1
|
Pham S, Zhao B, Neetu N, Sankaran B, Patil K, Ramani S, Song Y, Estes MK, Palzkill T, Prasad BVV. Conformational flexibility is a critical factor in designing broad-spectrum human norovirus protease inhibitors. J Virol 2025; 99:e0175724. [PMID: 39873493 PMCID: PMC11852804 DOI: 10.1128/jvi.01757-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 01/04/2025] [Indexed: 01/30/2025] Open
Abstract
Human norovirus (HuNoV) is a leading cause of gastroenteritis worldwide and is associated with significant morbidity, mortality, and economic impact. There are currently no licensed antiviral drugs for the treatment of HuNoV-associated gastroenteritis. The HuNoV protease plays a critical role in the initiation of virus replication by cleaving the polyprotein. Thus, it is an ideal target for developing antiviral small-molecule inhibitors. While rupintrivir, a potent small-molecule inhibitor of several picornavirus proteases, effectively inhibits GI.1 protease, it is an order of magnitude less effective against GII protease. Other GI.1 protease inhibitors also tend to be less effective against GII proteases. To understand the structural basis for the potency difference, we determined the crystal structures of proteases of GI.1, pandemic GII.4 (Houston and Sydney), and GII.3 in complex with rupintrivir. These structures show that the open substrate pocket in GI protease binds rupintrivir without requiring significant conformational changes, whereas, in GII proteases, the closed pocket flexibly extends, reorienting arginine-112 in the BII-CII loop to accommodate rupintrivir. Structures of R112A protease mutants with rupintrivir, coupled with enzymatic and inhibition studies, suggest R112 is involved in displacing both substrate and ligands from the active site, implying a role in the release of cleaved products during polyprotein processing. Thus, the primary determinant for differential inhibitor potency between the GI and GII proteases is the increased flexibility in the BII-CII loop of the GII proteases caused by the H-G mutation in this loop. Therefore, the inherent flexibility of the BII-CII loop in GII proteases is a critical factor to consider when developing broad-spectrum inhibitors for HuNoV proteases. IMPORTANCE Human noroviruses are a significant cause of sporadic and epidemic gastroenteritis worldwide. There are no vaccines or antiviral drugs currently available to treat infections. Our work elucidates the structural differences between GI.1 and GII proteases in response to inhibitor binding and will inform the future development of broad-spectrum norovirus protease inhibitors.
Collapse
Affiliation(s)
- Son Pham
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, Texas, USA
| | - Boyang Zhao
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, Texas, USA
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Neetu Neetu
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, Texas, USA
| | - Banumathi Sankaran
- Berkeley Center for Structural Biology, Molecular Biophysics, and Integrated Bioimaging, Lawrence Berkeley Laboratory, Berkeley, California, USA
| | - Ketki Patil
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Sasirekha Ramani
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Yongcheng Song
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, Texas, USA
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas, USA
| | - Mary K. Estes
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
- Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Timothy Palzkill
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, Texas, USA
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - B. V. Venkataram Prasad
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, Texas, USA
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
2
|
Hu X, Sun L, Han T, Zhao J, Qi X, Zhang Y, Lu P, Zhao J, Gao Y, Zhang Z, Li B, Du J, Jiao Y. The genetic diversity of genogroup I noroviruses causing acute gastroenteritis outbreaks in Beijing between 2014 and 2023. Heliyon 2024; 10:e39202. [PMID: 39640656 PMCID: PMC11620219 DOI: 10.1016/j.heliyon.2024.e39202] [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] [Scholar Register] [Received: 05/28/2024] [Revised: 10/08/2024] [Accepted: 10/09/2024] [Indexed: 12/07/2024] Open
Abstract
Background In the past decade, we have continuously conducted sporadic monitoring and outbreak detection of norovirus (NoV), which causes human acute gastroenteritis (AGE) in the capital of China. Accumulated data have shown that genogroup I (GI) NoVs not only cause sporadic cases but also cannot be ignored during outbreaks. This study aimed to update the genetic diversity of GI NoVs in the capital of China from 2014 to 2023. Methods Fecal or anal swab samples were collected from AGE outbreaks triggered by GI NoVs in Beijing Chaoyang District from 2014 to 2023. Both the partial coding genes of RNA-dependent RNA polymerase (RdRp) (∼283 bp) and viral protein 1 (VP1) (∼303 bp) were amplified via reverse transcription polymerase chain reaction (RT-PCR) and sequenced, followed by genotyping and phylogenetic analysis. Results A total of 421 fecal or anal swab samples were collected from 59 AGE outbreaks caused by GI NoVs. Genetic diversity was observed, with nine genotypes reported, including recombinant strains of GI.6[P11] and GI.3[P13], as well as multiple subtypes that cocirculated. In addition, we also reported a shift in the dominant genotype, with GI.6 [P11] in 2015-2018, GI.3 [P13] in 2019-2021, and GI.4 [P4] in 2023. Furthermore, alterations in amino acids were indirectly indicated through single nucleotide polymorphisms (SNPs) in certain VP1 areas of strains GI.3[P13] and GI.6[P11]. Epidemiologically, the peak of infection induced by GI NoVs occurs from March to May. Conclusions The sustained circulation and obvious genotype shift of GI NoVs in this region cannot be ignored, and GI.4[P4] NoVs are highly likely to become the main epidemic strain in the following years.
Collapse
Affiliation(s)
- Xiangyu Hu
- Baotou Medical College, Inner Mongolia Autonomous Region, China
- Beijing Chaoyang District Center for Disease Control and Prevention, Beijing, China
| | - Lingli Sun
- Beijing Chaoyang District Center for Disease Control and Prevention, Beijing, China
| | - Taoli Han
- Beijing Chaoyang District Center for Disease Control and Prevention, Beijing, China
| | - Jianhong Zhao
- Beijing Chaoyang District Center for Disease Control and Prevention, Beijing, China
| | - Xiao Qi
- Beijing Chaoyang District Center for Disease Control and Prevention, Beijing, China
| | - Yue Zhang
- Beijing Chaoyang District Center for Disease Control and Prevention, Beijing, China
| | - Pan Lu
- Beijing Chaoyang District Center for Disease Control and Prevention, Beijing, China
| | - Jiaxin Zhao
- Beijing Chaoyang District Center for Disease Control and Prevention, Beijing, China
| | - Yan Gao
- Beijing Chaoyang District Center for Disease Control and Prevention, Beijing, China
| | - Zheng Zhang
- Beijing Chaoyang District Center for Disease Control and Prevention, Beijing, China
| | - Beibei Li
- Beijing Chaoyang District Center for Disease Control and Prevention, Beijing, China
| | - Jialiang Du
- Division of Monoclonal Antibody Products, National Institutes for Food and Drug Control, State Key Laboratory of Drug Regulatory Science, NHC Key Laboratory of Research on Quality and Standardization of Biotech Products, NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, China
| | - Yang Jiao
- Beijing Chaoyang District Center for Disease Control and Prevention, Beijing, China
| |
Collapse
|
3
|
Sun Y, Liang M, Wu M, Su L. Inhibition of Norovirus GII.4 binding to HBGAs by Sargassum fusiforme polysaccharide. Biosci Rep 2024; 44:BSR20240092. [PMID: 39158037 PMCID: PMC11392911 DOI: 10.1042/bsr20240092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 08/12/2024] [Accepted: 08/16/2024] [Indexed: 08/20/2024] Open
Abstract
Norovirus (NoV) is the main pathogen that causes acute gastroenteritis and brings a heavy socio-economic burden worldwide. In this study, five polysaccharide fractions, labeled pSFP-1-5, were isolated and purified from Sargassum fusiforme (S. fusiforme). In vitro experiments demonstrated that pSFP-5 significantly prevented the binding of type A, B and H histo-blood group antigens (HBGAs) to NoV GII.4 virus-like particles (NoV GII.4 VLPs). In addition, in vivo experiments revealed that pSFP-5 was effective in reducing the accumulation of NoV in oysters, indicating that pSFP-5 could reduce the risk of NoV infection from oyster consumption. The results of transmission electron microscopy showed that the appearance of NoV GII.4 VLPs changed after pSFP-5 treatment, indicating that pSFP-5 may achieve antiviral ability by altering the morphological structure of the viral particles so that they could not bind to HBGAs. The results of the present study indicate that pSFP-5 may be an effective anti-NoV substance and can be used as a potential anti-NoV drug component.
Collapse
Affiliation(s)
- Yiqiang Sun
- College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China
- Zhejiang Provincial Key Laboratory for Water Environment and Marine Biological Resources Protection, Wenzhou University, Wenzhou 325035, China
| | - Meina Liang
- College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China
- Zhejiang Provincial Key Laboratory for Water Environment and Marine Biological Resources Protection, Wenzhou University, Wenzhou 325035, China
| | - Mingjiang Wu
- College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China
- Zhejiang Provincial Key Laboratory for Water Environment and Marine Biological Resources Protection, Wenzhou University, Wenzhou 325035, China
| | - Laijin Su
- College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China
- Zhejiang Provincial Key Laboratory for Water Environment and Marine Biological Resources Protection, Wenzhou University, Wenzhou 325035, China
| |
Collapse
|
4
|
Pham S, Zhao B, Neetu N, Sankaran B, Patil K, Ramani S, Song Y, Estes MK, Palzkill T, Prasad BV. CONFORMATIONAL FLEXIBILITY IS A CRITICAL FACTOR IN DESIGNING BROAD-SPECTRUM HUMAN NOROVIRUS PROTEASE INHIBITORS. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.16.613336. [PMID: 39345439 PMCID: PMC11430002 DOI: 10.1101/2024.09.16.613336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Human norovirus (HuNoV) infection is a global health and economic burden. Currently, there are no licensed HuNoV vaccines or antiviral drugs available. The protease encoded by the HuNoV genome plays a critical role in virus replication by cleaving the polyprotein and is, therefore, an excellent target for developing small molecule inhibitors. While rupintrivir, a potent small-molecule inhibitor of several picornavirus proteases, effectively inhibits GI.1 protease, it is an order of magnitude less effective against GII protease. Other GI.1 protease inhibitors also tend to be less effective against GII proteases. To understand the structural basis for the potency difference, we determined the crystal structures of proteases of GI.1, pandemic GII.4 (Houston and Sydney), and GII.3 in complex with rupintrivir. These structures show that the open substrate pocket in GI protease binds rupintrivir without requiring significant conformational changes, whereas, in GII proteases, the closed pocket flexibly extends, reorienting arginine-112 in the BII-CII loop to accommodate rupintrivir. Structures of R112A protease mutants with rupintrivir, coupled with enzymatic and inhibition studies, suggest R112 is involved in displacing both substrate and ligands from the active site, implying a role in the release of cleaved products during polyprotein processing. Thus, the primary determinant for differential inhibitor potency between the GI and GII proteases is the increased flexibility in the BII-CII loop of the GII proteases caused by H-G mutation in this loop. Therefore, the inherent flexibility of the BII-CII loop in GII proteases is a critical factor to consider when developing broad-spectrum inhibitors for HuNoV proteases.
Collapse
Affiliation(s)
- Son Pham
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, TX, USA
| | - Boyang Zhao
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Neetu Neetu
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, 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
| | - Ketki Patil
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Sasirekha Ramani
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Yongcheng Song
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, TX, USA
- Dan L Duncan Comprehensive Cancer Center, 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
| | - Timothy Palzkill
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - B.V. Venkataram Prasad
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
5
|
Tenge V, Ayyar BV, Ettayebi K, Crawford SE, Hayes NM, Shen YT, Neill FH, Atmar RL, Estes MK. Bile acid-sensitive human norovirus strains are susceptible to sphingosine-1-phosphate receptor 2 inhibition. J Virol 2024; 98:e0202023. [PMID: 38884472 PMCID: PMC11265423 DOI: 10.1128/jvi.02020-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 05/15/2024] [Indexed: 06/18/2024] Open
Abstract
Human noroviruses (HuNoVs) are a diverse group of RNA viruses that cause endemic and pandemic acute viral gastroenteritis. Previously, we reported that many HuNoV strains require bile or bile acid (BA) to infect human jejunal intestinal enteroid cultures. BA was not essential for the replication of a pandemic-causing GII.4 HuNoV strain. We found the hydrophobic BA glycochenodeoxycholic acid (GCDCA) promotes the replication of the BA-dependent strain GII.3 in jejunal enteroids. Furthermore, we found that inhibition of the G-protein-coupled BA receptor, sphingosine-1-phosphate receptor 2 (S1PR2), by JTE-013, reduced GII.3 infection dose-dependently and inhibited GII.3 cellular uptake in enteroids. Herein, we sought to determine whether S1PR2 is required for other BA-dependent HuNoV strains, the BA-independent GII.4, and whether S1PR2 is required for BA-dependent HuNoV infection in HIEs from other small intestinal segments. We found a second S1PR2 inhibitor, GLPG2938, reduces GII.3 infection dose-dependently, and an S1PR2 agonist (CYM-5520) enhances GII.3 replication in the absence of GCDCA. GII.3 replication also is abrogated in the presence of JTE-013 and CYM-5520. JTE-013 inhibition of S1PR2 in jejunal HIEs reduces GI.1, GII.3, and GII.17 (BA-dependent) but not GII.4 Sydney (BA-independent) infection, providing additional evidence of strain-specific differences in HuNoV infection. Finally, GII.3 infection of duodenal, jejunal, and ileal lines derived from the same individual is reduced with S1PR2 inhibition, indicating a common mechanism of BA-dependent infection among multiple segments of the small intestine. Our results support a model where BA-dependent HuNoVs exploit BA effects on S1PR2 to infect the entire small intestine.IMPORTANCEHuman noroviruses (HuNoVs) are important viral human pathogens that cause both outbreaks and sporadic gastroenteritis. These viruses are diverse, and many strains are capable of infecting humans. Our previous studies have identified strain-specific requirements for hydrophobic bile acids (BAs) to infect intestinal epithelial cells. Moreover, we identified a BA receptor, sphingosine-1-phosphate receptor 2 (S1PR2), required for infection by a BA-dependent strain. To better understand how various HuNoV strains enter and infect the small intestine and the role of S1PR2 in HuNoV infection, we evaluated infection by additional HuNoV strains using an expanded repertoire of intestinal enteroid cell lines. We found that multiple BA-dependent strains, but not a BA-independent strain, all require S1PR2 for infection. In addition, BA-dependent infection requires S1PR2 in multiple segments of the small intestine. Together, these results indicate that S1PR2 has value as a potential therapeutic target for BA-dependent HuNoV infection.
