any of the ligands studied to the RBD of the Wuhan S-protein. Our results put reported data on glycan binding into perspective and shed new light on the potential role of glycan-binding to the S-protein.
Collapse
Affiliation(s)
- Thorben Maass
- University of Lübeck: Universitat zu LubeckInstitute of Chemistry and MetabolomicsGERMANY
| | - George Ssebyatika
- University of Lübeck: Universitat zu LubeckInstitute of BiochemistryGERMANY
| | - Marlene Brückner
- University of Lübeck: Universitat zu LubeckInstitute of Chemistry and MetabolomicsGERMANY
| | - Lea Breckwoldt
- University of Lübeck: Universitat zu LubeckInstitute of Chemistry and MetabolomicsGERMANY
| | - Thomas Krey
- University of Lübeck: Universitat zu LubeckInstitute of BiochemistryGERMANY
| | - Alvaro Mallagaray
- University of Lübeck: Universitat zu LubeckInstitute of Chemistry and MetabolomicsGERMANY
| | - Thomas Peters
- Institute for Chemistry and MetabolomicsUniversität zu LübeckRatzeburger Allee 16023562LübeckGERMANY
| | | | - Robert Creutznacher
- University of Lübeck: Universitat zu LubeckInstitute of Chemistry and MetabolomicsGERMANY
| |
Collapse
|
27
|
Gutierrez MB, de Assis RMS, Arantes I, Fumian TM. Full genotype constellations analysis of unusual DS-1-like G12P[6] and G6P[8] rotavirus strains detected in Brazil, 2019. Virology 2022; 577:74-83. [PMID: 36323046 DOI: 10.1016/j.virol.2022.10.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/21/2022] [Revised: 10/20/2022] [Accepted: 10/21/2022] [Indexed: 11/07/2022]
Abstract
Rotavirus A (RVA) is a major cause of acute gastroenteritis (AGE) in children worldwide. We report unusual RVA G12P[6] and G6P[8] strains isolated from fecal samples from Brazilian children hospitalized for AGE. The characterized RVA have genome segments backbone: G12-P[6]/ G6-P[8]-I2-R2-C2-M2-A2-N2-T2-E2-H2 of DS-1-like genogroup. Our study describes the first identification of G6P[8], a DS-1-like genogroup strain. Nucleotide analysis of VP7 and VP4 genes revealed that all G12 Brazilian strains clustered into the sub-lineages IIIB, mostly associated with P[6] lineage I. Additionally, our G6 lineage I strains were closely related to German G6 genotypes, bound with P[8] lineage III, differing from both vaccine strains. The comparative sequence analysis of our strains with vaccine strains revealed amino acid substitutions located in immunodominant regions of VP7 and VP4 proteins. Continuous monitoring of RVA genotypes is essential to evaluate the impact of vaccination on the dynamic nature of RVA evolution.
Collapse
Affiliation(s)
- Meylin Bautista Gutierrez
- Laboratory of Comparative and Environmental Virology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation (Fiocruz), Av. Brasil, 4365, Rio de Janeiro, RJ 21040-360, Brazil
| | - Rosane Maria Santos de Assis
- Laboratory of Comparative and Environmental Virology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation (Fiocruz), Av. Brasil, 4365, Rio de Janeiro, RJ 21040-360, Brazil
| | - Ighor Arantes
- Laboratory of Respiratory Viruses and Measles, Oswaldo Cruz Institute, Oswaldo Cruz Foundation (Fiocruz), Av. Brasil, 4365, Rio de Janeiro, RJ 21040-360, Brazil
| | - Tulio Machado Fumian
- Laboratory of Comparative and Environmental Virology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation (Fiocruz), Av. Brasil, 4365, Rio de Janeiro, RJ 21040-360, Brazil.
| |
Collapse
|
28
|
Elbashir I, Aldoos NF, Mathew S, Al Thani AA, Emara MM, Yassine HM. Molecular epidemiology, genetic diversity, and vaccine availability of viral acute gastroenteritis in the middle East and North Africa (MENA) region. J Infect Public Health 2022; 15:1193-1211. [PMID: 36240530 DOI: 10.1016/j.jiph.2022.09.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/15/2022] [Revised: 08/26/2022] [Accepted: 09/11/2022] [Indexed: 11/18/2022] Open
Abstract
Acute gastroenteritis is the cause of considerable mortality and morbidity worldwide, particularly among children under five years in underdeveloped countries. Most acute gastroenteritis (AGE) cases are attributed to viral etiologies, including rotavirus, norovirus, adenovirus, astrovirus, and sapovirus. This paper aimed to determine the prevalence rate of different viral etiologies of AGE in the Middle East and North Africa (MENA) region. Moreover, this paper explored rotavirus phylogenetic relatedness, compared VP7 and VP4 antigenic regions of rotavirus with vaccine strains, and explored the availability of vaccines in the MENA region. The literature search identified 160 studies from 18 countries from 1980 to 2019. The overall prevalence of rotavirus, norovirus, adenovirus, astrovirus, and sapovirus were 29.8 %, 13.9 %, 6.3 %, 3.5 %, and 3.2 % of tested samples, respectively. The most common rotavirus genotype combinations in the MENA region were G1P[8], G9P[9], and G2P[4], whereas GII.4 was the predominant norovirus genotype all of which were reported in almost all the studies with genotyping data. The comparison of VP7 and VP4 between circulating rotavirus in the MENA region and vaccine strains has revealed discrete divergent regions, including the neutralizing epitopes. Rotavirus vaccine was introduced to most of the countries of the MENA region; however, only a few studies have assessed the effectiveness of vaccine introduction. This paper provides a comprehensive update on the prevalence of the different viral agents of AGE in the MENA region.
Collapse
Affiliation(s)
- Israa Elbashir
- Biomedical Research Center, Qatar University, 2713 Doha, Qatar.
| | - Noor F Aldoos
- Department of Laboratory Medicine and Pathology, Hamad Medical Corporation, Doha, Qatar.
| | - Shilu Mathew
- Biomedical Research Center, Qatar University, 2713 Doha, Qatar.
| | - Asmaa A Al Thani
- Biomedical Research Center, Qatar University, 2713 Doha, Qatar; Department of Biomedical Sciences, College of Health Science-QU Health, Qatar University, Doha 2713, Qatar
| | - Mohamed M Emara
- Basic Medical Sciences Department, College of Medicine, QU Health, Qatar University, 2713 Doha, Qatar.
| | - Hadi M Yassine
- Biomedical Research Center, Qatar University, 2713 Doha, Qatar.
| |
Collapse
|
29
|
Miao Q, Pan Y, Gong L, Guo L, Wu L, Jing Z, Zhang G, Tian J, Feng L. Full genome characterization of a human-porcine reassortment G12P[7] rotavirus and its pathogenicity in piglets. Transbound Emerg Dis 2022; 69:3506-3517. [PMID: 36150417 DOI: 10.1111/tbed.14712] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/29/2022] [Revised: 09/13/2022] [Accepted: 09/17/2022] [Indexed: 02/04/2023]
Abstract
In recent years, increasing numbers of cases of acute gastroenteritis caused by Group A rotavirus (RVA) G12 strains have been reported in humans from many countries around the world, but G12 RVA detection in animals is currently less reported. Pigs are an important animal reservoir of zoonotic RVs and a mixing vessel for RVs. In 2020, RVA infection cases in piglets increased in China, which attracted more attention. During an epidemiological survey, a new type of porcine G12P[7] strain (CN127) was detected in pig farms across several provinces. Complete genome analyses revealed that strain CN127 possessed a Wa-like backbone with a genotype constellation of G12-P[7]-I1-C1-M1-R1-A8-N1-T1-E1-H1. The A8 genotype is indicative of its porcine rotavirus origin. Sequence identities and phylogenetic analyses showed that the VP2, VP4, NSP1, NSP4 and NSP5 genes were most closely related to those of porcine rotaviruses, but the VP1, VP6, VP7 and NSP2-3 genes were most closely related to those of human rotaviruses. CN127 likely emerged due to genetic reassortment between porcine and human rotavirus. In vivo experiments showed that CN127 infection caused gastrointestinal tract lesions in piglets and histopathological changes in the lung, liver and mesenteric lymph nodes (MLNs). In the small intestine, RVA antigen was detected in the duodenum and jejunum but not in the ileum. In the extra-intestinal tissues, RVA antigen was detected in the lung but not in the MLNs. Viral RNA was detected in the intestinal and extra-intestinal tissues as well as blood. This study reveals that RVA G12P[7] may become an epidemic strain in China and also provides further evidence that cocirculating human and porcine strains could produce new genotype rotaviruses with high virulence in piglets.
Collapse
Affiliation(s)
- Qian Miao
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People's Republic of China
| | - Yudi Pan
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People's Republic of China
| | - Lang Gong
- Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, People's Republic of China
| | - Longjun Guo
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People's Republic of China
| | - Ling Wu
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People's Republic of China
| | - Zhaoyang Jing
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People's Republic of China
| | - Guihong Zhang
- Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, People's Republic of China
| | - Jin Tian
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People's Republic of China
| | - Li Feng
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People's Republic of China
| |
Collapse
|
30
|
Patić A, Vuković V, Kovačević G, Petrović V, Ristić M, Djilas M, Knežević P, Pustahija T, Štrbac M, Djekić Malbaša J, Rajčević S, Hrnjaković Cvjetković I. Detection and Molecular Characterization of Rotavirus Infections in Children and Adults with Gastroenteritis from Vojvodina, Serbia. Microorganisms 2022; 10:2050. [PMID: 36296326 PMCID: PMC9607116 DOI: 10.3390/microorganisms10102050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/30/2022] [Revised: 10/08/2022] [Accepted: 10/11/2022] [Indexed: 11/19/2022] Open
Abstract
Rotaviruses (RV) are the leading cause of gastroenteritis in infants, young children, and adults, responsible for serious disease burden. In the period 2012-2018, a cross-sectional study was conducted using stool samples collected from patients with acute gastroenteritis from Vojvodina, Serbia. We described age and gender distribution, as well as seasonal patterns of RV prevalence. Out of 1853 included stool samples, RV was detected in 29%. Hospitalized children between 1-2 years old were especially affected by RV infection (45%). The highest prevalence of infection was observed during the colder, winter/spring months. We compared sequenced representative G and P genotypes circulating in Serbia with vaccine strains and determined their genetic similarity. Genotype combination G2P[4] was the most prevalent (34.6%), followed by G2P[8] (24.1%) and G1P[8] (21.1%). Given that several epitopes were conserved, neutralization motifs among circulating strains can be characterized as sufficiently matching vaccine strains Rotarix™ and RotaTeq™, but existing antigenic disparities should not be overlooked. The present results contribute to a better insight into the prevalence of rotavirus infection in our region and point out the need for epidemiological surveillance of rotaviruses before the introduction of vaccines. These data can help formulate future vaccine strategies in Serbia.
Collapse
Affiliation(s)
- Aleksandra Patić
- Department of Microbiology with Parasitology and Immunology, Faculty of Medicine, University of Novi Sad, 21000 Novi Sad, Serbia
- Institute of Public Health of Vojvodina, 21000 Novi Sad, Serbia
| | - Vladimir Vuković
- Institute of Public Health of Vojvodina, 21000 Novi Sad, Serbia
- Department of Epidemiology, Faculty of Medicine, University of Novi Sad, 21000 Novi Sad, Serbia
| | | | - Vladimir Petrović
- Institute of Public Health of Vojvodina, 21000 Novi Sad, Serbia
- Department of Epidemiology, Faculty of Medicine, University of Novi Sad, 21000 Novi Sad, Serbia
| | - Mioljub Ristić
- Institute of Public Health of Vojvodina, 21000 Novi Sad, Serbia
- Department of Epidemiology, Faculty of Medicine, University of Novi Sad, 21000 Novi Sad, Serbia
| | - Milan Djilas
- Institute of Public Health of Vojvodina, 21000 Novi Sad, Serbia
| | - Petar Knežević
- Faculty of Sciences, University of Novi Sad, 21000 Novi Sad, Serbia
| | - Tatjana Pustahija
- Institute of Public Health of Vojvodina, 21000 Novi Sad, Serbia
- Department of Epidemiology, Faculty of Medicine, University of Novi Sad, 21000 Novi Sad, Serbia
| | - Mirjana Štrbac
- Institute of Public Health of Vojvodina, 21000 Novi Sad, Serbia
| | - Jelena Djekić Malbaša
- Department of Epidemiology, Faculty of Medicine, University of Novi Sad, 21000 Novi Sad, Serbia
- Institute for Pulmonary Diseases of Vojvodina, 21204 Sremska Kamenica, Serbia
| | - Smiljana Rajčević
- Institute of Public Health of Vojvodina, 21000 Novi Sad, Serbia
- Department of Epidemiology, Faculty of Medicine, University of Novi Sad, 21000 Novi Sad, Serbia
| | - Ivana Hrnjaković Cvjetković
- Department of Microbiology with Parasitology and Immunology, Faculty of Medicine, University of Novi Sad, 21000 Novi Sad, Serbia
- Institute of Public Health of Vojvodina, 21000 Novi Sad, Serbia
| |
Collapse
|
31
|
Li X, Bi X, Shi X, Rao L, Fu ML, Sun W, Yuan B. Effect of particulate matters on inactivation of bacteriophage MS2 under irradiation above 320 nm. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:73976-73986. [PMID: 35633451 DOI: 10.1007/s11356-022-20811-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 11/09/2021] [Accepted: 05/10/2022] [Indexed: 06/15/2023]
Abstract
The inactivation of bacteriophage MS2 under irradiation above 320 nm was investigated, focusing on different solution pH, ionic strength, and Suwannee River natural organic matter (SRNOM) concentrations when solutions contained organic or inorganic particle matters. Inorganic and organic particles were modeled using kaolinite (KAO) and Microcystis aeruginosa (MA), respectively. The results showed that the two types of particles influenced on MS2 inactivation under different conditions. The lower pH contributed to the greater MS2 aggregation within pH range of 3.0 to 8.0, leading to an increasing inactivation rate. The presence of KAO induced reactive oxygen species (ROS) under the action of irradiation above 320 nm, which promoted the inactivation of MS2. By comparison, the [Formula: see text] produced by MA after irradiation promoted the inactivation at pH < 6, whereas when the pH is ≥ 6, the inactivation effect of MS2 was lower than that of particle-free solution because MS2 was no longer aggregated and MA has a shading effect. In the presence of Na+ or Ca2+ cation, irradiation above 320 nm could not effectively inactivate the MS2 under particle-free solution. By comparison, KAO increased the inactivation efficiency as a photosensitizer. With the increase of Ca2+ concentration, MS2 was more easily adsorbed to MA than aggregation. Until Ca2+ concentration reached 20 mM, the inactivation effect in MA solution was enhanced. In the presence of SRNOM, the inactivation effect increased with the increase of SRNOM concentration. When the SRNOM was 20 mM, the inactivation increased in the particle-free solution due to the greater production of [Formula: see text]. Compared with the particle-free solution, the KAO and MA inactivation efficiency was lower.
Collapse
Affiliation(s)
- Xiaoxue Li
- Xiamen Key Laboratory of Municipal and Industrial Solid Waste Utilization and Pollution Control, College of Civil Engineering, Huaqiao University, Xiamen, Fujian, 361021, People's Republic of China
| | - Xiaochao Bi
- Xiamen Key Laboratory of Municipal and Industrial Solid Waste Utilization and Pollution Control, College of Civil Engineering, Huaqiao University, Xiamen, Fujian, 361021, People's Republic of China
| | - Xiaoyang Shi
- Xiamen Key Laboratory of Municipal and Industrial Solid Waste Utilization and Pollution Control, College of Civil Engineering, Huaqiao University, Xiamen, Fujian, 361021, People's Republic of China
| | - La Rao
- Xiamen Key Laboratory of Municipal and Industrial Solid Waste Utilization and Pollution Control, College of Civil Engineering, Huaqiao University, Xiamen, Fujian, 361021, People's Republic of China
| | - Ming-Lai Fu
- Xiamen Key Laboratory of Municipal and Industrial Solid Waste Utilization and Pollution Control, College of Civil Engineering, Huaqiao University, Xiamen, Fujian, 361021, People's Republic of China
| | - Wenjie Sun
- Department of Atmospheric and Hydrologic Science, St. Cloud State University, 720 4th Avenue South, St. Cloud, MN, 56301, USA
| | - Baoling Yuan
- Xiamen Key Laboratory of Municipal and Industrial Solid Waste Utilization and Pollution Control, College of Civil Engineering, Huaqiao University, Xiamen, Fujian, 361021, People's Republic of China.