Collapse
Affiliation(s)
- Victoria Tenge
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - B. Vijayalakshmi Ayyar
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Khalil Ettayebi
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Sue E. Crawford
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Nicole M. Hayes
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Yi-Ting Shen
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Frederick H. Neill
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Robert L. Atmar
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
- Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Mary K. Estes
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
- Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
6
|
Bykov R, Itani T, Starikova P, Skryabina S, Kilyachina A, Koltunov S, Romanov S, Semenov A. Genetic Diversity and Phylogenetic Relationship of Human Norovirus Sequences Derived from Municipalities within the Sverdlovsk Region of Russia. Viruses 2024; 16:1001. [PMID: 39066164 PMCID: PMC11281373 DOI: 10.3390/v16071001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 06/17/2024] [Accepted: 06/19/2024] [Indexed: 07/28/2024] Open
Abstract
Human noroviruses (HuNoVs) are highly contagious pathogens responsible of norovirus-associated acute gastroenteritis (AGE). GII.4 is the prevailing HuNoV genotype worldwide. Currently there are no studies on the molecular monitoring and phylogenetic analysis of HuNoVs in the territory of the Sverdlovsk region; therefore, it is not possible to objectively assess their genetic diversity. The aim of the study is to carry out genotyping and phylogenetic analysis of HuNoVs in the Sverdlovsk region from 2022 to 2023. Fecal samples (n = 510) were collected from children suffering from HuNoV-AGE in municipalities of the Sverdlovsk region and the capsid genotype was determined by amplifying the ORF1/ORF2 junction. Of the 196 HuNoVs typed, which represent 38% of the studied samples, the largest share of HuNoV genotypes belong to the GII genogroup-86%, followed by the GI genogroup-14%. Noroviruses GII.4 and GII.17 were the co-dominant capsid genotypes (33.2% each). Phylogenetic analysis demonstrates that the identified sequences on the territory of the Sverdlovsk region have the smallest genetic distance, which gives grounds for their unification into a common cluster. Routine monitoring and phylogenetic analysis of circulating norovirus pathogens spectrum will enable timely tracking of HuNoVs genetic diversity and evolutionary events. This will lead to the development of more effective anti-epidemic measures, ultimately reducing the burden of infectious diseases.
Collapse
Affiliation(s)
- Roman Bykov
- Federal Budgetary Institution of Science, «Federal Scientific Research Institute of Viral Infections «Virome»», Federal Service for Surveillance on Consumer Rights Protection and Human Wellbeing, Ekaterinburg 620030, Russia; (T.I.); (P.S.); (A.S.)
| | - Tarek Itani
- Federal Budgetary Institution of Science, «Federal Scientific Research Institute of Viral Infections «Virome»», Federal Service for Surveillance on Consumer Rights Protection and Human Wellbeing, Ekaterinburg 620030, Russia; (T.I.); (P.S.); (A.S.)
| | - Polina Starikova
- Federal Budgetary Institution of Science, «Federal Scientific Research Institute of Viral Infections «Virome»», Federal Service for Surveillance on Consumer Rights Protection and Human Wellbeing, Ekaterinburg 620030, Russia; (T.I.); (P.S.); (A.S.)
| | - Svetlana Skryabina
- Federal Service for Surveillance on Consumer Rights Protection and Human Wellbeing in the Sverdlovsk Region, Ekaterinburg 620078, Russia;
| | - Anastasia Kilyachina
- Federal Budgetary Healthcare Institution «Center of Hygiene and Epidemiology in the Sverdlovsk Region», Ekaterinburg 620078, Russia; (A.K.); (S.K.); (S.R.)
| | - Stanislav Koltunov
- Federal Budgetary Healthcare Institution «Center of Hygiene and Epidemiology in the Sverdlovsk Region», Ekaterinburg 620078, Russia; (A.K.); (S.K.); (S.R.)
| | - Sergey Romanov
- Federal Budgetary Healthcare Institution «Center of Hygiene and Epidemiology in the Sverdlovsk Region», Ekaterinburg 620078, Russia; (A.K.); (S.K.); (S.R.)
| | - Aleksandr Semenov
- Federal Budgetary Institution of Science, «Federal Scientific Research Institute of Viral Infections «Virome»», Federal Service for Surveillance on Consumer Rights Protection and Human Wellbeing, Ekaterinburg 620030, Russia; (T.I.); (P.S.); (A.S.)
- Department of Medical Microbiology and Clinical Laboratory Diagnostics, Ural State Medical University, Ekaterinburg 620109, Russia
- Institute of Natural Sciences and Mathematics, Ural Federal University Named after the First President of Russia B.N. Yeltsin, Ekaterinburg 620075, Russia
| |
Collapse
|
7
|
Lo M, Doan YH, Mitra S, Saha R, Miyoshi SI, Kitahara K, Dutta S, Oka T, Chawla-Sarkar M. Comprehensive full genome analysis of norovirus strains from eastern India, 2017-2021. Gut Pathog 2024; 16:3. [PMID: 38238807 PMCID: PMC10797879 DOI: 10.1186/s13099-023-00594-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 12/20/2023] [Indexed: 01/22/2024] Open
Abstract
BACKGROUND Worldwide, noroviruses are the leading cause of acute gastroenteritis (AGE) in people of all age groups. In India, norovirus rates between 1.4 to 44.4% have been reported. Only a very few complete norovirus genome sequences from India have been reported. OBJECTIVE To perform full genome sequencing of noroviruses circulating in India during 2017-2021, identify circulating genotypes, assess evolution including detection of recombination events. METHODOLOGY Forty-five archived norovirus-positive samples collected between October 2017 to July 2021 from patients with AGE from two hospitals in Kolkata, India were processed for full genome sequencing. Phylogenetic analysis, recombination breakpoint analysis and comprehensive mutation analysis were also performed. RESULTS Full genome analysis of norovirus sequences revealed that strains belonging to genogroup (G)I were genotyped as GI.3[P13]. Among the different norovirus capsid-polymerase combinations, GII.3[P16], GII.4 Sydney[P16], GII.4 Sydney[P31], GII.13[P16], GII.16[P16] and GII.17 were identified. Phylogenetic analysis confirmed phylogenetic relatedness with previously reported norovirus strains and all viruses were analyzed by Simplot. GII[P16] viruses with multiple residue mutations within the non-structural region were detected among circulating GII.4 and GII.3 strains. Comprehensive mutation analysis and selection pressure analysis of GII[P16] viruses showed positive as well as negative selection sites. A GII.17 strain (NICED-BCH-11889) had an untypeable polymerase type, closely related to GII[P38]. CONCLUSION This study highlights the circulation of diverse norovirus strains in eastern India. These findings are important for understanding norovirus epidemiology in India and may have implications for future vaccine development.
Collapse
Affiliation(s)
- Mahadeb Lo
- Division of Virology, ICMR-National Institute of Cholera and Enteric Diseases, P-33, C.I.T. Rd, Scheme-XM, Beliaghata, Kolkata, 700010, West Bengal, India
| | - Yen Hai Doan
- Center for Emergency Preparedness and Response, National Institute of Infectious Diseases, Gakuen 4-7-1, Musashi-Murayama, Tokyo, Japan
| | - Suvrotoa Mitra
- Division of Virology, ICMR-National Institute of Cholera and Enteric Diseases, P-33, C.I.T. Rd, Scheme-XM, Beliaghata, Kolkata, 700010, West Bengal, India
| | - Ritubrita Saha
- Division of Virology, ICMR-National Institute of Cholera and Enteric Diseases, P-33, C.I.T. Rd, Scheme-XM, Beliaghata, Kolkata, 700010, West Bengal, India
| | - Shin-Ichi Miyoshi
- Collaborative Research Center of Okayama University for Infectious Diseases in India, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata, India
- Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Kei Kitahara
- Collaborative Research Center of Okayama University for Infectious Diseases in India, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata, India
| | - Shanta Dutta
- Regional Virus Research and Diagnostic Laboratory, ICMR-National Institute of Cholera and Enteric Diseases, Beliaghata, Kolkata, West Bengal, India
| | - Tomoichiro Oka
- Department of Virology II, National Institute of Infectious Diseases, Gakuen 4-7-1, Musashi-Murayama, Tokyo, Japan.
| | - Mamta Chawla-Sarkar
- Division of Virology, ICMR-National Institute of Cholera and Enteric Diseases, P-33, C.I.T. Rd, Scheme-XM, Beliaghata, Kolkata, 700010, West Bengal, India.
| |
Collapse
|
8
|
Tenge V, Vijayalakshmi Ayyar B, Ettayebi K, Crawford SE, Shen YT, Neill FH, Atmar RL, Estes MK. Bile acid-sensitive human norovirus strains are susceptible to sphingosine-1-phosphate receptor 2 inhibition. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.02.573926. [PMID: 38260626 PMCID: PMC10802320 DOI: 10.1101/2024.01.02.573926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Human noroviruses (HuNoVs) are a diverse group of RNA viruses that cause both endemic and pandemic acute viral gastroenteritis. Previously we reported that many strains of HuNoV require bile or bile acid (BA) to infect human jejunal intestinal enteroid cultures. Of note, BA was not essential for replication of a pandemic-causing GII.4 HuNoV strain. Using the BA-requiring strain GII.3, we found that the hydrophobic BA GCDCA induces multiple cellular responses that promote replication in jejunal enteroids. Further, we found that chemical inhibition of the G-protein coupled receptor, sphingosine-1- phosphate receptor 2 (S1PR2), by JTE-013 reduced both GII.3 infection in a dose- dependent manner and cellular uptake in enteroids. Herein, we sought to determine if S1PR2 is required by other BA-dependent HuNoV strains and BA-independent GII.4, and if S1PR2 is required for BA-dependent HuNoV infection in other segments of the small intestine. We found JTE-013 inhibition of S1PR2 in jejunal HIEs reduces GI.1, GII.3, and GII.17 (BA-dependent) but not the GII.4 Sydney variant (BA-independent) infection, providing additional evidence of strain-specific differences in HuNoV infection. GII.3 infection of duodenal, jejunal and ileal lines derived from the same individual was also reduced with S1PR2 inhibition, indicating a common mechanism of BA-dependent infection among multiple segments of the small intestine. Our results support a model where BA-dependent HuNoV exploit the activation of S1PR2 by BA to infect the entire small intestine. Importance Human noroviruses (HuNoVs) are important viral human pathogens that cause both outbreaks and sporadic gastroenteritis. These viruses are diverse, and many strains are capable of infecting humans. Our previous studies have identified strain-specific requirements for hydrophobic bile acids (BAs) to infect intestinal epithelial cells. Moreover, we identified a BA receptor, sphingosine-1-phosphate receptor 2 (S1PR2), required for infection by a BA-dependent strain. To better understand how various HuNoV strains enter and infect the small intestine and the role of S1PR2 in HuNoV infection, we evaluated infection by additional HuNoV strains using an expanded repertoire of intestinal enteroid cell lines. We found that multiple BA-dependent strains, but not a BA- independent strain, all required S1PR2 for infection. Additionally, BA-dependent infection required S1PR2 in multiple segments of the small intestine. Together these results indicate S1PR2 has value as a potential therapeutic target for BA-dependent HuNoV infection.
Collapse
|
9
|
Yu JR, Xie DJ, Li JH, Koroma MM, Wang L, Wang Y, Jing DN, Xu JY, Yu JX, Du HS, Zhou FY, Liang ZY, Zhang XF, Dai YC. Serological surveillance of GI norovirus reveals persistence of blockade antibody in a Jidong community-based prospective cohort, 2014-2018. Front Cell Infect Microbiol 2023; 13:1258550. [PMID: 38188632 PMCID: PMC10766831 DOI: 10.3389/fcimb.2023.1258550] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 11/29/2023] [Indexed: 01/09/2024] Open
Abstract
Introduction Herd immunity against norovirus (NoV) is poorly understood in terms of its serological properties and vaccine designs. The precise neutralizing serological features of genotype I (GI) NoV have not been studied. Methods To expand insights on vaccine design and herd immunity of NoVs, seroprevalence and seroincidence of NoV genotypes GI.2, GI.3, and GI.9 were determined using blockade antibodies based on a 5-year longitudinal serosurveillance among 449 residents in Jidong community. Results Correlation between human histo-blood group antigens (HBGAs) and GI NoV, and dynamic and persistency of antibodies were also analyzed. Seroprevalence of GI.2, GI.3, and GI.9 NoV were 15.1%-18.0%, 35.0%-38.8%, and 17.6%-22.0%; seroincidences were 10.0, 21.0, and 11.0 per 100.0 person-year from 2014 to 2018, respectively. Blockade antibodies positive to GI.2 and GI.3 NoV were significantly associated with HBGA phenotypes, including blood types A, B (excluding GI.3), and O+; Lewis phenotypes Leb+/Ley+ and Lea+b+/Lex+y+; and secretors. The overall decay rate of anti-GI.2 antibody was -5.9%/year (95% CI: -7.1% to -4.8%/year), which was significantly faster than that of GI.3 [-3.6%/year (95% CI: -4.6% to -2.6%/year)] and GI.9 strains [-4.0%/year (95% CI: -4.7% to -3.3%/year)]. The duration of anti-GI.2, GI.3, and GI.9 NoV antibodies estimated by generalized linear model (GLM) was approximately 2.3, 4.2, and 4.8 years, respectively. Discussion In conclusion, enhanced community surveillance of GI NoV is needed, and even one-shot vaccine may provide coast-efficient health benefits against GI NoV infection.
Collapse
Affiliation(s)
- Jing-Rong Yu
- Department of Epidemiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
- Department of Public Health, Shenzhen Qianhai Shekou Free Trade Zone Hospital, Shenzhen, China
| | - Dong-Jie Xie
- The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, China
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Jia-Heng Li
- Department of Epidemiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Mark Momoh Koroma
- Department of Epidemiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Lu Wang
- Department of Epidemiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Yu Wang
- Department of Epidemiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Duo-Na Jing
- Department of Epidemiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Jia-Yi Xu
- Department of Epidemiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Jun-Xuan Yu
- Department of Epidemiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Hui-Sha Du
- Department of Epidemiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Fei-Yuan Zhou
- Department of Epidemiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Zhi-Yan Liang
- Department of Epidemiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Xu-Fu Zhang
- The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, China
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Ying-Chun Dai
- Department of Epidemiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| |
Collapse
|
10
|
Li C, Song S, Huang X, Liu X, Lv H, Shen Y, Wei X, Zhang W, Xu Y. Molecular epidemiology and genetic diversity of norovirus among hospitalized patients with acute gastroenteritis in Shandong, China, 2016-2018. J Med Virol 2023; 95:e29339. [PMID: 38130177 DOI: 10.1002/jmv.29339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 12/08/2023] [Accepted: 12/12/2023] [Indexed: 12/23/2023]
Abstract
Norovirus (NoV) infection is a leading cause of acute gastroenteritis (AGE) for people of all ages. Here, we reported the molecular epidemiology and genetic diversity of NoVs among hospitalized patients with AGE between 2016 and 2018 in Shandong Province, China. Two thousand sixty-nine AGE patients from sentinel hospitals were enrolled. The stool samples were collected and tested for NoVs by real-time RT-PCR. The RNA-dependent RNA polymerase (RdRp) and capsid gene of 163 strains were amplified and sequenced for genotyping. Phylogenetic analyses and genomic characterization were conducted with the VP1 and RdRp region of the full genome sequences. Four hundred seventy two (21.76%) samples were NoV-positive. The positive rate in 2016 was higher than those of 2017 and 2018. We observed diverse NoV genotypes. GII.2[P16] emerged in January 2017 and became the dominant genotype between May and June 2017. Phylogenetic analyses showed that our GII.2[P16] genomes clustered in the SC1 in VP1 region, while they belonged to the Emerging GⅡ.P16 (2015-2017) clade in RdRp region. Our GⅡ.4 strains displayed two amino acid mutations, positions R297H and D372N, in epitope A of the VP1 region. Our study highlighted that NoV is an important pathogen of viral AGE in Shandong and, therefore, it is necessary to strengthen its surveillance.