- Key Laboratory of Songliao Aquatic Environment, Ministry of Education, Jilin Jianzhu University, Changchun, 130118, People's Republic of China.
| |
Collapse
|
32
|
Abstract
Rotaviruses represent one of the most successful pathogens in the world, with high infectivity and efficient transmission between the young of many animal species, including humans. To overcome host defenses, rotaviruses have evolved a plethora of strategies to effectively evade the innate immune response, establish initial infection in the small intestine, produce progeny, and shed into the environment. Previously, studying the roles and relative contributions of specific rotaviral factors in innate immune evasion had been challenging without a plasmid-only reverse genetics system. Although still in its infancy, current reverse genetics technology will help address important research questions regarding rotavirus innate immune evasion, host range restriction, and viral pathogenesis. In this review, we summarize the current knowledge about the antiviral host innate immune defense mechanisms, countermeasures of rotavirus-encoded factors, and strategies to better understand these interactions using the rotavirus reverse genetics system.
Collapse
Affiliation(s)
- Avan Antia
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Amanda N. Pinski
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Siyuan Ding
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
33
|
Ren X, Saleem W, Haes R, Xie J, Theuns S, Nauwynck HJ. Milk lactose protects against porcine group A rotavirus infection. Front Microbiol 2022; 13:989242. [PMID: 36060735 PMCID: PMC9428151 DOI: 10.3389/fmicb.2022.989242] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/08/2022] [Accepted: 07/21/2022] [Indexed: 11/13/2022] Open
Abstract
Rotavirus A (RVA) is an important pathogen causing acute gastroenteritis in animals and humans. Attachment to the host receptor is a crucial step for virus replication. The VP8* domain is the distal terminal region of the RVA spike protein VP4 (expressed by the P gene) and is important for rotavirus binding and infectivity. Recent studies have indicated a role for non-sialylated glycans, including mucin core 2 and histo-blood group antigens (HBGAs), in the infectivity of human and animal group A rotaviruses. In the present study, we determined if porcine rotavirus-derived recombinant VP8* of the endemic strains 14R103 G5P[6], 13R054 G5P[7], 12R010 G4P[13], 12R046 G9P[23], and 12R022 G2P[27] interact with hitherto uncharacterized glycans. We successfully produced five recombinant GST-VP8* proteins of genotype P[6], P[7], P[13], P[23], and P[27]. The hemagglutination assay showed genotypes P[7] and P[23] hemagglutinate porcine and human red blood cells. In an array screen of > 300 glycans, recombinant VP8* of rotavirus genotype P[6], P[7], and P[13] showed specific binding to glycans with a Gal-β-1,4-Glc (β-lactose) motif, which forms the core structure of HBGAs. The specificity of glycan-binding was confirmed through an ELISA-based oligosaccharide binding assay. Further, 13R054 G5P[7] and 12R046 G9P[23] infectivity was significantly reduced by β-lactose in MA104 cells and primary porcine enterocytes. These data suggest that lactose, the main natural sugar in milk, plays an important role in protecting piglets from enteric viral replication and diarrhea.
Collapse
Affiliation(s)
- Xiaolei Ren
- Laboratory of Virology, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
- *Correspondence: Xiaolei Ren,
| | - Waqar Saleem
- Laboratory of Virology, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Robin Haes
- Laboratory of Virology, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Jiexiong Xie
- Laboratory of Virology, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Sebastiaan Theuns
- Laboratory of Virology, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
- PathoSense BV, Lier, Belgium
| | - Hans J. Nauwynck
- Laboratory of Virology, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| |
Collapse
|
34
|
Abstract
Rotavirus (RV) viroplasms are cytosolic inclusions where both virus genome replication and primary steps of virus progeny assembly take place. A stabilized microtubule cytoskeleton and lipid droplets are required for the viroplasm formation, which involves several virus proteins. The viral spike protein VP4 has not previously been shown to have a direct role in viroplasm formation. However, it is involved with virus-cell attachment, endocytic internalization, and virion morphogenesis. Moreover, VP4 interacts with actin cytoskeleton components, mainly in processes involving virus entrance and egress, and thereby may have an indirect role in viroplasm formation. In this study, we used reverse genetics to construct a recombinant RV, rRV/VP4-BAP, that contains a biotin acceptor peptide (BAP) in the K145-G150 loop of the VP4 lectin domain, permitting live monitoring. The recombinant virus was replication competent but showed a reduced fitness. We demonstrate that rRV/VP4-BAP infection, as opposed to rRV/wt infection, did not lead to a reorganized actin cytoskeleton as viroplasms formed were insensitive to drugs that depolymerize actin and inhibit myosin. Moreover, wild-type (wt) VP4, but not VP4-BAP, appeared to associate with actin filaments. Similarly, VP4 in coexpression with NSP5 and NSP2 induced a significant increase in the number of viroplasm-like structures. Interestingly, a small peptide mimicking loop K145-G150 rescued the phenotype of rRV/VP4-BAP by increasing its ability to form viroplasms and hence improve virus progeny formation. Collectively, these results provide a direct link between VP4 and the actin cytoskeleton to catalyze viroplasm assembly. IMPORTANCE The spike protein VP4 participates in diverse steps of the rotavirus (RV) life cycle, including virus-cell attachment, internalization, modulation of endocytosis, virion morphogenesis, and virus egress. Using reverse genetics, we constructed for the first time a recombinant RV, rRV/VP4-BAP, harboring a heterologous peptide in the lectin domain (loop K145-G150) of VP4. The rRV/VP4-BAP was replication competent but with reduced fitness due to a defect in the ability to reorganize the actin cytoskeleton, which affected the efficiency of viroplasm assembly. This defect was rescued by adding a permeable small-peptide mimicking the wild-type VP4 loop K145-G150. In addition to revealing a new role of VP4, our findings suggest that rRV harboring an engineered VP4 could be used as a new dual vaccination platform providing immunity against RV and additional heterologous antigens.
Collapse
|
35
|
Diebold O, Gonzalez V, Venditti L, Sharp C, Blake RA, Tan WS, Stevens J, Caddy S, Digard P, Borodavka A, Gaunt E. Using Species a Rotavirus Reverse Genetics to Engineer Chimeric Viruses Expressing SARS-CoV-2 Spike Epitopes. J Virol 2022; 96:e0048822. [PMID: 35758692 PMCID: PMC9327695 DOI: 10.1128/jvi.00488-22] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/22/2022] [Accepted: 05/31/2022] [Indexed: 02/02/2023] Open
Abstract
Species A rotavirus (RVA) vaccines based on live attenuated viruses are used worldwide in humans. The recent establishment of a reverse genetics system for rotoviruses (RVs) has opened the possibility of engineering chimeric viruses expressing heterologous peptides from other viral or microbial species in order to develop polyvalent vaccines. We tested the feasibility of this concept by two approaches. First, we inserted short SARS-CoV-2 spike peptides into the hypervariable region of the simian RV SA11 strain viral protein (VP) 4. Second, we fused the receptor binding domain (RBD) of the SARS-CoV-2 spike protein, or the shorter receptor binding motif (RBM) nested within the RBD, to the C terminus of nonstructural protein (NSP) 3 of the bovine RV RF strain, with or without an intervening Thosea asigna virus 2A (T2A) peptide. Mutating the hypervariable region of SA11 VP4 impeded viral replication, and for these mutants, no cross-reactivity with spike antibodies was detected. To rescue NSP3 mutants, we established a plasmid-based reverse genetics system for the bovine RV RF strain. Except for the RBD mutant that demonstrated a rescue defect, all NSP3 mutants delivered endpoint infectivity titers and exhibited replication kinetics comparable to that of the wild-type virus. In ELISAs, cell lysates of an NSP3 mutant expressing the RBD peptide showed cross-reactivity with a SARS-CoV-2 RBD antibody. 3D bovine gut enteroids were susceptible to infection by all NSP3 mutants, but cross-reactivity with SARS-CoV-2 RBD antibody was only detected for the RBM mutant. The tolerance of large SARS-CoV-2 peptide insertions at the C terminus of NSP3 in the presence of T2A element highlights the potential of this approach for the development of vaccine vectors targeting multiple enteric pathogens simultaneously. IMPORTANCE We explored the use of rotaviruses (RVs) to express heterologous peptides, using SARS-CoV-2 as an example. Small SARS-CoV-2 peptide insertions (<34 amino acids) into the hypervariable region of the viral protein 4 (VP4) of RV SA11 strain resulted in reduced viral titer and replication, demonstrating a limited tolerance for peptide insertions at this site. To test the RV RF strain for its tolerance for peptide insertions, we constructed a reverse genetics system. NSP3 was C-terminally tagged with SARS-CoV-2 spike peptides of up to 193 amino acids in length. With a T2A-separated 193 amino acid tag on NSP3, there was no significant effect on the viral rescue efficiency, endpoint titer, and replication kinetics. Tagged NSP3 elicited cross-reactivity with SARS-CoV-2 spike antibodies in ELISA. We highlight the potential for development of RV vaccine vectors targeting multiple enteric pathogens simultaneously.
Collapse
Affiliation(s)
- Ola Diebold
- Infection and Immunity Division, Roslin Institute, University of Edinburgh, Easter Bush Campus, Midlothian, United Kingdom
| | - Victoria Gonzalez
- Infection and Immunity Division, Roslin Institute, University of Edinburgh, Easter Bush Campus, Midlothian, United Kingdom
| | - Luca Venditti
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| | - Colin Sharp
- Infection and Immunity Division, Roslin Institute, University of Edinburgh, Easter Bush Campus, Midlothian, United Kingdom
| | - Rosemary A. Blake
- Infection and Immunity Division, Roslin Institute, University of Edinburgh, Easter Bush Campus, Midlothian, United Kingdom
| | - Wenfang S. Tan
- Infection and Immunity Division, Roslin Institute, University of Edinburgh, Easter Bush Campus, Midlothian, United Kingdom
| | - Joanne Stevens
- Infection and Immunity Division, Roslin Institute, University of Edinburgh, Easter Bush Campus, Midlothian, United Kingdom
| | - Sarah Caddy
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Paul Digard
- Infection and Immunity Division, Roslin Institute, University of Edinburgh, Easter Bush Campus, Midlothian, United Kingdom
| | - Alexander Borodavka
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| | - Eleanor Gaunt
- Infection and Immunity Division, Roslin Institute, University of Edinburgh, Easter Bush Campus, Midlothian, United Kingdom
| |
Collapse
|
36
|
Simsek C, Bloemen M, Jansen D, Descheemaeker P, Reynders M, Van Ranst M, Matthijnssens J. Rotavirus vaccine-derived cases in Belgium: Evidence for reversion of attenuating mutations and alternative causes of gastroenteritis. Vaccine 2022; 40:5114-5125. [PMID: 35871871 DOI: 10.1016/j.vaccine.2022.06.082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/15/2021] [Revised: 06/27/2022] [Accepted: 06/30/2022] [Indexed: 02/07/2023]
Abstract
Since the introduction of live-attenuated rotavirus vaccines in Belgium in 2006, surveillance has routinely detected rotavirus vaccine-derived strains. However, their genomic landscape and potential role in gastroenteritis have not been thoroughly investigated. We compared VP7 and VP4 nucleotide sequences obtained from rotavirus surveillance with the Rotarix vaccine sequence. As a result, we identified 80 vaccine-derived strains in 5125 rotavirus-positive infants with gastroenteritis from 2007 to 2018. Using both viral metagenomics and reverse transcription qPCR, we evaluated the vaccine strains and screened for co-infecting enteropathogens. Among the 45 patients with known vaccination status, 39 were vaccinated and 87% received the vaccine less than a month before the gastroenteritis episode. Reconstruction of 30 near complete vaccine-derived genomes revealed 0-11 mutations per genome, with 88% of them being non-synonymous. This, in combination with several shared amino acid changes among strains, pointed at selection of minor variant(s) present in the vaccine. We also found that some of these substitutions were true revertants (e.g., F167L on VP4, and I45T on NSP4). Finally, co-infections with known (e.g., Clostridioides difficile and norovirus) and divergent or emerging (e.g., human parechovirus A1, salivirus A2) pathogens were detected, and we estimated that 35% of the infants likely had gastroenteritis due to a 'non-rotavirus' cause. Conversely, we could not rule out the vaccine-derived gastroenteritis in over half of the cases. Continued studies inspecting reversion to pathogenicity should monitor the long-time safety of live-attenuated rotavirus vaccines. All in all, the complementary approach with NGS and qPCR provided a better understanding of rotavirus vaccine strain evolution in the Belgian population and epidemiology of co-infecting enteropathogens in suspected rotavirus vaccine-derived gastroenteritis cases.
Collapse
Affiliation(s)
- Ceren Simsek
- KU Leuven - University of Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Leuven, Belgium
| | - Mandy Bloemen
- KU Leuven - University of Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Leuven, Belgium
| | - Daan Jansen
- KU Leuven - University of Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Leuven, Belgium
| | - Patrick Descheemaeker
- Department of Laboratory Medicine, Medical Microbiology, AZ Sint-Jan, Brugge-Oostende AV, Bruges, Belgium
| | - Marijke Reynders
- Department of Laboratory Medicine, Medical Microbiology, AZ Sint-Jan, Brugge-Oostende AV, Bruges, Belgium
| | - Marc Van Ranst
- KU Leuven - University of Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Leuven, Belgium
| | - Jelle Matthijnssens
- KU Leuven - University of Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Leuven, Belgium.
| |
Collapse
|
37
|
Hu L, Salmen W, Sankaran B, Lasanajak Y, Smith DF, Crawford SE, Estes MK, Prasad BVV. Novel fold of rotavirus glycan-binding domain predicted by AlphaFold2 and determined by X-ray crystallography. Commun Biol 2022; 5:419. [PMID: 35513489 PMCID: PMC9072675 DOI: 10.1038/s42003-022-03357-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/07/2021] [Accepted: 04/12/2022] [Indexed: 02/08/2023] Open
Abstract
The VP8* domain of spike protein VP4 in group A and C rotaviruses, which cause epidemic gastroenteritis in children, exhibits a conserved galectin-like fold for recognizing glycans during cell entry. In group B rotavirus, which causes significant diarrheal outbreaks in adults, the VP8* domain (VP8*B) surprisingly lacks sequence similarity with VP8* of group A or group C rotavirus. Here, by using the recently developed AlphaFold2 for ab initio structure prediction and validating the predicted model by determining a 1.3-Å crystal structure, we show that VP8*B exhibits a novel fold distinct from the galectin fold. This fold with a β-sheet clasping an α-helix represents a new fold for glycan recognition based on glycan array screening, which shows that VP8*B recognizes glycans containing N-acetyllactosamine moiety. Although uncommon, our study illustrates how evolution can incorporate structurally distinct folds with similar functionality in a homologous protein within the same virus genus.
Collapse
Affiliation(s)
- Liya Hu
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX, USA.
| | - Wilhelm Salmen
- 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
| | - Yi Lasanajak
- Emory Glycomics and Molecular Interactions Core (EGMIC), Emory University School of Medicine, Atlanta, GA, USA
| | - David F Smith
- Emory Glycomics and Molecular Interactions Core (EGMIC), Emory University School of Medicine, Atlanta, GA, 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
| | - 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
|
38
|
Mwangi PN, Page NA, Seheri ML, Mphahlele MJ, Nadan S, Esona MD, Kumwenda B, Kamng'ona AW, Donato CM, Steele DA, Ndze VN, Dennis FE, Jere KC, Nyaga MM. Evolutionary changes between pre- and post-vaccine South African group A G2P[4] rotavirus strains, 2003-2017. Microb Genom 2022; 8. [PMID: 35446251 PMCID: PMC9453071 DOI: 10.1099/mgen.0.000809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/18/2022] Open
Abstract
The transient upsurge of G2P[4] group A rotavirus (RVA) after Rotarix vaccine introduction in several countries has been a matter of concern. To gain insight into the diversity and evolution of G2P[4] strains in South Africa pre- and post-RVA vaccination introduction, whole-genome sequencing was performed for RVA positive faecal specimens collected between 2003 and 2017 and samples previously sequenced were obtained from GenBank (n=103; 56 pre- and 47 post-vaccine). Pre-vaccine G2 sequences predominantly clustered within sub-lineage IVa-1. In contrast, post-vaccine G2 sequences clustered mainly within sub-lineage IVa-3, whereby a radical amino acid (AA) substitution, S15F, was observed between the two sub-lineages. Pre-vaccine P[4] sequences predominantly segregated within sub-lineage IVa while post-vaccine sequences clustered mostly within sub-lineage IVb, with a radical AA substitution R162G. Both S15F and R162G occurred outside recognised antigenic sites. The AA residue at position 15 is found within the signal sequence domain of Viral Protein 7 (VP7) involved in translocation of VP7 into endoplasmic reticulum during infection process. The 162 AA residue lies within the hemagglutination domain of Viral Protein 4 (VP4) engaged in interaction with sialic acid-containing structure during attachment to the target cell. Free energy change analysis on VP7 indicated accumulation of stable point mutations in both antigenic and non-antigenic regions. The segregation of South African G2P[4] strains into pre- and post-vaccination sub-lineages is likely due to erstwhile hypothesized stepwise lineage/sub-lineage evolution of G2P[4] strains rather than RVA vaccine introduction. Our findings reinforce the need for continuous whole-genome RVA surveillance and investigation of contribution of AA substitutions in understanding the dynamic G2P[4] epidemiology.