Collapse
Affiliation(s)
- Chao Li
- Department of Microbiology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Shaoxia Song
- Shandong Center for Disease Control and Prevention, Jinan, Shandong, China
- Shandong Provincial Key Laboratory for Infectious Disease Prevention and Control, Preventive healthcare Research Institute of Shandong University, Jinan, China
| | - Xianglin Huang
- Department of Microbiology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Xiaolin Liu
- Shandong Center for Disease Control and Prevention, Jinan, Shandong, China
- Shandong Provincial Key Laboratory for Infectious Disease Prevention and Control, Preventive healthcare Research Institute of Shandong University, Jinan, China
| | - Hui Lv
- Shandong Center for Disease Control and Prevention, Jinan, Shandong, China
- Shandong Provincial Key Laboratory for Infectious Disease Prevention and Control, Preventive healthcare Research Institute of Shandong University, Jinan, China
| | - Yuanyuan Shen
- Department of Microbiology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Xuemin Wei
- Department of Microbiology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Wenqiang Zhang
- Shandong Center for Disease Control and Prevention, Jinan, Shandong, China
- Shandong Provincial Key Laboratory for Infectious Disease Prevention and Control, Preventive healthcare Research Institute of Shandong University, Jinan, China
| | - Yifei Xu
- Department of Microbiology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Suzhou Research Institute of Shandong University, Suzhou, Jiangsu, China
| |
Collapse
|
11
|
Bai GH, Tsai MC, Lin SC, Hsu YH, Chen SY. Unraveling the interplay between norovirus infection, gut microbiota, and novel antiviral approaches: a comprehensive review. Front Microbiol 2023; 14:1212582. [PMID: 37485533 PMCID: PMC10359435 DOI: 10.3389/fmicb.2023.1212582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 06/15/2023] [Indexed: 07/25/2023] Open
Abstract
Norovirus infection is a leading cause of acute gastroenteritis worldwide and can also cause harmful chronic infections in individuals with weakened immune systems. The role of the gut microbiota in the interactions between the host and noroviruses has been extensively studied. While most past studies were conducted in vitro or focused on murine noroviruses, recent research has expanded to human noroviruses using in vivo or ex vivo human intestinal enteroids culture studies. The gut microbiota has been observed to have both promoting and inhibiting effects on human noroviruses. Understanding the interaction between noroviruses and the gut microbiota or probiotics is crucial for studying the pathogenesis of norovirus infection and its potential implications, including probiotics and vaccines for infection control. Recently, several clinical trials of probiotics and norovirus vaccines have also been published. Therefore, in this review, we discuss the current understanding and recent updates on the interactions between noroviruses and gut microbiota, including the impact of norovirus on the microbiota profile, pro-viral and antiviral effects of microbiota on norovirus infection, the use of probiotics for treating norovirus infections, and human norovirus vaccine development.
Collapse
Affiliation(s)
- Geng-Hao Bai
- Department of Internal Medicine, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Meng-Chen Tsai
- Department of General Medicine, Taipei Medical University Hospital, Taipei, Taiwan
| | - Sheng-Chieh Lin
- Department of Pediatrics, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Pediatrics, Division of Allergy, Asthma and Immunology, Shuang Ho Hospital, New Taipei, Taiwan
| | - Yi-Hsiang Hsu
- Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, United States
- Broad Institute of MIT and Harvard, Cambridge, MA, United States
| | - Shih-Yen Chen
- Department of Pediatrics, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Pediatrics, Division of Pediatric Gastroenterology and Hepatology, Shuang Ho Hospital, New Taipei, Taiwan
- TMU Research Center for Digestive Medicine, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
12
|
Li TT, Xu Q, Liu MC, Wang T, Che TL, Teng AY, Lv CL, Wang GL, Hong F, Liu W, Fang LQ. Prevalence and Etiological Characteristics of Norovirus Infection in China: A Systematic Review and Meta-Analysis. Viruses 2023; 15:1336. [PMID: 37376635 DOI: 10.3390/v15061336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 06/04/2023] [Accepted: 06/04/2023] [Indexed: 06/29/2023] Open
Abstract
Norovirus is a common cause of sporadic cases and outbreaks of gastroenteritis worldwide, although its prevalence and the dominant genotypes responsible for gastroenteritis outbreaks remain obscure. A systematic review was conducted on norovirus infection in China between January 2009 and March 2021. A meta-analysis and beta-binomial regression model were used to explore the epidemiological and clinical characteristics of norovirus infection and the potential factors contributing to the attack rate of the norovirus outbreaks, respectively. A total of 1132 articles with 155,865 confirmed cases were included, with a pooled positive test rate of 11.54% among 991,786 patients with acute diarrhea and a pooled attack rate of 6.73% in 500 norovirus outbreaks. GII.4 was the predominant genotype in both the etiological surveillance and outbreaks, followed by GII.3 in the etiological surveillance, and GII.17 in the outbreaks, with the proportion of recombinant genotypes increasing in recent years. A higher attack rate in the norovirus outbreaks was associated with age group (older adults), settings (nurseries, primary schools, etc.) and region (North China). The nation-wide pooled positive rate in the etiological surveillance of norovirus is lower than elsewhere in the global population, while the dominant genotypes are similar in both the etiological surveillance and the outbreak investigations. This study contributes to the understanding of norovirus infection with different genotypes in China. The prevention and control of norovirus outbreaks during the cold season should be intensified, with special attention paid to and enhanced surveillance performed in nurseries, schools and nursing homes from November to March.
Collapse
Affiliation(s)
- Ting-Ting Li
- School of Public Health, Guizhou Medical University, Guiyang 550025, China
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Qiang Xu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Mei-Chen Liu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
- School of Public Health, Anhui Medical University, Hefei 230032, China
| | - Tao Wang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Tian-Le Che
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Ai-Ying Teng
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Chen-Long Lv
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Guo-Lin Wang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Feng Hong
- School of Public Health, Guizhou Medical University, Guiyang 550025, China
| | - Wei Liu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
- School of Public Health, Anhui Medical University, Hefei 230032, China
| | - Li-Qun Fang
- School of Public Health, Guizhou Medical University, Guiyang 550025, China
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| |
Collapse
|
13
|
Yu Z, Shao Q, Xu Z, Chen C, Li M, Jiang Y, Cheng D. Immunogenicity and Blocking Efficacy of Norovirus GII.4 Recombinant P Protein Vaccine. Vaccines (Basel) 2023; 11:1053. [PMID: 37376442 DOI: 10.3390/vaccines11061053] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 05/24/2023] [Accepted: 05/30/2023] [Indexed: 06/29/2023] Open
Abstract
Noroviruses (NoVs) are the main cause of acute gastroenteritis in all ages worldwide. The aim of this study was to produce the recombinant P protein of norovirus and to demonstrate its blocking effect. In this study, the engineered strains were induced to express the P protein of NoVs GII.4, which was identified using SDS-PAGE and ELISA as having the capacity to bind to histo-blood group antigens (HBGAs). Rabbits were immunized to obtain neutralizing antibodies. ELISA and ISC-RT-qPCR were used to determine the blocking efficacy of the neutralizing antibody to human norovirus (HuNoV) and murine norovirus (MNV). The recombinant P protein (35 KD) was obtained, and the neutralizing antibody was successfully prepared. The neutralizing antibody could block the binding of the P protein and HuNoV to HBGAs. Neutralizing antibodies can also block MNV invasion into host cells RAW264.7. The recombinant P protein expressed in E. coli can induce antibodies to block HuNoV and MNV. The recombinant P protein of NoVs GII.4 has the value of vaccine development.
Collapse
Affiliation(s)
- Zhendi Yu
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Qingyi Shao
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Zhangkai Xu
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Chenghao Chen
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Mingfan Li
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Yi Jiang
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Dongqing Cheng
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou 310053, China
| |
Collapse
|
14
|
Hung CH, Yen JB, Chang PJ, Chen LW, Huang TY, Tsai WJ, Tsai YC. Characterization of Human Norovirus Nonstructural Protein NS1.2 Involved in the Induction of the Filamentous Endoplasmic Reticulum, Enlarged Lipid Droplets, LC3 Recruitment, and Interaction with NTPase and NS4. Viruses 2023; 15:v15030812. [PMID: 36992520 PMCID: PMC10053803 DOI: 10.3390/v15030812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 03/09/2023] [Accepted: 03/20/2023] [Indexed: 03/31/2023] Open
Abstract
Human noroviruses (HuNVs) are the leading cause of gastroenteritis worldwide. NS1.2 is critical for HuNV pathogenesis, but the function is still unclear. The GII NS1.2 of HuNVs, unlike GI NS1.2, was localized to the endoplasmic reticulum (ER) and lipid droplets (LDs) and is accompanied by a distorted-filamentous ER morphology and aggregated-enlarged LDs. LC3 was recruited to the NS1.2-localized membrane through an autophagy-independent pathway. NS1.2, expressed from a cDNA clone of GII.4 norovirus, formed complexes with NTPase and NS4, which exhibited aggregated vesicle-like structures that were also colocalized with LC3 and LDs. NS1.2 is structurally divided into three domains from the N terminus: an inherently disordered region (IDR), a region that contains a putative hydrolase with the H-box/NC catalytic center (H-box/NC), and a C-terminal 251-330 a.a. region containing membrane-targeting domain. All three functional domains of NS1.2 were required for the induction of the filamentous ER. The IDR was essential for LC3 recruitment by NS1.2. Both the H-Box/NC and membrane-targeting domains are required for the induction of aggregated-enlarged LDs, NS1.2 self-assembly, and interaction with NTPase. The membrane-targeting domain was sufficient to interact with NS4. The study characterized the NS1.2 domain required for membrane targeting and protein-protein interactions, which are crucial for forming a viral replication complex.
Collapse
Affiliation(s)
- Chien-Hui Hung
- Graduate Institute of Clinical Medical Sciences, Chang Gung University, Taoyuan 33302, Taiwan
- Department of Internal Medicine, Division of Infectious Diseases, Chang Gung Memorial Hospital, Chiayi 61363, Taiwan
| | - Ju-Bei Yen
- Department of Pediatrics, Chang Gung Memorial Hospital, Chiayi 61363, Taiwan
| | - Pey-Jium Chang
- Graduate Institute of Clinical Medical Sciences, Chang Gung University, Taoyuan 33302, Taiwan
| | - Lee-Wen Chen
- Department of Respiratory Care, Chung Gung University of Science and Technology, Chiayi 61363, Taiwan
| | - Tsung-Yu Huang
- Department of Internal Medicine, Division of Infectious Diseases, Chang Gung Memorial Hospital, Chiayi 61363, Taiwan
| | - Wan-Ju Tsai
- Graduate Institute of Clinical Medical Sciences, Chang Gung University, Taoyuan 33302, Taiwan
| | - Yu-Chin Tsai
- Graduate Institute of Clinical Medical Sciences, Chang Gung University, Taoyuan 33302, Taiwan
| |
Collapse
|
15
|
Functional and structural characterization of Norovirus GII.6 in recognizing histo-blood group antigens. Virol Sin 2023; 38:56-65. [PMID: 36216242 PMCID: PMC10006186 DOI: 10.1016/j.virs.2022.09.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 07/04/2022] [Indexed: 11/11/2022] Open
Abstract
Noroviruses (NoVs) are the primary cause of acute gastroenteritis worldwide. Histo-blood group antigens (HBGAs) are receptors or attachment factors that affect the prevalence and host susceptibility of NoVs. GII.6 NoV is one of the predominant genotypes in humans, which recognizes the type ABO secretor of HBGAs. However, the structural basis of GII.6 NoV's interaction with HBGAs receptors remains elusive. In this study, we investigated the binding features of the GII.6 strain to HBGAs using saliva- and glycan-ELISA assays and characterized the molecular basis of the GII.6 virus that recognizes H disaccharide. We showed that the GII.6 P domain recognized some A and O secretor's saliva samples, most B secretor's saliva samples, and H disaccharide antigen, but did not bind non-secretors' saliva. Further, we determined the crystal structures of GII.6 and its complex with H disaccharides at 1.7 Å, revealing that the P domain of GII.6 shares the conventional binding interface and mode of GII HBGAs. Single residue mutations at the GII.6-H binding sites could inhibit the binding of GII.6 to HBGAs, demonstrating that the interaction residues were crucial in maintaining NoV-glycan integrity. Finally, structural and sequence analyses showed that the major residues of the GII.6-H interaction were conserved among NoVs in the GII genogroup. Taken together, our study characterized the functional and structural features of GII.6 that allow it to interact with HBGAs, and shed light on NoV evolution, epidemiology, and anti-viral drug development.
Collapse
|
16
|
Tohma K, Ushijima H. [Molecular epidemiology and evolution of human noroviruses]. Uirusu 2023; 73:17-32. [PMID: 39343517 DOI: 10.2222/jsv.73.17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Noroviruses are the most common viral cause of acute gastroenteritis after the introduction of rotavirus vaccines. Norovirus infection can cause severe symptoms in vulnerable populations including young children and the elderly. Thus, it is still a leading cause of death from diarrhea in children in developing countries. Recent advancement of genomics platforms facilitated understanding of the epidemiology of norovirus, while the whole picture of norovirus diversity is still undetermined. Currently, there are no approved vaccines for norovirus, but state-of-the-art norovirus cultivation systems could elucidate the antigenic diversity of this fast-evolving virus. In this review, we will summarize the historical and latest findings of norovirus epidemiology, diversity, and evolution.
Collapse
Affiliation(s)
- Kentaro Tohma
- Division of Viral Products, US Food and Drug Administration, Maryland, Unites States
| | - Hiroshi Ushijima
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo, Japan
| |
Collapse
|
17
|
Liao Y, Xue L, Gao J, Zuo Y, Liang Y, Jiang Y, Cai W, Yang J, Zhang J, Ding Y, Chen M, Wu A, Kou X, Wu Q. Rapid screening for antigenic characterization of GII.17 norovirus strains with variations in capsid gene. Gut Pathog 2022; 14:31. [PMID: 35879724 PMCID: PMC9309444 DOI: 10.1186/s13099-022-00504-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 06/11/2022] [Indexed: 11/13/2022] Open
Abstract
The emergence of the novel GII.17 Kawasaki 2014 norovirus variant raising the interest of the public, has replaced GII.4 as the predominant cause of noroviruses outbreaks in East Asia during 2014–2015. Antigenic variation of the capsid protein is considered as one of the key mechanisms of norovirus evolution. In this study, we screened a panel of GII.17 mutants. First, we produced norovirus P proteins using cell-free protein synthesis (CFPS) system, comparing the results to pure proteins expressed in a cell-based system. Next, we determined the binding capability of specific monoclonal antibody (mAb) 2D11 using a unique set of wild-type GII.17 strains. Results of the EIA involving a panel of mutant cell-free proteins indicated that Q298 was the key residue within loop 1. These data highlighted the essential residues in the linear antibody binding characteristics of novel GII.17. Furthermore, it supported the CFPS as a promising tool for rapidly screening mutants via the scalable expression of norovirus P proteins.