Collapse
Affiliation(s)
- Peter N Mwangi
- Next Generation Sequencing Unit and Division of Virology, Faculty of Health Sciences, University of the Free State, Bloemfontein 9300, South Africa
| | - Nicola A Page
- Centre for Enteric Disease, National Institute for Communicable Diseases, Private Bag X4, Sandringham, 2131, Johannesburg, South Africa.,Department of Medical Virology, Faculty of Health Sciences, University of Pretoria, Private Bag X323, Arcadia, 0007, Pretoria, South Africa
| | - Mapaseka L Seheri
- Diarrheal Pathogens Research Unit, Sefako Makgatho Health Sciences University, Medunsa 0204, Pretoria, South Africa
| | - M Jeffrey Mphahlele
- Diarrheal Pathogens Research Unit, Sefako Makgatho Health Sciences University, Medunsa 0204, Pretoria, South Africa.,Office of the Deputy Vice Chancellor for Research and Innovation, North-West University, Potchefstroom 2351, South Africa.,South African Medical Research Council, Pretoria 0001, South Africa
| | - Sandrama Nadan
- Centre for Enteric Disease, National Institute for Communicable Diseases, Private Bag X4, Sandringham, 2131, Johannesburg, South Africa
| | - Mathew D Esona
- Diarrheal Pathogens Research Unit, Sefako Makgatho Health Sciences University, Medunsa 0204, Pretoria, South Africa
| | - Benjamin Kumwenda
- Department of Biomedical Sciences, School of Life Sciences and Applied Health Professions, Kamuzu University of Health Sciences, Private Bag 360, Chichiri, Blantyre 3, Malawi
| | - Arox W Kamng'ona
- Department of Biomedical Sciences, School of Life Sciences and Applied Health Professions, Kamuzu University of Health Sciences, Private Bag 360, Chichiri, Blantyre 3, Malawi
| | - Celeste M Donato
- Department of Medical Laboratory Sciences, School of Life Sciences and Applied Health Professions, Kamuzu University of Health Sciences, Private Bag 360, Chichiri, Blantyre3, Malawi.,Enteric Diseases Group, Murdoch Children's Research Institute, 50 Flemington Road, Parkville, Melboune 3052, Australia.,Department of Paediatrics, the University of Melbourne, Parkville 3010, Australia
| | - Duncan A Steele
- Diarrheal Pathogens Research Unit, Sefako Makgatho Health Sciences University, Medunsa 0204, Pretoria, South Africa
| | - Valantine N Ndze
- Faculty of Health Sciences, University of Buea, P.O Box 63 Buea, Cameroon
| | - Francis E Dennis
- Department of Electron Microscopy and Histopathology, Noguchi Memorial Institute for Medical Research, University of Ghana, P.O Box LG581, Legon, Ghana
| | - Khuzwayo C Jere
- Center for Global Vaccine Research, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, L697BE, Liverpool, UK.,Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Blantyre 312225, Malawi
| | - Martin M Nyaga
- Next Generation Sequencing Unit and Division of Virology, Faculty of Health Sciences, University of the Free State, Bloemfontein 9300, South Africa
| |
Collapse
|
39
|
|
40
|
Zhang Y, Cui Y, Sun J, Zhou ZH. Multiple conformations of trimeric spikes visualized on a non-enveloped virus. Nat Commun 2022; 13:550. [PMID: 35087065 PMCID: PMC8795420 DOI: 10.1038/s41467-022-28114-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/14/2021] [Accepted: 01/10/2022] [Indexed: 11/18/2022] Open
Abstract
Many viruses utilize trimeric spikes to gain entry into host cells. However, without in situ structures of these trimeric spikes, a full understanding of this dynamic and essential process of viral infections is not possible. Here we present four in situ and one isolated cryoEM structures of the trimeric spike of the cytoplasmic polyhedrosis virus, a member of the non-enveloped Reoviridae family and a virus historically used as a model in the discoveries of RNA transcription and capping. These structures adopt two drastically different conformations, closed spike and opened spike, which respectively represent the penetration-inactive and penetration-active states. Each spike monomer has four domains: N-terminal, body, claw, and C-terminal. From closed to opened state, the RGD motif-containing C-terminal domain is freed to bind integrins, and the claw domain rotates to expose and project its membrane insertion loops into the cellular membrane. Comparison between turret vertices before and after detachment of the trimeric spike shows that the trimeric spike anchors its N-terminal domain in the iris of the pentameric RNA-capping turret. Sensing of cytosolic S-adenosylmethionine (SAM) and adenosine triphosphate (ATP) by the turret triggers a cascade of events: opening of the iris, detachment of the spike, and initiation of endogenous transcription. Zhang and Cui et al. present in situ cryoEM structures of the trimeric spike of cytoplasmic polyhedrosis virus in both open and close conformations, and demonstrate that spike detachment from the capsid is triggered by the presence of SAM and ATP.
Collapse
Affiliation(s)
- Yinong Zhang
- Subtropical Sericulture and Mulberry Resources Protection and Safety Engineering Research Center, Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, 510642, China.,California Nanosystems Institute, University of California, Los Angeles (UCLA), Los Angeles, CA, 90095, USA.,Department of Microbiology, Immunology and Molecular Genetics, UCLA, Los Angeles, CA, 90095, USA
| | - Yanxiang Cui
- California Nanosystems Institute, University of California, Los Angeles (UCLA), Los Angeles, CA, 90095, USA
| | - Jingchen Sun
- Subtropical Sericulture and Mulberry Resources Protection and Safety Engineering Research Center, Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, 510642, China. .,Department of Microbiology, Immunology and Molecular Genetics, UCLA, Los Angeles, CA, 90095, USA.
| | - Z Hong Zhou
- California Nanosystems Institute, University of California, Los Angeles (UCLA), Los Angeles, CA, 90095, USA. .,Department of Microbiology, Immunology and Molecular Genetics, UCLA, Los Angeles, CA, 90095, USA.
| |
Collapse
|
41
|
Esona MD, Gautam R, Chhabra P, Vinjé J, Bowen MD, Burke RM. Gastrointestinal Tract Infections: Viruses. ENCYCLOPEDIA OF INFECTION AND IMMUNITY 2022:82-106. [DOI: 10.1016/b978-0-12-818731-9.00217-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 01/05/2025]
|
42
|
Amimo JO, Raev SA, Chepngeno J, Mainga AO, Guo Y, Saif L, Vlasova AN. Rotavirus Interactions With Host Intestinal Epithelial Cells. Front Immunol 2021; 12:793841. [PMID: 35003114 PMCID: PMC8727603 DOI: 10.3389/fimmu.2021.793841] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/12/2021] [Accepted: 12/06/2021] [Indexed: 12/13/2022] Open
Abstract
Rotavirus (RV) is the foremost enteric pathogen associated with severe diarrheal illness in young children (<5years) and animals worldwide. RV primarily infects mature enterocytes in the intestinal epithelium causing villus atrophy, enhanced epithelial cell turnover and apoptosis. Intestinal epithelial cells (IECs) being the first physical barrier against RV infection employs a range of innate immune strategies to counteract RVs invasion, including mucus production, toll-like receptor signaling and cytokine/chemokine production. Conversely, RVs have evolved numerous mechanisms to escape/subvert host immunity, seizing translation machinery of the host for effective replication and transmission. RV cell entry process involve penetration through the outer mucus layer, interaction with cell surface molecules and intestinal microbiota before reaching the IECs. For successful cell attachment and entry, RVs use sialic acid, histo-blood group antigens, heat shock cognate protein 70 and cell-surface integrins as attachment factors and/or (co)-receptors. In this review, a comprehensive summary of the existing knowledge of mechanisms underlying RV-IECs interactions, including the role of gut microbiota, during RV infection is presented. Understanding these mechanisms is imperative for developing efficacious strategies to control RV infections, including development of antiviral therapies and vaccines that target specific immune system antagonists within IECs.
Collapse
Affiliation(s)
- Joshua Oluoch Amimo
- Center for Food Animal Health, Department of Animal Sciences, College of Food Agricultural and Environmental Sciences, The Ohio State University, Wooster, OH, United States
- Department of Animal Production, Faculty of Veterinary Medicine, University of Nairobi, Nairobi, Kenya
| | - Sergei Alekseevich Raev
- Center for Food Animal Health, Department of Animal Sciences, College of Food Agricultural and Environmental Sciences, The Ohio State University, Wooster, OH, United States
| | - Juliet Chepngeno
- Center for Food Animal Health, Department of Animal Sciences, College of Food Agricultural and Environmental Sciences, The Ohio State University, Wooster, OH, United States
| | - Alfred Omwando Mainga
- Center for Food Animal Health, Department of Animal Sciences, College of Food Agricultural and Environmental Sciences, The Ohio State University, Wooster, OH, United States
- Department of Public Health, Pharmacology and Toxicology, Faculty of Veterinary Medicine, University of Nairobi, Nairobi, Kenya
| | - Yusheng Guo
- Center for Food Animal Health, Department of Animal Sciences, College of Food Agricultural and Environmental Sciences, The Ohio State University, Wooster, OH, United States
| | - Linda Saif
- Center for Food Animal Health, Department of Animal Sciences, College of Food Agricultural and Environmental Sciences, The Ohio State University, Wooster, OH, United States
| | - Anastasia N. Vlasova
- Center for Food Animal Health, Department of Animal Sciences, College of Food Agricultural and Environmental Sciences, The Ohio State University, Wooster, OH, United States
| |
Collapse
|
43
|
Patra U, Mukhopadhyay U, Mukherjee A, Dutta S, Chawla-Sarkar M. Treading a HOSTile path: Mapping the dynamic landscape of host cell-rotavirus interactions to explore novel host-directed curative dimensions. Virulence 2021; 12:1022-1062. [PMID: 33818275 PMCID: PMC8023246 DOI: 10.1080/21505594.2021.1903198] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/21/2020] [Revised: 01/20/2021] [Accepted: 03/10/2021] [Indexed: 12/27/2022] Open
Abstract
Viruses are intracellular pathogens and are dependent on host cellular resources to carry out their cycles of perpetuation. Obtaining an integrative view of host-virus interaction is of utmost importance to understand the complex and dynamic interplay between viral components and host machineries. Besides its obvious scholarly significance, a comprehensive host-virus interaction profile also provides a platform where from host determinants of pro-viral and antiviral importance can be identified and further be subjected to therapeutic intervention. Therefore, adjunct to conventional methods of prophylactic vaccination and virus-directed antivirals, this host-targeted antiviral approach holds promising therapeutic potential. In this review, we present a comprehensive landscape of host cellular reprogramming in response to infection with rotavirus (RV) which causes profuse watery diarrhea in neonates and infants. In addition, an emphasis is given on how host determinants are either usurped or subverted by RV in course of infection and how therapeutic manipulation of specific host factors can effectively modulate the RV life cycle.
Collapse
Affiliation(s)
- Upayan Patra
- Division of Virology, National Institute of Cholera and Enteric Diseases, Beliaghata, Kolkata, India
| | - Urbi Mukhopadhyay
- Division of Virology, National Institute of Cholera and Enteric Diseases, Beliaghata, Kolkata, India
| | - Arpita Mukherjee
- Division of Virology, National Institute of Cholera and Enteric Diseases, Beliaghata, Kolkata, India
| | - Shanta Dutta
- Division of Bacteriology, National Institute of Cholera and Enteric Diseases, Beliaghata, Kolkata, India
| | - Mamta Chawla-Sarkar
- Division of Virology, National Institute of Cholera and Enteric Diseases, Beliaghata, Kolkata, India
| |
Collapse
|
44
|
Ranshing S, Ganorkar N, Ramji S, Gopalkrishna V. Complete genomic analysis of uncommon G12P[11] rotavirus causing a nosocomial outbreak of acute diarrhea in the newborns in New Delhi, India. J Med Virol 2021; 94:2613-2623. [PMID: 34811775 DOI: 10.1002/jmv.27468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/18/2021] [Accepted: 11/19/2021] [Indexed: 11/06/2022]
Abstract
Rotaviruses (RVs) are the major causative agents of acute gastroenteritis in children, but in neonates, RV infections are generally nosocomial in origin and mostly asymptomatic. However, there have been infrequent reports of nosocomial outbreaks of clinical disease in this population. In this study, we describe uncommon RV genotype; G12P[11] associated with an outbreak of acute gastroenteritis in the neonatal ward and neonatal intensive care unit (NICU) in New Delhi, North India. Full-genome analyses of the pathogenic G12P[11] strain was carried out to map the genotype constellation and further to explore the variations in the antigenic epitopes on the immunodominant VP7 and VP4 proteins, the amino acid sequences were compared with neonatal strains; ROTAVAC® (G9P[11]) and asymptomatic G12P[11] and also other G/P-type matched strains. The study revealed G12-P[11]-I1-R1-C1-M1-A1-N1-T1-E1-H1 human Wa-like genotype constellation and highlights evidence of gene reassortment. No significant differences were observed in the sequences of structural (except VP3) and nonstructural encoding genes of G12P[11] strains recovered from symptomatic and asymptomatic neonates. Presence of additional N-linked glycosylation site was noted in the G12 strains, as a consequence of a change from Asp→Asn at amino acid position 238. Interestingly, only two and four amino acids substitution within the 7-1a and 8-1 antigenic epitope were observed, respectively, compared with asymptomatic G12P[11] strain. The study emphasizes the importance of close monitoring of RV outbreaks in neonates for early alarming of novel strain.
Collapse
Affiliation(s)
- Sujata Ranshing
- Enteric Viruses Group, ICMR-National Institute of Virology, Pune, India
| | - Nital Ganorkar
- Enteric Viruses Group, ICMR-National Institute of Virology, Pune, India
| | - Siddarth Ramji
- Maulana Azad Medical College and Lok Nayak Hospital, New Delhi, India
| | | |
Collapse
|
45
|
Manouana GP, Niendorf S, Tomazatos A, Mbong Ngwese M, Nzamba Maloum M, Nguema Moure PA, Bingoulou Matsougou G, Ategbo S, Rossatanga EG, Bock CT, Borrmann S, Mordmüller B, Eibach D, Kremsner PG, Velavan TP, Adegnika AA. Molecular surveillance and genetic divergence of rotavirus A antigenic epitopes in Gabonese children with acute gastroenteritis. EBioMedicine 2021; 73:103648. [PMID: 34706308 PMCID: PMC8551588 DOI: 10.1016/j.ebiom.2021.103648] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/08/2021] [Revised: 10/08/2021] [Accepted: 10/11/2021] [Indexed: 01/21/2023] Open
Abstract
Background Rotavirus A (RVA) causes acute gastroenteritis in children <5 years of age in sub-Saharan Africa. In this study, we described the epidemiology and genetic diversity of RVA infecting Gabonese children and examined the antigenic variability of circulating strains in relation to available vaccine strains to maximize the public health benefits of introducing rotavirus vaccine through the Expanded Programme on Immunization (EPI) in Gabon. Methods Stool samples were collected consecutively between April 2018 and November 2019 from all hospitalized children <5 years with gastroenteritis and community controls without gastroenteritis. Children were tested for rotavirus A by quantitative RT-PCR and subsequently sequenced to identify circulating rotavirus A genotypes in the most vulnerable population. The VP7 and VP4 (VP8*) antigenic epitopes were mapped to homologs of vaccine strains to assess structural variability and potential impact on antigenicity. Findings Infections were mostly acquired during the dry season. Rotavirus A was detected in 98/177 (55%) hospitalized children with gastroenteritis and 14/67 (21%) of the control children. The most common RVA genotypes were G1 (18%), G3 (12%), G8 (18%), G9 (2%), G12 (25%), with G8 and G9 reported for the first time in Gabon. All were associated either with P[6] (31%) or P[8] (38%) genotypes. Several non-synonymous substitutions were observed in the antigenic epitopes of VP7 (positions 94 and 147) and VP8* (positions 89, 116, 146 and 150), which may modulate the elicited immune responses. Interpretation This study contributes to the epidemiological surveillance of rotavirus A required before the introduction of rotavirus vaccination in the EPI for Gabonese children.