Collapse
|
18
|
Yu F, Jiang B, Guo X, Hou L, Tian Y, Zhang J, Li Q, Jia L, Yang P, Wang Q, Pang X, Gao Z. Norovirus outbreaks in China, 2000-2018: A systematic review. Rev Med Virol 2022; 32:e2382. [PMID: 35946340 DOI: 10.1002/rmv.2382] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 07/14/2022] [Accepted: 07/21/2022] [Indexed: 11/09/2022]
Abstract
To understand epidemiological characteristics of norovirus outbreaks in China from 2000 to 2018 the literature on norovirus outbreaks was identified by searching WANFANG, CNKI, PubMed, and Web of Science databases before 31 December 2018. Statistical analyses were performed using Statistical Product Service Solutions software. RStudio1.4.1717 and ArcGIS trial version were used for plotting bar graphs and maps. A total of 419 norovirus outbreaks were reported in the 394 included articles, which occurred between June 2000 and October 2018, showing an overall increasing trend. The majority of outbreaks occurred in schools (52.28%, 218/417) and kindergartens (55/417, 13.19%). Person-to-person transmission (41.64%, 137/329) was most common, followed by food-borne transmission (75/329, 22.80%) and water-borne transmission (72/329, 21.88%). GII was the most predominant norovirus genogroup, with GII.4, GII.17 and GII.2 being the dominant genotypes in 2007-2013, 2014-2015, 2016-2017, respectively. Increased outbreaks were associated with the prevalence of new variants. Most norovirus outbreaks were reported in the southeast of the country. The number of norovirus outbreaks was positively associated with the per capita gross domestic product and the year-end resident population. Norovirus outbreaks have become an important public health problem in China. It is necessary to establish surveillance in hospitals and nursing homes. Genotyping of noroviruses is important for monitoring the circulating strains and improving the vaccine design, so it should be carried out in more regions.
Collapse
Affiliation(s)
- Fan Yu
- China Medical University School of Public Health, Shenyang, China.,Beijing Center for Disease Prevention and Control, Beijing, China
| | - Bo Jiang
- Capital Medical University School of Public Health, Beijing, China
| | - Xinhui Guo
- Fangshan District Center for Disease Prevention and Control, Beijing, China
| | - Liyu Hou
- Capital Medical University School of Public Health, Beijing, China
| | - Yi Tian
- Beijing Center for Disease Prevention and Control, Beijing, China
| | - Jiaying Zhang
- Capital Medical University School of Public Health, Beijing, China
| | - Qianqian Li
- Shanghai Institute of Technology, Shanghai, China
| | - Lei Jia
- Beijing Center for Disease Prevention and Control, Beijing, China
| | - Peng Yang
- Beijing Center for Disease Prevention and Control, Beijing, China
| | - Quanyi Wang
- Beijing Center for Disease Prevention and Control, Beijing, China
| | - Xinghuo Pang
- Beijing Center for Disease Prevention and Control, Beijing, China
| | - Zhiyong Gao
- China Medical University School of Public Health, Shenyang, China.,Beijing Center for Disease Prevention and Control, Beijing, China
| |
Collapse
|
19
|
Estienney M, Tarris G, Abou-Hamad N, Rouleau A, Boireau W, Chassagnon R, Ayouni S, Daval-Frerot P, Martin L, Bouyer F, Le Pendu J, de Rougemont A, Belliot G. Epidemiological Impact of GII.17 Human Noroviruses Associated With Attachment to Enterocytes. Front Microbiol 2022; 13:858245. [PMID: 35572680 PMCID: PMC9094630 DOI: 10.3389/fmicb.2022.858245] [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] [Scholar Register] [Received: 01/19/2022] [Accepted: 03/23/2022] [Indexed: 01/19/2023] Open
Abstract
For the last 30 years, molecular surveys have shown that human norovirus (HuNoV), predominantly the GII.4 genotype, is one of the main causative agents of gastroenteritis. However, epidemiological surveys have revealed the worldwide emergence of GII.17 HuNoVs. Genetic analysis confirmed that GII.17 strains are distributed into three variants (i.e., Kawasaki 308, Kawasaki 323, and CS-E1). Here, virus-like particles (VLPs) were baculovirus-expressed from these variants to study putative interactions with HBGA. Qualitative analysis of the HBGA binding profile of each variant showed that the most recent and predominant GII.17 variant, Kawasaki 308, possesses a larger binding spectrum. The retrospective study of GII.17 strains documented before the emergence of the dominant Kawasaki 308 variant showed that the emergence of a new GII.17 variant could be related to an increased binding capacity toward HBGA. The use of duodenal histological sections confirmed that recognition of enterocytes involved HBGA for the three GII.17 variants. Finally, we observed that the relative affinity of recent GII.17 VLPs for HBGA remains lower than that of the GII.4-2012 variant. These observations suggest a model whereby a combination of virological factors, such as polymerase fidelity and increased affinity for HBGA, and immunological factors was responsible for the incomplete and non-persistent replacement of GII.4 by new GII.17 variants.
Collapse
Affiliation(s)
- Marie Estienney
- National Reference Centre for Gastroenteritis Viruses, Laboratory of Virology, University Hospital of Dijon, Dijon, France.,UMR PAM A 02.102 Procédés Alimentaires et Microbiologiques, Université de Bourgogne, Franche-Comté/AgroSup Dijon, Dijon, France
| | - Georges Tarris
- Department of Pathology, University Hospital of Dijon, Dijon, France.,Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Dijon, France
| | - Nicole Abou-Hamad
- National Reference Centre for Gastroenteritis Viruses, Laboratory of Virology, University Hospital of Dijon, Dijon, France.,UMR PAM A 02.102 Procédés Alimentaires et Microbiologiques, Université de Bourgogne, Franche-Comté/AgroSup Dijon, Dijon, France.,Laboratoire Interdisciplinaire Carnot de Bourgogne, UMR CNRS 6303, Université Bourgogne Franche-Comté, Dijon, France
| | - Alain Rouleau
- FEMTO-ST Institute, CNRS UMR-6174, Université de Bourgogne Franche-Comté, Besançon, France
| | - Wilfrid Boireau
- FEMTO-ST Institute, CNRS UMR-6174, Université de Bourgogne Franche-Comté, Besançon, France
| | - Rémi Chassagnon
- Laboratoire Interdisciplinaire Carnot de Bourgogne, UMR CNRS 6303, Université Bourgogne Franche-Comté, Dijon, France
| | - Siwar Ayouni
- National Reference Centre for Gastroenteritis Viruses, Laboratory of Virology, University Hospital of Dijon, Dijon, France
| | - Philippe Daval-Frerot
- National Reference Centre for Gastroenteritis Viruses, Laboratory of Virology, University Hospital of Dijon, Dijon, France
| | - Laurent Martin
- Department of Pathology, University Hospital of Dijon, Dijon, France.,Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Dijon, France
| | - Frédéric Bouyer
- Laboratoire Interdisciplinaire Carnot de Bourgogne, UMR CNRS 6303, Université Bourgogne Franche-Comté, Dijon, France
| | | | - Alexis de Rougemont
- National Reference Centre for Gastroenteritis Viruses, Laboratory of Virology, University Hospital of Dijon, Dijon, France.,UMR PAM A 02.102 Procédés Alimentaires et Microbiologiques, Université de Bourgogne, Franche-Comté/AgroSup Dijon, Dijon, France
| | - Gael Belliot
- National Reference Centre for Gastroenteritis Viruses, Laboratory of Virology, University Hospital of Dijon, Dijon, France.,UMR PAM A 02.102 Procédés Alimentaires et Microbiologiques, Université de Bourgogne, Franche-Comté/AgroSup Dijon, Dijon, France
| |
Collapse
|
20
|
Kendra JA, Tohma K, Parra GI. Global and regional circulation trends of norovirus genotypes and recombinants, 1995-2019: A comprehensive review of sequences from public databases. Rev Med Virol 2022; 32:e2354. [PMID: 35481689 PMCID: PMC9542180 DOI: 10.1002/rmv.2354] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 03/16/2022] [Accepted: 04/01/2022] [Indexed: 12/25/2022]
Abstract
Human noroviruses are the leading global cause of viral gastroenteritis. Attempts at developing effective vaccines and treatments against norovirus disease have been stymied by the extreme genetic diversity and rapid geographic distribution of these viruses. The emergence and replacement of predominantly circulating norovirus genotypes has primarily been attributed to mutations on the VP1 capsid protein leading to genetic drift, and more recently to recombination events between the ORF1/ORF2 junction. However, large‐scale research into the historical and geographic distribution of recombinant norovirus strains has been limited in the literature. We performed a comprehensive historical analysis on 30,810 human norovirus sequences submitted to public databases between the years 1995 and 2019. During this time, 37 capsid genotypes and 56 polymerase types were detected across 90 different countries, and 97 unique recombinant genomes were also identified. GII.4, both capsid and polymerase, was the predominately circulating type worldwide for the majority of this time span, save for a brief swell of GII.17 and GII.2 capsid genotypes and a near‐total eclipse by GII.P16, GII.P21 and GII.P31 beginning in 2013. Interestingly, an analysis of 4067 recombinants found that 50.2% (N = 2039) of all recorded sequences belonged to three recently emerged recombinant strains: GII.2[P16], GII.4[P31], and GII.4[P16]. This analysis should provide an important historical foundation for future studies that evaluate the emergence and distribution of noroviruses, as well as the design of cross‐protective vaccines.
Collapse
Affiliation(s)
- Joseph A Kendra
- Division of Viral Products, CBER, FDA, Silver Spring, Maryland, USA
| | - Kentaro Tohma
- Division of Viral Products, CBER, FDA, Silver Spring, Maryland, USA
| | - Gabriel I Parra
- Division of Viral Products, CBER, FDA, Silver Spring, Maryland, USA
| |
Collapse
|
21
|
Zhu X, He Y, Wei X, Kong X, Zhang Q, Li J, Jin M, Duan Z. Molecular Epidemiological Characteristics of Gastroenteritis Outbreaks Caused by Norovirus GII.4 Sydney [P31] Strains - China, October 2016-December 2020. China CDC Wkly 2021; 3:1127-1132. [PMID: 35036035 PMCID: PMC8742140 DOI: 10.46234/ccdcw2021.276] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 12/10/2021] [Indexed: 11/14/2022] Open
Abstract
Introduction Human noroviruses are the leading cause of acute viral gastroenteritis (AGE) worldwide in all age groups. GII.4 strains have been the predominant genotype circulating globally over the last 2 decades and since 2012. GII.4 Sydney viruses have emerged and caused the majority of AGE outbreaks worldwide. Methods Data from norovirus outbreaks from the laboratory-based surveillance of norovirus outbreaks in China (CaliciNet China) between October 2016-December 2020 were analyzed. Results During October 2016-December 2020, 1,954 norovirus outbreaks were reported, and positive fecal samples from 1,352 (69.19%) outbreaks were genotyped. GII.4 Sydney [P31] viruses accounted for 2.1% (October 2016-August 2017), 5.5% (September 2017-August 2018), 3.3% (September 2018-August 2018), 26.6% (September 2019-August 2020), and and 1.1% (September 2020-December 2020) of GII outbreaks, respectively. Compared to reference strains of GII.4 Sydney [P31] from 2012 to 2013, 7 amino acid mutations in epitopes[A (297, 372 and 373), B (333), E (414), and H (309 and 310)] and 1 in human histo-blood group antigens binding site at site II 372 were found by analyzing 9 GII.4 Sydney [P31] complete genomic sequences. Conclusions This report identified the genomic variation of GII.4 Sydney [P31] from CaliciNet China. Continued surveillance with prompt genotyping and genetic analysis is necessary to monitor the emergence of novel GII.4 variants.
Collapse
Affiliation(s)
- Xi Zhu
- Key Laboratory of Medical Virology and Viral Diseases, Ministry of Health of the People's Republic of China, Beijing, China; National Institute for Viral Disease Control and Prevention, China CDC, Beijing, China
| | - Yaqing He
- Shenzhen Center for Disease Control and Prevention, Shenzhen, Guangdong, China
| | - Xingyan Wei
- Key Laboratory of Medical Virology and Viral Diseases, Ministry of Health of the People's Republic of China, Beijing, China; National Institute for Viral Disease Control and Prevention, China CDC, Beijing, China
| | - Xiangyu Kong
- Key Laboratory of Medical Virology and Viral Diseases, Ministry of Health of the People's Republic of China, Beijing, China; National Institute for Viral Disease Control and Prevention, China CDC, Beijing, China
| | - Qing Zhang
- Key Laboratory of Medical Virology and Viral Diseases, Ministry of Health of the People's Republic of China, Beijing, China; National Institute for Viral Disease Control and Prevention, China CDC, Beijing, China
| | - Jingxin Li
- Key Laboratory of Medical Virology and Viral Diseases, Ministry of Health of the People's Republic of China, Beijing, China; National Institute for Viral Disease Control and Prevention, China CDC, Beijing, China
| | - Miao Jin
- Key Laboratory of Medical Virology and Viral Diseases, Ministry of Health of the People's Republic of China, Beijing, China; National Institute for Viral Disease Control and Prevention, China CDC, Beijing, China
| | - Zhaojun Duan
- Key Laboratory of Medical Virology and Viral Diseases, Ministry of Health of the People's Republic of China, Beijing, China; National Institute for Viral Disease Control and Prevention, China CDC, Beijing, China
| |
Collapse
|
22
|
Li W, Yan H, Liu B, Tian Y, Chen Y, Jia L, Gao Z, Wang Q. Epidemiological characteristics and genetic diversity of norovirus infections among outpatient children with diarrhea under 5 years of age in Beijing, China, 2011-2018. Gut Pathog 2021; 13:77. [PMID: 34952625 PMCID: PMC8709959 DOI: 10.1186/s13099-021-00473-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 12/07/2021] [Indexed: 11/10/2022] Open
Abstract
Background Human noroviruses are the leading cause of sporadic cases and outbreaks of viral acute gastroenteritis in all age groups worldwide. Methods Epidemiological data and fecal specimens were collected between January 2011 and December 2018 from 4911 children < 5 years of age with diarrhea in three districts of Beijing. From 2011 to 2013, One-Step Reverse Transcription Polymerase Chain Reaction (RT-PCR) was used to detect noroviruses, and from January 2014 to December 2018, norovirus GI and GII were screened using duplex quantitative real-time RT-PCR (qRT-PCR). One-Step RT-PCR and RT-seminested PCR were performed to amplify the RNA-dependent polymerase and capsid genes of noroviruses in positive sample. Amplified products were sequenced directly; norovirus was typed using the online Norovirus Genotyping Tool v2.0 and phylogenetic analyses were conducted using MEGA-X. Results From 2011 to 2018, noroviruses were detected in 16.5% of specimens from children with diarrhea. The highest prevalence was observed in children aged 12 to 23 months (22.4%, 319/1421), followed by children aged 6 to 11 months (17.6%, 253/1441). The highest prevalence of norovirus infections occurred in autumn followed by winter, spring, and summer. From 2011 to 2018, the most prevalent dual types (genotype and polymerase type) were GII.4 Sydney[P31] (51.6%, 239/463), followed by GII.3[P12] (24.0%, 111/463), GII.4 2006b[P4 2006b] (7.3%, 34/463), GII.2[P16] (5.0%, 23/463), GII.17[P17] (2.6%, 12/463) and GII.6[P7] (2.6%, 12/463). GII.4 2006b[P4 2006b] predominated in 2011 and 2012. GII.4 Sydney[P31] predominated from 2013 to 2018. In total, 15 genotypes, 15 P-types and 19 dual types were detected in this study, reflecting the genetic diversity. Conclusions There were significant epidemiological characteristics and genetic diversity among outpatient children with norovirus infections < 5 years of age in Beijing from 2011 to 2018. These characteristics differ from those of norovirus outbreaks in Beijing. The complete genome sequences of each genotype are needed to better understand norovirus evolutionary mechanisms.