Collapse
Affiliation(s)
- Gédéon Prince Manouana
- Institute of Tropical Medicine, University of Tübingen, Wilhelmstrasse 27, Tübingen 72074, Germany; Centre de Recherche Médicales de Lambaréné, Lambaréné, Gabon
| | - Sandra Niendorf
- Department of Infectious Diseases, Robert Koch Institute, Berlin, Germany
| | - Alexandru Tomazatos
- Institute of Tropical Medicine, University of Tübingen, Wilhelmstrasse 27, Tübingen 72074, Germany; Department of Arbovirology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | | | | | | | - Gedeon Bingoulou Matsougou
- Département de Pédiatrie, Faculté de Médecine, Université des Sciences de la Santé (USS), BP 4009, Libreville, Gabon
| | - Simon Ategbo
- Département de Pédiatrie, Faculté de Médecine, Université des Sciences de la Santé (USS), BP 4009, Libreville, Gabon
| | | | - C Thomas Bock
- Institute of Tropical Medicine, University of Tübingen, Wilhelmstrasse 27, Tübingen 72074, Germany; Department of Infectious Diseases, Robert Koch Institute, Berlin, Germany
| | - Steffen Borrmann
- Institute of Tropical Medicine, University of Tübingen, Wilhelmstrasse 27, Tübingen 72074, Germany; Centre de Recherche Médicales de Lambaréné, Lambaréné, Gabon; German Center for Infection Research (DZIF), Tübingen, Germany
| | - Benjamin Mordmüller
- Institute of Tropical Medicine, University of Tübingen, Wilhelmstrasse 27, Tübingen 72074, Germany; Centre de Recherche Médicales de Lambaréné, Lambaréné, Gabon; Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, the Netherland
| | - Daniel Eibach
- Infectious Disease Epidemiology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Peter G Kremsner
- Institute of Tropical Medicine, University of Tübingen, Wilhelmstrasse 27, Tübingen 72074, Germany; Centre de Recherche Médicales de Lambaréné, Lambaréné, Gabon; German Center for Infection Research (DZIF), Tübingen, Germany
| | - Thirumalaisamy P Velavan
- Institute of Tropical Medicine, University of Tübingen, Wilhelmstrasse 27, Tübingen 72074, Germany; Vietnamese-German Center for Medical Research (VG-CARE), Hanoi, Viet Nam.
| | - Ayola Akim Adegnika
- Institute of Tropical Medicine, University of Tübingen, Wilhelmstrasse 27, Tübingen 72074, Germany; Centre de Recherche Médicales de Lambaréné, Lambaréné, Gabon; Centre Hospitalier Régional Georges Rawiri de Lambaréné, Lambaréné, Gabon; Fondation pour la Recherche Scientifique, Cotonou, Bénin.
| |
Collapse
|
46
|
Dang L, Su Y, Qi J, Wu Z, Li D, Wang M, Zhang Q, Wang H, Bai R, Duan Z, Sun X. Structural and functional characterization of bovine G1P[5] rotavirus VP8* protein. Virology 2021; 563:116-125. [PMID: 34509703 DOI: 10.1016/j.virol.2021.08.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/04/2021] [Revised: 08/07/2021] [Accepted: 08/17/2021] [Indexed: 11/26/2022]
Abstract
The widely used rotavirus (RV) vaccine, Rotateq, contained reassortment strains of human and bovine G1/2/3/4P[5] RVs. The functional and structural features of bovine G1P[5] VP8* were investigated. Bovine G1P[5] VP8* was identified to interact with sialic acids and sialic acid-containing glycans. In addition, P[5] VP8* recognized α-Gal histo-blood group antigens (HBGAs). Bovine G1P[5] VP8* did not hemagglutinate the tested red blood cells. The crystal structure of P[5] VP8* was determined at 1.7 Å. Structural superimposition revealed that P[5] VP8* was most close to human P[8] VP8*, while much further to VP8*s of porcine P[7] and rhesus P[3]. Sequence alignment showed that amino acids of the putative glycan binding site in P[5] VP8* were different to those in P[3]/P[7] VP8*s, indicating that P[5] VP8* may interact with glycans using different mechanism. This study provided more understanding of P[5] RV infection and the interactions of RV VP8* and glycans.
Collapse
Affiliation(s)
- Lei Dang
- National Health Commission Key Laboratory for Medical Virology and Viral Diseases, Beijing, 102206, China; National Institute for Viral Disease Control and Prevention, China CDC, Beijing, 102206, China; Inner Mongolia Hospital of Traditional Chinese Medicine, Hohhot, 010059, China
| | - Yunxi Su
- National Health Commission Key Laboratory for Medical Virology and Viral Diseases, Beijing, 102206, China; National Institute for Viral Disease Control and Prevention, China CDC, Beijing, 102206, China
| | - Jianxun Qi
- Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Zheng Wu
- National Health Commission Key Laboratory for Medical Virology and Viral Diseases, Beijing, 102206, China; National Institute for Viral Disease Control and Prevention, China CDC, Beijing, 102206, China
| | - Dandi Li
- National Health Commission Key Laboratory for Medical Virology and Viral Diseases, Beijing, 102206, China; National Institute for Viral Disease Control and Prevention, China CDC, Beijing, 102206, China
| | - Mengxuan Wang
- National Health Commission Key Laboratory for Medical Virology and Viral Diseases, Beijing, 102206, China; National Institute for Viral Disease Control and Prevention, China CDC, Beijing, 102206, China
| | - Qing Zhang
- National Health Commission Key Laboratory for Medical Virology and Viral Diseases, Beijing, 102206, China; National Institute for Viral Disease Control and Prevention, China CDC, Beijing, 102206, China
| | - Hong Wang
- National Health Commission Key Laboratory for Medical Virology and Viral Diseases, Beijing, 102206, China; National Institute for Viral Disease Control and Prevention, China CDC, Beijing, 102206, China
| | - Ruixia Bai
- Inner Mongolia Medical University, Hohhot, 010059, China
| | - Zhaojun Duan
- National Health Commission Key Laboratory for Medical Virology and Viral Diseases, Beijing, 102206, China; National Institute for Viral Disease Control and Prevention, China CDC, Beijing, 102206, China.
| | - Xiaoman Sun
- National Health Commission Key Laboratory for Medical Virology and Viral Diseases, Beijing, 102206, China; National Institute for Viral Disease Control and Prevention, China CDC, Beijing, 102206, China.
| |
Collapse
|
47
|
Liu X, Yan N, Yue H, Wang Y, Zhang B, Tang C. Detection and molecular characteristics of bovine rotavirus A in dairy calves in China. J Vet Sci 2021; 22:e69. [PMID: 34423605 PMCID: PMC8460460 DOI: 10.4142/jvs.2021.22.e69] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/05/2021] [Revised: 07/11/2021] [Accepted: 07/26/2021] [Indexed: 11/29/2022] Open
Abstract
Background Bovine group A rotavirus (BoRVA) is a major cause of severe gastroenteritis in newborn dairy calves. Only one study has investigated the G and P genotypes among dairy calves in a few regions of China, which were G6 and P[5]. Therefore, data on the prevalence and molecular characteristics of BoRVA in dairy calves in China remains limited. Objectives The purpose of this study was to investigate the prevalence and molecular characteristics of BoRVA in dairy calves in China. Methods 269 dairy calves diarrheic samples from 23 farms in six provinces in China were collected to detect BoRVA using reverse transcription polymerase chain reaction. Results 71% of samples were determined to be BoRVA-positive. Two G genotypes (G6, G10) and two P genotypes (P[1], P[5]) were identified, and G6P[1] BoRVA was the predominant strain. Moreover, the VP7 and VP4 gene sequences of these dairy calf BoRVA strains revealed abundant genetic diversity. Interestingly, eight out of 17 complete G6 VP7 sequences were clustered into G6 lineage VI and analysis showed the strains were closely related to Chinese yak BoRVA strains. Conclusions The results of this study show that BoRVA circulates widely among dairy calves in China, and the dominant genotype in circulation is G6P[1], first report on molecular characteristics of complete P[5] VP4 genes in chinese dairy calves. These results will help us to further understand the prevalence and genetic evolution of BoRVA among dairy calves in China and, thus, prevent the disease more effectively.