Collapse
Affiliation(s)
- Weihong Li
- Institute for Infectious Disease and Endemic Disease Control, Beijing Center for Disease Prevention and Control and Beijing Research Center for Preventive Medicine, Beijing, China
| | - Hanqiu Yan
- Institute for Infectious Disease and Endemic Disease Control, Beijing Center for Disease Prevention and Control and Beijing Research Center for Preventive Medicine, Beijing, China
| | - Baiwei Liu
- Institute for Infectious Disease and Endemic Disease Control, Beijing Center for Disease Prevention and Control and Beijing Research Center for Preventive Medicine, Beijing, China
| | - Yi Tian
- Institute for Infectious Disease and Endemic Disease Control, Beijing Center for Disease Prevention and Control and Beijing Research Center for Preventive Medicine, Beijing, China
| | - Yanwei Chen
- Institute for Infectious Disease and Endemic Disease Control, Beijing Center for Disease Prevention and Control and Beijing Research Center for Preventive Medicine, Beijing, China
| | - Lei Jia
- Institute for Infectious Disease and Endemic Disease Control, Beijing Center for Disease Prevention and Control and Beijing Research Center for Preventive Medicine, Beijing, China
| | - Zhiyong Gao
- Institute for Infectious Disease and Endemic Disease Control, Beijing Center for Disease Prevention and Control and Beijing Research Center for Preventive Medicine, Beijing, China.
| | - Quanyi Wang
- Institute for Infectious Disease and Endemic Disease Control, Beijing Center for Disease Prevention and Control and Beijing Research Center for Preventive Medicine, Beijing, China.
| |
Collapse
|
23
|
Han JC, Li QX, Fang JB, Zhang JY, Li YQ, Li SZ, Cheng C, Xie CZ, Nan FL, Zhang H, Li ZX, Jin NY, Zhu GZ, Lu HJ. GII.P16-GII.2 Recombinant Norovirus VLPs Polarize Macrophages Into the M1 Phenotype for Th1 Immune Responses. Front Immunol 2021; 12:781718. [PMID: 34868056 PMCID: PMC8637406 DOI: 10.3389/fimmu.2021.781718] [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] [Scholar Register] [Received: 09/23/2021] [Accepted: 11/02/2021] [Indexed: 11/13/2022] Open
Abstract
Norovirus (NoV) is a zoonotic virus that causes diarrhea in humans and animals. Outbreaks in nosocomial settings occur annually worldwide, endangering public health and causing serious social and economic burdens. The latter quarter of 2016 witnessed the emergence of the GII.P16-GII.2 recombinant norovirus throughout Asia. This genotype exhibits strong infectivity and replication characteristics, proposing its potential to initiate a pandemic. There is no vaccine against GII.P16-GII.2 recombinant norovirus, so it is necessary to design a preventive vaccine. In this study, GII.P16-GII.2 type norovirus virus-like particles (VLPs) were constructed using the baculovirus expression system and used to conduct immunizations in mice. After immunization of mice, mice were induced to produce memory T cells and specific antibodies, indicating that the VLPs induced specific cellular and humoral immune responses. Further experiments were then initiated to understand the underlying mechanisms involved in antigen presentation. Towards this, we established co-cultures between dendritic cells (DCs) or macrophages (Mø) and naïve CD4+T cells and simulated the antigen presentation process by incubation with VLPs. Thereafter, we detected changes in cell surface molecules, cytokines and related proteins. The results indicated that VLPs effectively promoted the phenotypic maturation of Mø but not DCs, as indicated by significant changes in the expression of MHC-II, costimulatory factors and related cytokines in Mø. Moreover, we found VLPs caused Mø to polarize to the M1 type and release inflammatory cytokines, thereby inducing naïve CD4+ T cells to perform Th1 immune responses. Therefore, this study reveals the mechanism of antigen presentation involving GII.P16-GII.2 recombinant norovirus VLPs, providing a theoretical basis for both understanding responses to norovirus infection as well as opportunities for vaccine development.
Collapse
Affiliation(s)
- Ji Cheng Han
- Academician Workstation, Changchun University of Chinese Medicine, Changchun, China
| | - Qiu Xuan Li
- Academician Workstation, Changchun University of Chinese Medicine, Changchun, China.,College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Jin Bo Fang
- Academician Workstation, Changchun University of Chinese Medicine, Changchun, China
| | - Jin Yong Zhang
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China.,Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Yi Quan Li
- Academician Workstation, Changchun University of Chinese Medicine, Changchun, China
| | - Shan Zhi Li
- Academician Workstation, Changchun University of Chinese Medicine, Changchun, China
| | - Cheng Cheng
- Academician Workstation, Changchun University of Chinese Medicine, Changchun, China.,College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Chang Zhan Xie
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China.,Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Fu Long Nan
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China.,Department of Specialty Medicine, School of Basic Medicine, Qingdao University, Qingdao, China
| | - He Zhang
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Zhuo Xin Li
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China.,Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Ning Yi Jin
- Academician Workstation, Changchun University of Chinese Medicine, Changchun, China.,Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Guang Ze Zhu
- Academician Workstation, Changchun University of Chinese Medicine, Changchun, China
| | - Hui Jun Lu
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| |
Collapse
|
24
|
Xu Y, Zhu Y, Lei Z, Rui J, Zhao Z, Lin S, Wang Y, Xu J, Liu X, Yang M, Chen H, Pan X, Lu W, Du Y, Li H, Fang L, Zhang M, Zhou L, Yang F, Chen T. Investigation and analysis on an outbreak of norovirus infection in a health school in Guangdong Province, China. INFECTION, GENETICS AND EVOLUTION : JOURNAL OF MOLECULAR EPIDEMIOLOGY AND EVOLUTIONARY GENETICS IN INFECTIOUS DISEASES 2021; 96:105135. [PMID: 34781036 DOI: 10.1016/j.meegid.2021.105135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 10/14/2021] [Accepted: 11/08/2021] [Indexed: 06/13/2023]
Abstract
Our objective was to describe the epidemiological features of an outbreak of norovirus infection in a health school in Guangdong province, China, to identify the cause of such a large scale outbreak of norovirus among older students, to simulate the transmission dynamics, and to evaluate the effect of intervention measures of GII.17 [P17] genotype norovirus infection. We identified all cases during the outbreak. Descriptive epidemiological, analytical epidemiological and hygiene survey methods were used to described the outbreak epidemic course and identify the cause of the outbreak of norovirus infection. We also used dynamical model to simulate the transmission dynamics of norovirus infection and evaluate the effect of intervention measures. Norovirus genotyping was assigned to the newly obtained strains, with a maximum likelihood phylogenetic analysis conducted. There were 360 cases of 42 classes in five grades with a 12.99% attack rate. Proportionally, more students were in contact with sick students and vomit in the suspected case group than the control group (χ2 = 5.535, P = 0.019 and χ2 = 5.549, P = 0.019, respectively). The basic reproduction number was 8.32 before and 0.49 after the intervention. Dynamical modeling showed that if the isolation rate was higher or case isolation began earlier, the total attack rate would decrease. Molecular characterization identified the GII.17 [P17] genotype in all stains obtained from the health school, which were clustered with high support in the phylogenetic tree. This was an outbreak of norovirus infection caused by contact transmission. The main reasons for the spread of the epidemic were the later control time, irregular treatment of vomit and no case isolation. The transmission dynamics of contact transmission was high, more efficient control measures should be employed.
Collapse
Affiliation(s)
- Yucheng Xu
- Futian District Center for Disease Control and Prevention, Shenzhen, People's Republic of China; Guangdong Field Epidemiology Training Program, Guangzhou, People's Republic of China
| | - Yuanzhao Zhu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen City 361102, Fujian Province, People's Republic of China
| | - Zhao Lei
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen City 361102, Fujian Province, People's Republic of China
| | - Jia Rui
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen City 361102, Fujian Province, People's Republic of China
| | - Zeyu Zhao
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen City 361102, Fujian Province, People's Republic of China
| | - Shengnan Lin
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen City 361102, Fujian Province, People's Republic of China
| | - Yao Wang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen City 361102, Fujian Province, People's Republic of China
| | - Jingwen Xu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen City 361102, Fujian Province, People's Republic of China
| | - Xingchun Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen City 361102, Fujian Province, People's Republic of China
| | - Meng Yang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen City 361102, Fujian Province, People's Republic of China
| | - Hongsheng Chen
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, People's Republic of China
| | - Xuemei Pan
- Lianzhou District Center for Disease Control and Prevention, Qingyuan, People's Republic of China
| | - Wentao Lu
- Qingyuan City Center for Disease Control and Prevention, Qingyuan, People's Republic of China
| | - Yuzhong Du
- Qingyuan City Center for Disease Control and Prevention, Qingyuan, People's Republic of China
| | - Hui Li
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, People's Republic of China
| | - Ling Fang
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, People's Republic of China
| | - Meng Zhang
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, People's Republic of China
| | - Lina Zhou
- Department of Nephrology, The second Hospital of Xiamen Medical college, Xiamen 361021, China
| | - Fen Yang
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, People's Republic of China.
| | - Tianmu Chen
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen City 361102, Fujian Province, People's Republic of China.
| |
Collapse
|
25
|
Antigenic cartography reveals complexities of genetic determinants that lead to antigenic differences among pandemic GII.4 noroviruses. Proc Natl Acad Sci U S A 2021; 118:2015874118. [PMID: 33836574 PMCID: PMC7980451 DOI: 10.1073/pnas.2015874118] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Noroviruses are the predominant cause of acute gastroenteritis, with a single genotype (GII.4) responsible for the majority of infections. This prevalence is characterized by the periodic emergence of new variants that present substitutions at antigenic sites of the major structural protein (VP1), facilitating escape from herd immunity. Notably, the contribution of intravariant mutations to changes in antigenic properties is unknown. We performed a comprehensive antigenic analysis on a virus-like particle panel representing major chronological GII.4 variants to investigate diversification at the inter- and intravariant level. Immunoassays, neutralization data, and cartography analyses showed antigenic similarities between phylogenetically related variants, with major switches to antigenic properties observed over the evolution of GII.4 variants. Genetic analysis indicated that multiple coevolving amino acid changes-primarily at antigenic sites-are associated with the antigenic diversification of GII.4 variants. These data highlight complexities of the genetic determinants and provide a framework for the antigenic characterization of emerging GII.4 noroviruses.
Collapse
|
26
|
Zuo Y, Xue L, Gao J, Liao Y, Jiang Y, Li Y, Liang Y, Wang L, Cai W, Cheng T, Wang J, Chen M, Zhang J, Ding Y, Wu Q. Development and Application of a Novel Rapid and Throughput Method for Broad-Spectrum Anti-Foodborne Norovirus Antibody Testing. Front Microbiol 2021; 12:670488. [PMID: 34539594 PMCID: PMC8446669 DOI: 10.3389/fmicb.2021.670488] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Accepted: 08/13/2021] [Indexed: 11/22/2022] Open
Abstract
Foodbone norovirus (NoV) is the leading cause of acute gastroenteritis worldwide. Candidate vaccines are being developed, however, no licensed vaccines are currently available for managing NoV infections. Screening for stimulated antibodies with broad-spectrum binding activities can be performed for the development of NoV polyvalent vaccines. In this study, we aimed to develop an indirect enzyme-linked immunosorbent assay (ELISA) for testing the broad spectrum of anti-NoV antibodies. Capsid P proteins from 28 representative NoV strains (GI.1–GI.9 and GII.1–GII.22 except GII.11, GII.18, and GII.19) were selected, prepared, and used as coating antigens on one microplate. Combined with incubation and the horseradish peroxidase chromogenic reaction, the entire process for testing the spectrum of unknown antibodies required 2 h for completion. The intra-assay and inter-assay coefficients of variation were less than 10%. The new method was successfully performed with monoclonal antibodies and polyclonal antibodies induced by multiple antigens. In conclusion, the indirect ELISA assay developed in this study had a good performance of reliability, convenience, and high-throughput screening for broad-spectrum antibodies.
Collapse
Affiliation(s)
- Yueting Zuo
- School of Bioscience and Bioengineering, South China University of Technology, Guangzhou, China.,Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Liang Xue
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Junshan Gao
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Yingyin Liao
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Yueting Jiang
- Department of Laboratory Medicine, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ying Li
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Yanhui Liang
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Linping Wang
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Weicheng Cai
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Tong Cheng
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Juan Wang
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Moutong Chen
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Jumei Zhang
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Yu Ding
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Qingping Wu
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| |
Collapse
|
27
|
Tohma K, Lepore CJ, Martinez M, Degiuseppe JI, Khamrin P, Saito M, Mayta H, Nwaba AUA, Ford-Siltz LA, Green KY, Galeano ME, Zimic M, Stupka JA, Gilman RH, Maneekarn N, Ushijima H, Parra GI. Genome-wide analyses of human noroviruses provide insights on evolutionary dynamics and evidence of coexisting viral populations evolving under recombination constraints. PLoS Pathog 2021; 17:e1009744. [PMID: 34255807 PMCID: PMC8318288 DOI: 10.1371/journal.ppat.1009744] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 07/28/2021] [Accepted: 06/23/2021] [Indexed: 12/14/2022] Open
Abstract
Norovirus is a major cause of acute gastroenteritis worldwide. Over 30 different genotypes, mostly from genogroup I (GI) and II (GII), have been shown to infect humans. Despite three decades of genome sequencing, our understanding of the role of genomic diversification across continents and time is incomplete. To close the spatiotemporal gap of genomic information of human noroviruses, we conducted a large-scale genome-wide analyses that included the nearly full-length sequencing of 281 archival viruses circulating since the 1970s in over 10 countries from four continents, with a major emphasis on norovirus genotypes that are currently underrepresented in public genome databases. We provided new genome information for 24 distinct genotypes, including the oldest genome information from 12 norovirus genotypes. Analyses of this new genomic information, together with those publicly available, showed that (i) noroviruses evolve at similar rates across genomic regions and genotypes; (ii) emerging viruses evolved from transiently-circulating intermediate viruses; (iii) diversifying selection on the VP1 protein was recorded in genotypes with multiple variants; (iv) non-structural proteins showed a similar branching on their phylogenetic trees; and (v) contrary to the current understanding, there are restrictions on the ability to recombine different genomic regions, which results in co-circulating populations of viruses evolving independently in human communities. This study provides a comprehensive genetic analysis of diverse norovirus genotypes and the role of non-structural proteins on viral diversification, shedding new light on the mechanisms of norovirus evolution and transmission. Norovirus is a highly diverse enteric pathogen. The large genomic database accumulated in the last three decades advanced our understanding of norovirus diversity; however, this information is limited by geographical bias, sporadic times of collection, and missing or incomplete genome sequences. In this multinational collaborative study, we mined archival samples collected since the 1970s and sequenced nearly full-length new genomes from 281 historical noroviruses, including the first full-length genomic sequences for three genotypes. Using this novel dataset, we found evidence for restrictions in the recombination of genetically disparate viruses and that diversifying selection results in new variants with different epidemiological profiles. These new insights on the diversification of noroviruses could provide baseline information for the study of future epidemics and ultimately the prevention of norovirus infections.