Collapse
Affiliation(s)
- Xiaoying Liu
- College of Animal & Veterinary Sciences, Southwest Minzu University, Chengdu 610041, China
| | - Nan Yan
- College of Animal & Veterinary Sciences, Southwest Minzu University, Chengdu 610041, China
| | - Hua Yue
- College of Animal & Veterinary Sciences, Southwest Minzu University, Chengdu 610041, China.,Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Chengdu 610041, China
| | - Yuanwei Wang
- College of Animal & Veterinary Sciences, Southwest Minzu University, Chengdu 610041, China
| | - Bin Zhang
- College of Animal & Veterinary Sciences, Southwest Minzu University, Chengdu 610041, China
| | - Cheng Tang
- College of Animal & Veterinary Sciences, Southwest Minzu University, Chengdu 610041, China.,Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Chengdu 610041, China.
| |
Collapse
|
48
|
Yang J, Park J, Koehler M, Simpson J, Luque D, Rodríguez JM, Alsteens D. Rotavirus Binding to Cell Surface Receptors Directly Recruiting α
2
Integrin. ADVANCED NANOBIOMED RESEARCH 2021. [DOI: 10.1002/anbr.202100077] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 01/21/2023] Open
Affiliation(s)
- Jinsung Yang
- Louvain Institute of Biomolecular Science and Technology Université Catholique de Louvain Louvain-la-Neuve 1348 Belgium
| | | | - Melanie Koehler
- Louvain Institute of Biomolecular Science and Technology Université Catholique de Louvain Louvain-la-Neuve 1348 Belgium
| | - Joshua Simpson
- Louvain Institute of Biomolecular Science and Technology Université Catholique de Louvain Louvain-la-Neuve 1348 Belgium
| | - Daniel Luque
- Centro Nacional de Microbiología/ISCIII Madrid 28220 Spain
| | | | - David Alsteens
- Louvain Institute of Biomolecular Science and Technology Université Catholique de Louvain Louvain-la-Neuve 1348 Belgium
| |
Collapse
|
49
|
Sun X, Li D, Duan Z. Structural Basis of Glycan Recognition of Rotavirus. Front Mol Biosci 2021; 8:658029. [PMID: 34307449 PMCID: PMC8296142 DOI: 10.3389/fmolb.2021.658029] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/24/2021] [Accepted: 06/21/2021] [Indexed: 11/13/2022] Open
Abstract
Rotavirus (RV) is an important pathogen causing acute gastroenteritis in young humans and animals. Attachment to the host receptor is a crucial step for the virus infection. The recent advances in illustrating the interactions between RV and glycans promoted our understanding of the host range and epidemiology of RVs. VP8*, the distal region of the RV outer capsid spike protein VP4, played a critical role in the glycan recognition. Group A RVs were classified into different P genotypes based on the VP4 sequences and recognized glycans in a P genotype-dependent manner. Glycans including sialic acid, gangliosides, histo-blood group antigens (HBGAs), and mucin cores have been reported to interact with RV VP8*s. The glycan binding specificities of VP8*s of different RV genotypes have been studied. Here, we mainly discussed the structural basis for the interactions between RV VP8*s and glycans, which provided molecular insights into the receptor recognition and host tropism, offering new clues to the design of RV vaccine and anti-viral agents.
Collapse
Affiliation(s)
- Xiaoman Sun
- National Health Commission Key Laboratory for Medical Virology and Viral Diseases, Beijing, China.,National Institute for Viral Disease Control and Prevention, China CDC, Beijing, China
| | - Dandi Li
- National Health Commission Key Laboratory for Medical Virology and Viral Diseases, Beijing, China.,National Institute for Viral Disease Control and Prevention, China CDC, Beijing, China
| | - Zhaojun Duan
- National Health Commission Key Laboratory for Medical Virology and Viral Diseases, Beijing, China.,National Institute for Viral Disease Control and Prevention, China CDC, Beijing, China
| |
Collapse
|
50
|
Li D, Wang M, Mao T, Wang M, Zhang Q, Wang H, Pang L, Sun X, Duan Z. The Functional Characterization of Bat and Human P[3] Rotavirus VP8*s. Virol Sin 2021; 36:1187-1196. [PMID: 34057680 PMCID: PMC8165343 DOI: 10.1007/s12250-021-00400-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/06/2021] [Accepted: 04/12/2021] [Indexed: 11/11/2022] Open
Abstract
P[3] rotavirus (RV) has been identified in many species, including human, simian, dog, and bat. Several glycans, including sialic acid, histo-blood group antigens (HBGAs) are reported as RV attachment factors. The glycan binding specificity of different P[3] RV VP8*s were investigated in this study. Human HCR3A and dog P[3] RV VP8*s recognized glycans with terminal sialic acid and hemagglutinated the red blood cells, while bat P[3] VP8* showed neither binding to glycans nor hemagglutination. However, the bat P[3] VP8* mutant of C189Y obtained the ability to hemagglutinate the red blood cells, while human P[3] HCR3A/M2-102 mutants of Y189C lost the ability. Sequence alignment and structural analysis indicated that residue 189 played an important role in the ligand recognition and may contribute to the cross-species transmission. Structural superimposition exhibited that bat P[3] VP8* model was quite different from the simian P[3] Rhesus rotavirus (RRV) P[3] VP8*, indicating that bat P[3] RV was relatively distinct and partially contributed to the no binding to tested glycans. These results promote our understanding of P[3] VP8*/glycans interactions and the potential transmission of bat/human P[3] RVs, offering more insight into the RV infection and prevalence.
Collapse
Affiliation(s)
- Dandi Li
- National Health Commission Key Laboratory for Medical Virology and Viral Diseases, Beijing, 102206, China.,National Institute for Viral Disease Control and Prevention, China CDC, Beijing, 102206, China
| | - Mengxuan Wang
- National Health Commission Key Laboratory for Medical Virology and Viral Diseases, Beijing, 102206, China.,National Institute for Viral Disease Control and Prevention, China CDC, Beijing, 102206, China
| | - Tongyao Mao
- National Health Commission Key Laboratory for Medical Virology and Viral Diseases, Beijing, 102206, China.,National Institute for Viral Disease Control and Prevention, China CDC, Beijing, 102206, China
| | - Mingwen Wang
- National Health Commission Key Laboratory for Medical Virology and Viral Diseases, Beijing, 102206, China.,National Institute for Viral Disease Control and Prevention, China CDC, Beijing, 102206, China
| | - Qing Zhang
- National Health Commission Key Laboratory for Medical Virology and Viral Diseases, Beijing, 102206, China.,National Institute for Viral Disease Control and Prevention, China CDC, Beijing, 102206, China
| | - Hong Wang
- National Health Commission Key Laboratory for Medical Virology and Viral Diseases, Beijing, 102206, China.,National Institute for Viral Disease Control and Prevention, China CDC, Beijing, 102206, China
| | - Lili Pang
- National Health Commission Key Laboratory for Medical Virology and Viral Diseases, Beijing, 102206, China.,National Institute for Viral Disease Control and Prevention, China CDC, Beijing, 102206, China
| | - Xiaoman Sun
- National Health Commission Key Laboratory for Medical Virology and Viral Diseases, Beijing, 102206, China. .,National Institute for Viral Disease Control and Prevention, China CDC, Beijing, 102206, China.
| | - Zhaojun Duan
- National Health Commission Key Laboratory for Medical Virology and Viral Diseases, Beijing, 102206, China. .,National Institute for Viral Disease Control and Prevention, China CDC, Beijing, 102206, China.
| |
Collapse
|