Collapse
Affiliation(s)
- Kentaro Tohma
- Division of Viral Products, CBER, FDA, Silver Spring, Maryland, United States of America
| | - Cara J. Lepore
- Division of Viral Products, CBER, FDA, Silver Spring, Maryland, United States of America
| | - Magaly Martinez
- Division of Viral Products, CBER, FDA, Silver Spring, Maryland, United States of America
- IICS, National University of Asuncion, Asuncion, Paraguay
| | | | - Pattara Khamrin
- Department of Microbiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Mayuko Saito
- Department of Virology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Holger Mayta
- Department of Cellular and Molecular Sciences, Faculty of Sciences, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Amy U. Amanda Nwaba
- Division of Viral Products, CBER, FDA, Silver Spring, Maryland, United States of America
| | - Lauren A. Ford-Siltz
- Division of Viral Products, CBER, FDA, Silver Spring, Maryland, United States of America
| | - Kim Y. Green
- Laboratory of Infectious Diseases, NIAID, NIH, Bethesda, Maryland, United States of America
| | | | - Mirko Zimic
- Department of Cellular and Molecular Sciences, Faculty of Sciences, Universidad Peruana Cayetano Heredia, Lima, Peru
| | | | - Robert H. Gilman
- Department of International Health, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Niwat Maneekarn
- Department of Microbiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Hiroshi Ushijima
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo, Japan
| | - Gabriel I. Parra
- Division of Viral Products, CBER, FDA, Silver Spring, Maryland, United States of America
- * E-mail:
| |
Collapse
|
28
|
Jin M, Wu S, Kong X, Xie H, Fu J, He Y, Feng W, Liu N, Li J, Rainey JJ, Hall AJ, Vinjé J, Duan Z. Norovirus Outbreak Surveillance, China, 2016-2018. Emerg Infect Dis 2021; 26:437-445. [PMID: 32091361 PMCID: PMC7045832 DOI: 10.3201/eid2603.191183] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
CaliciNet China, a network of provincial, county, and city laboratories coordinated by the Chinese Centers for Disease Control and Prevention, was launched in October 2016 to monitor the epidemiology and genotype distribution of norovirus outbreaks in China. During October 2016–September 2018, a total of 556 norovirus outbreaks were reported, and positive fecal samples from 470 (84.5%) outbreaks were genotyped. Most of these outbreaks were associated with person-to-person transmission (95.1%), occurred in childcare centers or schools (78.2%), and were reported during November–March of each year (63.5%). During the 2-year study period, 81.2% of all norovirus outbreaks were typed as GII.2[P16]. In China, most norovirus outbreaks are reported by childcare centers or schools; GII.2[P16] is the predominant genotype. Ongoing surveillance by CaliciNet China will provide information about the evolving norovirus genotype distribution and outbreak characteristics important for the development of effective interventions, including vaccines.
Collapse
|
29
|
Molecular epidemiology and genetic diversity of norovirus infection in children hospitalized with acute gastroenteritis in East Java, Indonesia in 2015-2019. INFECTION GENETICS AND EVOLUTION 2021; 88:104703. [PMID: 33401005 DOI: 10.1016/j.meegid.2020.104703] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 12/02/2020] [Accepted: 12/30/2020] [Indexed: 12/12/2022]
Abstract
Noroviruses are recognized as a leading cause of outbreaks and sporadic cases of acute gastroenteritis (AGE) among individuals of all ages worldwide, especially in children <5 years old. We investigated the epidemiology of noroviruses among hospitalized children at two hospitals in East Java, Indonesia. Stool samples were collected from 966 children with AGE during September 2015-July 2019. All samples were analyzed by reverse transcription-polymerase chain reaction (RT-PCR) for the amplification of both the RNA-dependent RNA polymerase (RdRp) and the capsid genes of noroviruses. The genotypes were determined by phylogenetic analyses. In 2015-2019, noroviruses were detected in 12.3% (119/966) of the samples. Children <2 years old showed a significantly higher prevalence than those ≥2 years old (P = 0.01). NoV infections were observed throughout the year, with the highest prevalence in December. Based on our genetic analyses of RdRp, GII.[P31] (43.7%, 31/71) was the most prevalent RdRp genotype, followed by GII.[P16] (36.6%, 26/71). GII.[P31] was a dominant genotype in 2016 and 2018, whereas GII.[P16] was a dominant genotype in 2015 and 2017. Among the capsid genotypes, the most predominant norovirus genotype from 2015 to 2018 was GII.4 Sydney_2012 (33.6%, 40/119). The most prevalent genotype in each year was GII.13 in 2015, GII.4 Sydney_2012 in 2016 and 2018, and GII.3 in 2017. Based on the genetic analyses of RdRp and capsid sequences, the strains were clustered into 13 RdRp/capsid genotypes; 12 of them were discordant, e.g., GII.4 Sydney[P31], GII.3[P16], and GII.13[P16]. The predominant genotype in each year was GII.13[P16] in 2015, GII.4 Sydney[P31] in 2016, GII.3[P16] in 2017, and GII.4 Sydney[P31] in 2018. Our results demonstrate high detection rates and genetic diversity of norovirus GII genotypes in pediatric AGE samples from Indonesia. These findings strengthen the importance of the continuous molecular surveillance of emerging norovirus strains.
Collapse
|
30
|
Interaction between norovirus and Histo-Blood Group Antigens: A key to understanding virus transmission and inactivation through treatments? Food Microbiol 2020; 92:103594. [PMID: 32950136 DOI: 10.1016/j.fm.2020.103594] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 06/27/2020] [Accepted: 07/02/2020] [Indexed: 02/06/2023]
Abstract
Human noroviruses (HuNoVs) are a main cause of acute gastroenteritis worldwide. They are frequently involved in foodborne and waterborne outbreaks. Environmental transmission of the virus depends on two main factors: the ability of viral particles to remain infectious and their adhesion capacity onto different surfaces. Until recently, adhesion of viral particles to food matrices was mainly investigated by considering non-specific interactions (e.g. electrostatic, hydrophobic) and there was only limited information about infectious HuNoVs because of the absence of a reliable in vitro HuNoV cultivation system. Many HuNoV strains have now been described as having specific binding interactions with human Histo-Blood Group Antigens (HBGAs) and non-HBGA ligands found in food and the environment. Relevant approaches to the in vitro replication of HuNoVs were also proposed recently. On the basis of the available literature data, this review discusses the opportunities to use this new knowledge to obtain a better understanding of HuNoV transmission to human populations and better evaluate the hazard posed by HuNoVs in foodstuffs and the environment.
Collapse
|
31
|
Derya SM, Spiegel H, Hanisch FG, Morozov V, Schroten H, Jennewein S, Parschat K. Biotechnologically produced fucosylated oligosaccharides inhibit the binding of human noroviruses to their natural receptors. J Biotechnol 2020; 318:31-38. [PMID: 32387450 DOI: 10.1016/j.jbiotec.2020.05.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 04/10/2020] [Accepted: 05/01/2020] [Indexed: 01/17/2023]
Abstract
Norovirus infections cause severe gastroenteritis in millions of people every year. Infection requires the recognition of histo-blood group antigens (HBGAs), but such interactions can be inhibited by human milk oligosaccharides (HMOs), which act as structurally-similar decoys. HMO supplements could help to prevent norovirus infections, but the industrial production of complex HMOs is challenging. Here we describe a large-scale fermentation process that yields several kilograms of lacto-N-fucopentaose I (LNFP I). The product was synthesized in Escherichia coli BL21(DE3) cells expressing a recombinant N-acetylglucosaminyltransferase, β(1,3)galactosyltransferase and α(1,2)fucosyltransferase. Subsequent in vitro enzymatic conversion produced HBGA types A1 and B1 for norovirus inhibition assays. These carbohydrates inhibited the binding of GII.17 virus-like particles (VLPs) to type A1 and B1 trisaccharides more efficiently than simpler fucosylated HMOs, which were in turn more effective than any non-fucosylated structures. However, we found that the simpler fucosylated HMOs were more effective than complex molecules such as LNFP I when inhibiting the binding of GII.17 and GII.4 VLPs to human gastric mucins and mucins from human amniotic fluid. Our results show that complex fucosylated HMOs can be produced by large-scale fermentation and that a combination of simple and complex fucosylated structures is more likely to prevent norovirus infections.
Collapse
Affiliation(s)
- Sami M Derya
- Jennewein Biotechnologie GmbH, 53619 Rheinbreitbach, Germany.
| | - Holger Spiegel
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, 52074 Aachen, Germany.
| | - Franz-Georg Hanisch
- Institute of Biochemistry II, Medical Faculty, University of Cologne, 50931 Köln, Germany.
| | - Vasily Morozov
- Pediatric Infectious Disease Unit, University Children's Hospital Mannheim, 68167 Mannheim, Germany.
| | - Horst Schroten
- Pediatric Infectious Disease Unit, University Children's Hospital Mannheim, 68167 Mannheim, Germany.
| | - Stefan Jennewein
- Jennewein Biotechnologie GmbH, 53619 Rheinbreitbach, Germany; Fraunhofer Institute for Molecular Biology and Applied Ecology IME, 52074 Aachen, Germany.
| | - Katja Parschat
- Jennewein Biotechnologie GmbH, 53619 Rheinbreitbach, Germany.
| |
Collapse
|
32
|
Chen C, Wu B, Zhang H, Li KF, Liu R, Wang HL, Yan JB. Molecular evolution of GII.P17-GII.17 norovirus associated with sporadic acute gastroenteritis cases during 2013-2018 in Zhoushan Islands, China. Virus Genes 2020; 56:279-287. [PMID: 32065329 DOI: 10.1007/s11262-020-01744-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2019] [Accepted: 02/10/2020] [Indexed: 12/13/2022]
Abstract
In this study, we investigated the molecular characteristics and spatio-temporal dynamics of GII.P17-GII.17 norovirus in Zhoushan Islands during 2013-2018. We collected 1849 samples from sporadic acute gastroenteritis patients between January 2013 and August 2018 in Zhoushan Islands, China. Among the 1849 samples, 134 (7.24%) samples were positive for human norovirus (HuNoV). The complete sequence of GII.17 VP1 gene was amplified from 31 HuNoV-positive samples and sequenced. A phylogenetic tree was constructed based on the full-length sequence of the VP1 gene. Phylogenetic analysis revealed that the GII.17 genotype detected during 2014-2018 belongs to the new GII.17 Kawasaki variant. Divergence analysis revealed that the time of the most recent common ancestor (TMRCA) of GII.17 in Zhoushan Islands was estimated to be between 1997 and 1998. The evolutionary rate of the VP1 gene of the GII.17 genotype norovirus was 1.14 × 10-3 (95% HPD: 0.62-1.73 × 10-3) nucleotide substitutions/site/year. The spatio-temporal diffusion analysis of the GII.17 genotype identified Hong Kong as the epicenter for GII.17 dissemination. The VP1 gene sequence of Zhoushan Island isolates correlated with that of Hong Kong and Japan isolates.
Collapse
Affiliation(s)
- Can Chen
- Zhoushan Center for Disease Control and Prevention, Zhoushan, Zhejiang Province, China.,Zhejiang Provincial Key Laboratory of Health Risk Factors for Seafood, Zhoushan Center for Disease Control and Prevention, Zhoushan, Zhejiang Province, China.,Jiangxi Province Key Laboratory of Preventive Medicine, School of Public Health, Nanchang University, Nanchang, 330006, Jiangxi, China.,State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affifiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Bing Wu
- Zhoushan Center for Disease Control and Prevention, Zhoushan, Zhejiang Province, China.,Zhejiang Provincial Key Laboratory of Health Risk Factors for Seafood, Zhoushan Center for Disease Control and Prevention, Zhoushan, Zhejiang Province, China
| | - Hui Zhang
- Zhoushan Center for Disease Control and Prevention, Zhoushan, Zhejiang Province, China.,Zhejiang Provincial Key Laboratory of Health Risk Factors for Seafood, Zhoushan Center for Disease Control and Prevention, Zhoushan, Zhejiang Province, China
| | - Ke-Feng Li
- Zhoushan Center for Disease Control and Prevention, Zhoushan, Zhejiang Province, China.,Zhejiang Provincial Key Laboratory of Health Risk Factors for Seafood, Zhoushan Center for Disease Control and Prevention, Zhoushan, Zhejiang Province, China
| | - Rong Liu
- Jiangxi Province Key Laboratory of Preventive Medicine, School of Public Health, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Hong-Ling Wang
- Zhoushan Center for Disease Control and Prevention, Zhoushan, Zhejiang Province, China. .,Zhejiang Provincial Key Laboratory of Health Risk Factors for Seafood, Zhoushan Center for Disease Control and Prevention, Zhoushan, Zhejiang Province, China.
| | - Jian-Bo Yan
- Zhoushan Center for Disease Control and Prevention, Zhoushan, Zhejiang Province, China. .,Zhejiang Provincial Key Laboratory of Health Risk Factors for Seafood, Zhoushan Center for Disease Control and Prevention, Zhoushan, Zhejiang Province, China. .,Jiangxi Province Key Laboratory of Preventive Medicine, School of Public Health, Nanchang University, Nanchang, 330006, Jiangxi, China.
| |
Collapse
|
33
|
Matsushima Y, Mizukoshi F, Sakon N, Doan YH, Ueki Y, Ogawa Y, Motoya T, Tsukagoshi H, Nakamura N, Shigemoto N, Yoshitomi H, Okamoto-Nakagawa R, Suzuki R, Tsutsui R, Terasoma F, Takahashi T, Sadamasu K, Shimizu H, Okabe N, Nagasawa K, Aso J, Ishii H, Kuroda M, Ryo A, Katayama K, Kimura H. Evolutionary Analysis of the VP1 and RNA-Dependent RNA Polymerase Regions of Human Norovirus GII.P17-GII.17 in 2013-2017. Front Microbiol 2019; 10:2189. [PMID: 31611853 PMCID: PMC6777354 DOI: 10.3389/fmicb.2019.02189] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 09/05/2019] [Indexed: 01/05/2023] Open
Abstract
Human norovirus (HuNoV) GII.P17-GII.17 (Kawasaki2014 variant) reportedly emerged in 2014 and caused gastroenteritis outbreaks worldwide. To clarify the evolution of both VP1 and RNA-dependent RNA polymerase (RdRp) regions of GII.P17-GII.17, we analyzed both global and novel Japanese strains detected during 2013-2017. Time-scaled phylogenetic trees revealed that the ancestral GII.17 VP1 region diverged around 1949, while the ancestral GII.P17 RdRp region diverged around 2010. The evolutionary rates of the VP1 and RdRp regions were estimated at ~2.7 × 10-3 and ~2.3 × 10-3 substitutions/site/year, respectively. The phylogenetic distances of the VP1 region exhibited no overlaps between intra-cluster and inter-cluster peaks in the GII.17 strains, whereas those of the RdRp region exhibited a unimodal distribution in the GII.P17 strains. Conformational epitope positions in the VP1 protein of the GII.P17-GII.17 strains were similar, although some substitutions, insertions and deletions had occurred. Strains belonging to the same cluster also harbored substitutions around the binding sites for the histo-blood group antigens of the VP1 protein. Moreover, some amino acid substitutions were estimated to be near the interface between monomers and the active site of the RdRp protein. These results suggest that the GII.P17-GII.17 virus has produced variants with the potential to alter viral antigenicity, host-binding capability, and replication property over the past 10 years.
Collapse
Affiliation(s)
- Yuki Matsushima
- Division of Virology, Kawasaki City Institute for Public Health, Kawasaki, Japan
| | - Fuminori Mizukoshi
- Department of Microbiology, Tochigi Prefectural Institute of Public Health and Environmental Science, Utsunomiya, Japan
| | - Naomi Sakon
- Department of Microbiology, Osaka Institute of Public Health, Osaka, Japan
| | - Yen Hai Doan
- Department of Virology II, National Institute of Infectious Diseases, Musashimurayama, Japan
| | - Yo Ueki
- Department of Microbiology, Miyagi Prefectural Institute of Public Health and Environment, Sendai, Japan
| | - Yasutaka Ogawa
- Division of Virology, Saitama Institute of Public Health, Saitama, Japan
| | - Takumi Motoya
- Ibaraki Prefectural Institute of Public Health, Mito, Japan
| | - Hiroyuki Tsukagoshi
- Gunma Prefectural Institute of Public Health and Environmental Sciences, Maebashi, Japan
| | | | - Naoki Shigemoto
- Hiroshima Prefectural Technology Research Institute Public Health and Environment Center, Hiroshima, Japan
| | - Hideaki Yoshitomi
- Fukuoka Institute of Health and Environmental Sciences, Dazaifu, Japan
| | | | - Rieko Suzuki
- Kanagawa Prefectural Institute of Public Health, Chigasaki, Japan
| | - Rika Tsutsui
- Aomori Prefecture Public Health and Environment Center, Aomori, Japan
| | - Fumio Terasoma
- Wakayama Prefectural Research Center of Environment and Public Health, Wakayama, Japan
| | - Tomoko Takahashi
- Iwate Prefectural Research Institute for Environmental Sciences and Public Health, Morioka, Japan
| | - Kenji Sadamasu
- Department of Microbiology, Tokyo Metropolitan Institute of Public Health, Shinjuku, Japan
| | - Hideaki Shimizu
- Division of Virology, Kawasaki City Institute for Public Health, Kawasaki, Japan
| | - Nobuhiko Okabe
- Division of Virology, Kawasaki City Institute for Public Health, Kawasaki, Japan
| | | | - Jumpei Aso
- Graduate School of Health Sciences, Gunma Paz University, Takasaki, Japan
- Department of Respiratory Medicine, Kyorin University School of Medicine, Mitaka, Japan
| | - Haruyuki Ishii
- Department of Respiratory Medicine, Kyorin University School of Medicine, Mitaka, Japan
| | - Makoto Kuroda
- Pathogen Genomics Center, National Institute of Infectious Diseases, Musashimurayama, Japan
| | - Akihide Ryo
- Department of Microbiology, Yokohama City University, Graduate School of Medicine, Yokohama, Japan
| | - Kazuhiko Katayama
- Laboratory of Viral Infection I, Kitasato Institute for Life Sciences, Graduate School of Infection Control Sciences, Kitasato University, Minato, Japan
| | - Hirokazu Kimura
- Graduate School of Health Sciences, Gunma Paz University, Takasaki, Japan
- Department of Microbiology, Yokohama City University, Graduate School of Medicine, Yokohama, Japan
| |
Collapse
|
34
|
Li B, Xiao D, Li Y, Wu X, Qi L, Tang W, Li Q. Epidemiological analysis of norovirus infectious diarrhea outbreaks in Chongqing, China, from 2011 to 2016. J Infect Public Health 2019; 13:46-50. [PMID: 31548166 DOI: 10.1016/j.jiph.2019.06.019] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 06/01/2019] [Accepted: 06/13/2019] [Indexed: 11/17/2022] Open
Abstract
OBJECTIVE We investigated the epidemiological characteristics of norovirus infection from 2011 to 2016 in Chongqing, China, in order to provide evidence for strategies on epidemic prevention and control. METHODS We collected data on norovirus infectious diarrhea epidemics in 38 districts and counties, and analyzed the information using descriptive epidemiological methods. RESULTS In 2011, the first case of norovirus infectious diarrhea in Chongqing was reported. From 2011 to 2015, 38 districts and counties in Chongqing reported a total of 4 epidemics. In 2016, however, the city reported 117 outbreaks. From 2011 to 2016, there were 1637 cases of norovirus infection but no deaths. In 2016, most outbreaks occurred over a 5-month period with a clear peak in December and higher incidence in major urban areas than smaller communities (83.61% vs. 16.39%). Of these 1637 cases, 99.18% occurred in urban schools and nurseries, and 80% were transmitted person-to-person. Infection by genogroup II genotype 2 (GII.2) viruses accounted for 98.71% of cases. Leukocytes were increased in 67.81% of patients, neutrophils in 65%, and lymphocytes in 50%. Medical treatment was sought by 70% of patients or guardians but only 3.66% of cases were hospitalized. The most frequent misdiagnosis was "suspected food poisoning". CONCLUSION The frequency of norovirus infectious diarrhea epidemics increased over 20-fold from 2011 to 2016 in Chongqing, China. These epidemics occurred predominantly in urban schools and nurseries. However, epidemics showed little spread to outlying districts and counties, so prevention and control pressures were relatively high. SUGGESTIONS Healthcare professionals and institutions should strengthen health education for groups at high-risk of norovirus infection, such as school children, and increase norovirus testing capacity to further improve emergency investigation. Prevention and control knowledge should be disseminated to the general public to reduce transmission risk and total disease burden. Finally, governments and health administrative departments should invest special funds to prevent and control norovirus epidemics.
Collapse
Affiliation(s)
- Baisong Li
- Infectious Disease Control Office, The Chongqing Center for Disease Control and Prevention, Chongqing 400010, China
| | - Dayong Xiao
- Infectious Disease Control Office, The Chongqing Center for Disease Control and Prevention, Chongqing 400010, China
| | - Yanlin Li
- Hainan Medical University, Haikou 570100, China
| | - Xianlan Wu
- The Department of Clinical Laboratory Medicine, Southwest Hospital, Army Medical University, Chongqing 400000, China
| | - Li Qi
- Infectious Disease Control Office, The Chongqing Center for Disease Control and Prevention, Chongqing 400010, China
| | - Wenge Tang
- Infectious Disease Control Office, The Chongqing Center for Disease Control and Prevention, Chongqing 400010, China
| | - Qin Li
- Infectious Disease Control Office, The Chongqing Center for Disease Control and Prevention, Chongqing 400010, China.
| |
Collapse
|
35
|
Norovirus outbreaks in Beijing, China, from 2014 to 2017. J Infect 2019; 79:159-166. [DOI: 10.1016/j.jinf.2019.05.019] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 02/15/2019] [Accepted: 05/26/2019] [Indexed: 11/21/2022]
|
36
|
GII.13/21 Noroviruses Recognize Glycans with a Terminal β-Galactose via an Unconventional Glycan Binding Site. J Virol 2019; 93:JVI.00723-19. [PMID: 31118252 PMCID: PMC6639292 DOI: 10.1128/jvi.00723-19] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 05/01/2019] [Indexed: 01/17/2023] Open
Abstract
Evidence from both phenotypic binding assay and structural study support the observed interactions of human noroviruses (huNoVs) with histo-blood group antigens (HBGAs) as receptors or attachment factors, affecting their host susceptibility. GII.13 and GII.21 genotypes form a unique genetic lineage that differs from the mainstream GII huNoVs in their unconventional glycan binding site. Unlike the previous findings that GII.13/21 genotypes recognize only Lea antigen, we found in this study that they can interact with a group of glycans with a common terminal β-Gal, including Lec, lactose, and mucin core 2. However, this wide glycan binding spectrum in a unique binding mode of the GII.13/21 huNoVs appears not to increase their prevalence, probably due to the existence of decoy glycan receptors in human gastrointestinal tract limiting their infection. Our findings shed light on the host interaction and epidemiology of huNoVs, which would impact the strategy of huNoV control and prevention. Human noroviruses (huNoVs) recognize histo-blood group antigens (HBGAs) as host susceptibility factors. GII.13 and GII.21 huNoVs form a unique genetic lineage that emerged from mainstream GII NoVs via development of a new, nonconventional glycan binding site (GBS) that binds Lea antigen. This previous finding raised the question of whether the new GII.13/21 GBS really has such a narrow glycan binding spectrum. In this study, we provide solid phenotypic and structural evidence indicating that this new GBS recognizes a group of glycans with a common terminal β-galactose (β-Gal). First, we found that P domain proteins of GII.13/21 huNoVs circulating at different times bound three glycans sharing a common terminal β-Gal, including Lec, lactose, and mucin core 2. Second, we solved the crystal structures of the GII.13 P dimers in complex with Lec and mucin core 2, which showed that β-Gal is the major binding saccharide. Third, nonfat milk and lactose blocked the GII.13/21 P domain-glycan binding, which may explain the low prevalence of GII.13/21 viruses. Our data provide new insight into the host interactions and epidemiology of huNoVs, which would help in the control and prevention of NoV-associated diseases. IMPORTANCE Evidence from both phenotypic binding assay and structural study support the observed interactions of human noroviruses (huNoVs) with histo-blood group antigens (HBGAs) as receptors or attachment factors, affecting their host susceptibility. GII.13 and GII.21 genotypes form a unique genetic lineage that differs from the mainstream GII huNoVs in their unconventional glycan binding site. Unlike the previous findings that GII.13/21 genotypes recognize only Lea antigen, we found in this study that they can interact with a group of glycans with a common terminal β-Gal, including Lec, lactose, and mucin core 2. However, this wide glycan binding spectrum in a unique binding mode of the GII.13/21 huNoVs appears not to increase their prevalence, probably due to the existence of decoy glycan receptors in human gastrointestinal tract limiting their infection. Our findings shed light on the host interaction and epidemiology of huNoVs, which would impact the strategy of huNoV control and prevention.
Collapse
|
37
|
Lian Y, Wu S, Luo L, Lv B, Liao Q, Li Z, Rainey JJ, Hall AJ, Ran L. Epidemiology of Norovirus Outbreaks Reported to the Public Health Emergency Event Surveillance System, China, 2014⁻2017. Viruses 2019; 11:v11040342. [PMID: 30979014 PMCID: PMC6520956 DOI: 10.3390/v11040342] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 04/08/2019] [Accepted: 04/09/2019] [Indexed: 01/18/2023] Open
Abstract
We conducted a retrospective analysis of norovirus outbreaks reported to the National Public Health Emergency Event Surveillance System (PHEESS) in China from January 1, 2014 to December 31, 2017. We reviewed all acute gastroenteritis outbreaks (n = 692) submitted to PHEESS to identify the frequency, seasonality, geographic distribution, setting, and transmission mode of outbreaks due to norovirus. A total of 616 norovirus outbreaks resulting in 30,848 cases were reported. Among these outbreaks, 571 (93%) occurred in school settings including 239 (39%) in primary schools, 136 (22%) in childcare facilities, and 121 (20%) in secondary schools. The majority of outbreaks (63%) were due to person-to-person transmission, followed by multiple modes of transmission (11%), foodborne (5%) and waterborne (3%) transmission. These findings highlight the importance of improving hand hygiene and environmental disinfection in high-risk settings. Developing a standard and quantitative outbreak reporting structure could improve the usefulness of PHEESS for monitoring norovirus outbreaks.
Collapse
Affiliation(s)
- Yiyao Lian
- Division of Infectious Disease, Key Laboratory of Surveillance and Early-warning on Infectious Disease, Chinese Centre for Disease Control and Prevention, Beijing 102206, China.
| | - Shuyu Wu
- Division of Global Health Protection, Center for Global Health, U.S. Centers for Disease Control and Prevention, Beijing 100600, China.
| | - Li Luo
- Division of Infectious Disease, Key Laboratory of Surveillance and Early-warning on Infectious Disease, Chinese Centre for Disease Control and Prevention, Beijing 102206, China.
| | - Bin Lv
- Xiaogan Center for Disease Control and Prevention, Xiaogan 432000, China.
| | - Qiaohong Liao
- Division of Infectious Disease, Key Laboratory of Surveillance and Early-warning on Infectious Disease, Chinese Centre for Disease Control and Prevention, Beijing 102206, China.
| | - Zhongjie Li
- Division of Infectious Disease, Key Laboratory of Surveillance and Early-warning on Infectious Disease, Chinese Centre for Disease Control and Prevention, Beijing 102206, China.
| | - Jeanette J Rainey
- Division of Global Health Protection, Center for Global Health, U.S. Centers for Disease Control and Prevention, Beijing 100600, China.
| | - Aron J Hall
- Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, U.S. Centers for Disease Control and Prevention, Atlanta, GA 30333, USA.
| | - Lu Ran
- Division of Infectious Disease, Key Laboratory of Surveillance and Early-warning on Infectious Disease, Chinese Centre for Disease Control and Prevention, Beijing 102206, China.
| |
Collapse
|
38
|
Hoque SA, Thongprachum A, Takanashi S, Mostafa SM, Saito H, Anwar KS, Nomura A, Hoque SA, Begum R, Sultana UN, Hossain T, Khamrin P, Okitsu S, Hayakawa S, Ushijima H. Alarming Situation of Spreading Enteric Viruses Through Sewage Water in Dhaka City: Molecular Epidemiological Evidences. FOOD AND ENVIRONMENTAL VIROLOGY 2019; 11:65-75. [PMID: 30607905 DOI: 10.1007/s12560-018-09363-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 12/13/2018] [Indexed: 06/09/2023]
Abstract
Global burden of acute viral gastroenteritis remains high, particularly in developing countries including Bangladesh. Sewage water (SW) is an important node to monitor enteric pathogens both in the environment and among the population. Analysis of SW in Dhaka city deems crucially important because a large number of urban-city dwellers live in Dhaka city, the capital of Bangladesh, under a constant threat of precarious sewerage system. In this study, we collected raw SW from five locations of Dhaka city every month from June 2016 to May 2017. It was concentrated with polyethylene glycol (PEG) and investigated for three major enteric viruses, rotavirus A (RVA), norovirus GII (NoV GII) and adenovirus (AdV) using polymerase chain reaction (PCR). Most of these SW samples collected from both hospitals and non-hospital areas yielded enteric viruses: 76% samples were positive for AdV, followed by 53% NoV GII and 38% RVA. Viral load was determined as much as 1 × 107 copies/ml for RVA and 3.5 × 103 copies/ml for NoV GII. Importantly, NoV GII and AdV that can affect people of all ages were predominated during monsoon also when SW overflows and spreads over a wide and crowded area. Genotypes G1, G2, G3, G8, and G9 for RVA, GII.4 for NoV, and type 41 for AdV were detected representing the current profile of circulating genotypes in the population. This study provides the first evidence of distribution of major diarrheal viruses in SW in Dhaka city which is alarming showing grave risk of impending outbreaks through exposure.
Collapse
Affiliation(s)
- Sheikh Ariful Hoque
- Cell and Tissue Culture Laboratory, Centre for Advanced Research in Sciences (CARS), University of Dhaka, Dhaka, Bangladesh.
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo, Japan.
| | - Aksara Thongprachum
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo, Japan
- Faculty of Public Health, Chiang Mai University, Chiang Mai, Thailand
| | - Sayaka Takanashi
- Department of Developmental Medical Sciences, School of International Health, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Salwa Mohd Mostafa
- Cell and Tissue Culture Laboratory, Centre for Advanced Research in Sciences (CARS), University of Dhaka, Dhaka, Bangladesh
| | - Hiroyuki Saito
- Akita Prefectural Research Center for Public Health and Environment, Akita, Japan
| | - Kazi Selim Anwar
- Department of Infectious Diseases, International University of Health and Welfare (IUHW), Narita Campus, Narita, Chiba, Japan
| | - Akiko Nomura
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo, Japan
| | - Sk Azimul Hoque
- National Institute Neuroscience and Hospital, Agargaon, Dhaka, Bangladesh
| | - Rokeya Begum
- Genetic Engineering and Biotechnology Research Laboratory, Centre for Advanced Research in Sciences (CARS), University of Dhaka, Dhaka, Bangladesh
| | - Ummay Nasrin Sultana
- Cell and Tissue Culture Laboratory, Centre for Advanced Research in Sciences (CARS), University of Dhaka, Dhaka, Bangladesh
| | - Tania Hossain
- Cell and Tissue Culture Laboratory, Centre for Advanced Research in Sciences (CARS), University of Dhaka, Dhaka, Bangladesh
| | - Pattara Khamrin
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo, Japan
- Department of Microbiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Shoko Okitsu
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo, Japan
- Faculty of Public Health, Chiang Mai University, Chiang Mai, Thailand
| | - Satoshi Hayakawa
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo, Japan
| | - Hiroshi Ushijima
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo, Japan
- Faculty of Public Health, Chiang Mai University, Chiang Mai, Thailand
| |
Collapse
|
39
|
Qian Y, Song M, Jiang X, Xia M, Meller J, Tan M, Chen Y, Li X, Rao Z. Structural Adaptations of Norovirus GII.17/13/21 Lineage through Two Distinct Evolutionary Paths. J Virol 2019; 93:e01655-18. [PMID: 30333166 PMCID: PMC6288326 DOI: 10.1128/jvi.01655-18] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 09/22/2018] [Indexed: 11/20/2022] Open
Abstract
Human noroviruses (huNoVs), which cause epidemic acute gastroenteritis, recognize histo-blood group antigens (HBGAs) as host attachment factors affecting host susceptibility. HuNoVs are genetically diverse, containing at least 31 genotypes in the two major genogroups (genogroup I [GI] and GII). Three GII genotypes, GII genotype 17 (GII.17), GII.13, and GII.21, form a unique genetic lineage, in which the GII.17 genotype retains the conventional GII HBGA binding site (HBS), while the GII.13/21 genotypes acquire a completely new HBS. To understand the molecular bases behind these evolutionary changes, we solved the crystal structures of the HBGA binding protruding domains of (i) an early GII.17 variant (the 1978 variant) that does not bind or binds weakly to HBGAs, (ii) the new GII.17 variant (the 2014/15 variant) that binds A/B/H antigens strongly via an optimized GII HBS, and (iii) a GII.13 variant (the 2010 variant) that binds the Lewis a (Lea) antigen via the new HBS. These serial, high-resolution structural data enable a comprehensive structural comparison to understand the evolutionary changes of the GII.17/13/21 lineage, including the emergence of the new HBS of the GII.13/21 sublineage and the possible HBS optimization of the recent GII.17 variant for an enhanced HBGA binding ability. Our study elucidates the structural adaptations of the GII.17/13/21 lineage through distinct evolutionary paths, which may allow a theory explaining huNoV adaptations and evolutions to be put forward.IMPORTANCE Our understanding of the molecular bases behind the interplays between human noroviruses and their host glycan ligands, as well as their evolutionary changes over time with alterations in their host ligand binding capability and host susceptibility, remains limited. By solving the crystal structures of the glycan ligand binding protruding (P) domains with or without glycan ligands of three representative noroviruses of the GII.17/13/21 genetic lineage, we elucidated the molecular bases of the human norovirus-glycan interactions of this special genetic lineage. We present solid evidence on how noroviruses of this genetic lineage evolved via different evolutionary paths to (i) optimize their glycan binding site for higher glycan binding function and (ii) acquire a completely new glycan binding site for new ligands. Our data shed light on the mechanism of the structural adaptations of human noroviruses through different evolutionary paths, facilitating our understanding of human norovirus adaptations, evolutions, and epidemiology.
Collapse
Affiliation(s)
- Ying Qian
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Mohan Song
- College of Life Science, Nankai University, Tianjin, China
| | - Xi Jiang
- Division of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Ming Xia
- Division of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Jarek Meller
- Division of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Environmental Health, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Ming Tan
- Division of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Yutao Chen
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Xuemei Li
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Zihe Rao
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
40
|
Malm M, Tamminen K, Vesikari T, Blazevic V. Norovirus GII.17 Virus-Like Particles Bind to Different Histo-Blood Group Antigens and Cross-React with Genogroup II-Specific Mouse Sera. Viral Immunol 2018; 31:649-657. [DOI: 10.1089/vim.2018.0115] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
Affiliation(s)
- Maria Malm
- Vaccine Research Center, Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
| | - Kirsi Tamminen
- Vaccine Research Center, Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
| | - Timo Vesikari
- Vaccine Research Center, Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
| | - Vesna Blazevic
- Vaccine Research Center, Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
| |
Collapse
|
41
|
Pang LL, Wang MX, Sun XM, Yuan Y, Qing Y, Xin Y, Zhang JY, Li DD, Duan ZJ. Glycan binding patterns of human rotavirus P[10] VP8* protein. Virol J 2018; 15:161. [PMID: 30340611 PMCID: PMC6195756 DOI: 10.1186/s12985-018-1065-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 09/25/2018] [Indexed: 12/19/2022] Open
Abstract
Background Rotaviruses (RVs) are a major cause of acute children gastroenteritis. The rotavirus P [10] belongs to P[I] genogroup of group A rotaviruses that mainly infect animals, while the rotavirus P [10] was mainly identified from human infection. The rotavirus P [10] is an unusual genotype and the recognition pattern of cellular receptors remains unclear. Methods We expressed and purified the RV P [10] VP8* protein and investigated the saliva and oligosaccharide binding profiles of the protein. A homology model of the P [10] VP8* core protein was built and the superimposition structural analysis of P [10] VP8* protein on P [19] VP8* in complex with mucin core 2 was performed to explore the possible docking structural basis of P [10] VP8* and mucin cores. Results Our data showed that rotavirus P [10] VP8* protein bound to all ABO secretor and non-secretor saliva. The rotavirus P [10] could bind strongly to mucin core 2 and weakly to mucin core 4. The homology modeling indicated that RV P [10] VP8* binds to mucin core 2 using a potential glycan binding site that is the same to P [19] VP8* belonging to P[II] genogroup. Conclusion Our results suggested an interaction of rotavirus P [10] VP8* protein with mucin core 2 and mucin core 4. These findings offer potential for elucidating the mechanism of RV A host specificity, evolution and epidemiology.
Collapse
Affiliation(s)
- Li-Li Pang
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206, China.,Key Laboratory of Medical Virology and Viral Diseases, Ministry of Health of the People's Republic of China, Beijing, 102206, China
| | - Meng-Xuan Wang
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206, China.,Department of Food and Chemical Engineering, Lushan College of Guangxi University of Science and Technology, Liuzhou, 545616, Guangxi, China
| | - Xiao-Man Sun
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206, China.,Key Laboratory of Medical Virology and Viral Diseases, Ministry of Health of the People's Republic of China, Beijing, 102206, China
| | - Yue Yuan
- Basic Medical College, Inner Mongolia Medical University, Hohhot, 010000, China
| | - Yu Qing
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206, China.,Department of Food and Chemical Engineering, Lushan College of Guangxi University of Science and Technology, Liuzhou, 545616, Guangxi, China
| | - Yan Xin
- Basic Medical College, Inner Mongolia Medical University, Hohhot, 010000, China
| | - Jia-Yan Zhang
- Department of Food and Chemical Engineering, Lushan College of Guangxi University of Science and Technology, Liuzhou, 545616, Guangxi, China
| | - Dan-di Li
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206, China. .,Key Laboratory of Medical Virology and Viral Diseases, Ministry of Health of the People's Republic of China, Beijing, 102206, China.
| | - Zhao-Jun Duan
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206, China. .,Key Laboratory of Medical Virology and Viral Diseases, Ministry of Health of the People's Republic of China, Beijing, 102206, China.
| |
Collapse
|
42
|
Molecular epidemiology of noroviruses in children in South Greece, 2013‐2015. J Med Virol 2018; 90:1703-1711. [DOI: 10.1002/jmv.25251] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 05/16/2018] [Indexed: 12/21/2022]
|
43
|
Tohma K, Lepore CJ, Ford-Siltz LA, Parra GI. Evolutionary dynamics of non-GII genotype 4 (GII.4) noroviruses reveal limited and independent diversification of variants. J Gen Virol 2018; 99:1027-1035. [DOI: 10.1099/jgv.0.001088] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Affiliation(s)
- Kentaro Tohma
- Division of Viral Products, Food and Drug Administration, Silver Spring, MD, USA
| | - Cara J. Lepore
- Division of Viral Products, Food and Drug Administration, Silver Spring, MD, USA
| | - Lauren A. Ford-Siltz
- Division of Viral Products, Food and Drug Administration, Silver Spring, MD, USA
| | - Gabriel I. Parra
- Division of Viral Products, Food and Drug Administration, Silver Spring, MD, USA
| |
Collapse
|
44
|
He Z, Liu B, Tao Y, Li C, Xia M, Zhong W, Jiang X, Liu H, Tan M. Norovirus GII.17 Natural Infections in Rhesus Monkeys, China. Emerg Infect Dis 2018; 23:316-319. [PMID: 28102802 PMCID: PMC5324814 DOI: 10.3201/eid2302.161077] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Noroviruses are a leading viral cause of acute gastroenteritis among humans. During the 2014–15 epidemic season, norovirus GII.17 was detected in rhesus monkeys in China. Genetic, structural, and challenge studies revealed virus mutations and verified the infections. Thus, cross-species transmission may occur, and monkeys may be a virus reservoir.
Collapse
|
45
|
Complete Genome Sequence of a Novel Recombinant GII.P16-GII.1 Norovirus Associated with a Gastroenteritis Outbreak in Shandong Province, China, in 2017. GENOME ANNOUNCEMENTS 2018; 6:6/6/e01483-17. [PMID: 29439040 PMCID: PMC5805878 DOI: 10.1128/genomea.01483-17] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
We report here the complete genome sequence of a novel recombinant GII.P16-GII.1 norovirus identified from eight fecal samples collected during an acute gastroenteritis outbreak in Jinan, Shandong Province, China, in 2017. The virus had nucleotide identities of 99% and 91% in the ORF1 and ORF2 genes of related strains, respectively.
Collapse
|
46
|
Dai YC, Xia M, Huang Q, Tan M, Qin L, Zhuang YL, Long Y, Li JD, Jiang X, Zhang XF. Characterization of Antigenic Relatedness between GII.4 and GII.17 Noroviruses by Use of Serum Samples from Norovirus-Infected Patients. J Clin Microbiol 2017; 55:3366-3373. [PMID: 28904188 PMCID: PMC5703803 DOI: 10.1128/jcm.00865-17] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2017] [Accepted: 09/08/2017] [Indexed: 11/20/2022] Open
Abstract
A novel GII.17 norovirus variant caused major gastroenteritis epidemics in China in 2014 to 2016. To explore the host immune factors in selection of the emergence of this new variant, we characterized its antigenic relatedness with the GII.4 noroviruses that have dominated in China for decades. Through an enzyme-linked immunosorbent assay (ELISA) and a histo-blood group antigen (HBGA) blocking assay using sera from GII.4 and the GII.17 variant-infected patients, respectively, we observed limited cross-immune reactivity by the ELISA but little reactivity by the HBGA blocking assay between GII.4 norovirus and the new GII.17 variant. Our data suggest that, among other possible factors, GII.4-specific herd immunity had little role in the emergence of the new GII.17 variant. Thus, GII.17 may be an important active antigenic type or immunotype that needs to be considered for future vaccine strategies against human noroviruses.
Collapse
Affiliation(s)
- Ying-Chun Dai
- Department of Epidemiology, School of Public Health, Southern Medical University (Guangdong Provincial Key Laboratory of Tropical Disease Research), Guangzhou, Guangdong, China
| | - Ming Xia
- Division of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Qiong Huang
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, China
| | - Ming Tan
- Division of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Lin Qin
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, China
| | - Ya-Li Zhuang
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, China
| | - Yan Long
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Jian-Dong Li
- Department of Epidemiology, School of Public Health, Southern Medical University (Guangdong Provincial Key Laboratory of Tropical Disease Research), Guangzhou, Guangdong, China
| | - Xi Jiang
- Division of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Xu-Fu Zhang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
47
|
Lindesmith LC, Kocher JF, Donaldson EF, Debbink K, Mallory ML, Swann EW, Brewer-Jensen PD, Baric RS. Emergence of Novel Human Norovirus GII.17 Strains Correlates With Changes in Blockade Antibody Epitopes. J Infect Dis 2017; 216:1227-1234. [PMID: 28973354 PMCID: PMC5853573 DOI: 10.1093/infdis/jix385] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 07/25/2017] [Indexed: 12/20/2022] Open
Abstract
Background Human norovirus is a significant public health burden, with >30 genotypes causing endemic levels of disease and strains from the GII.4 genotype causing serial pandemics as the virus evolves new ligand binding and antigenicity features. During 2014-2015, genotype GII.17 cluster IIIb strains emerged as the leading cause of norovirus infection in select global locations. Comparison of capsid sequences indicates that GII.17 is evolving at previously defined GII.4 antibody epitopes. Methods Antigenicity of virus-like particles (VLPs) representative of clusters I, II, and IIIb GII.17 strains were compared by a surrogate neutralization assay based on antibody blockade of ligand binding. Results Sera from mice immunized with a single GII.17 VLP identified antigenic shifts between each cluster of GII.17 strains. Ligand binding of GII.17 cluster IIIb VLP was blocked only by antisera from mice immunized with cluster IIIb VLPs. Exchange of residues 393-396 from GII.17.2015 into GII.17.1978 ablated ligand binding and altered antigenicity, defining an important varying epitope in GII.17. Conclusions The capsid sequence changes in GII.17 strains result in loss of blockade antibody binding, indicating that viral evolution, specifically at residues 393-396, may have contributed to the emergence of cluster IIIb strains and the persistence of GII.17 in human populations.
Collapse
Affiliation(s)
- Lisa C Lindesmith
- Department of Epidemiology, University of North Carolina, Chapel Hill
| | - Jacob F Kocher
- Department of Epidemiology, University of North Carolina, Chapel Hill
| | - Eric F Donaldson
- Department of Epidemiology, University of North Carolina, Chapel Hill
| | - Kari Debbink
- Department of Natural Sciences, Bowie State University, Maryland
| | - Michael L Mallory
- Department of Epidemiology, University of North Carolina, Chapel Hill
| | - Excel W Swann
- Department of Epidemiology, University of North Carolina, Chapel Hill
| | | | - Ralph S Baric
- Department of Epidemiology, University of North Carolina, Chapel Hill,Correspondence: R. S. Baric, PhD, 3304 Hooker Research Center, 135 Dauer Dr, CB7435, School of Public Health, University of North Carolina–Chapel Hill, Chapel Hill, NC 27599 ()
| |
Collapse
|
48
|
Ao Y, Wang J, Ling H, He Y, Dong X, Wang X, Peng J, Zhang H, Jin M, Duan Z. Norovirus GII.P16/GII.2-Associated Gastroenteritis, China, 2016. Emerg Infect Dis 2017; 23:1172-1175. [PMID: 28430563 PMCID: PMC5512504 DOI: 10.3201/eid2307.170034] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
During October–December 2016, the number of norovirus outbreaks in China increased sharply from the same period during the previous 4 years. We identified a recombinant norovirus strain, GII.P16-GII.2, as the cause of 44 (79%) of the 56 outbreaks, signaling that this strain could replace the predominant GII.4 viruses.
Collapse
|