1
|
Cortés-Trigueros JA, Ossio A, Heredia N, Casillas-Vega N, García S, Merino-Mascorro JA. Norovirus GI.5 [P4]: first report of the rare norovirus recombinant variant in Northeastern Mexico and its global epidemiological context. Virus Genes 2025; 61:294-302. [PMID: 39985634 DOI: 10.1007/s11262-025-02144-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Accepted: 02/14/2025] [Indexed: 02/24/2025]
Abstract
Norovirus is the primary cause of acute gastroenteritis outbreaks, considerably impacting children under 5 years, followed by older adults and immunocompromised individuals. As an RNA virus, norovirus exhibits high genetic variability, driven by recombination events at the ORF1-ORF2 junction. This study reports the first detection of the rare norovirus GI.5 [P4] variant in Northeastern Mexico, identified in a single positive isolate (MTY0115; GenBank: PQ369661) from a sample group of 386 individuals, with a prevalence of 0.25%. Notably, norovirus GII was not detected. Phylogenetic analysis of the partial RdRp/VP1 region revealed clustering with global GI.5 [P4] sequences, revealing evolutionary relationships with isolates from Asia, Europe, and America. A recombination event was identified at position 5307 (breakpoint based on reference sequences of GI.5 [P5] and GI.4 [P4]) within ORF1, with genetic inheritance from a GI.5 [P5] isolate from Moscow, Russia, and a GI.4 [P4] isolate from France. Typing classification through sequencing of overlapping ORF1 and ORF2 regions is valuable for understanding genomic variations and their epidemiological impact on at-risk and non-risk populations.
Collapse
Affiliation(s)
- José Antonio Cortés-Trigueros
- Laboratorio de Bioquímica y Genética de Microorganismos, Departamento de Microbiología, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, 66455, San Nicolás de los Garza, N.L., México
| | - Axel Ossio
- Laboratorio de Bioquímica y Genética de Microorganismos, Departamento de Microbiología, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, 66455, San Nicolás de los Garza, N.L., México
| | - Norma Heredia
- Laboratorio de Bioquímica y Genética de Microorganismos, Departamento de Microbiología, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, 66455, San Nicolás de los Garza, N.L., México
| | - Néstor Casillas-Vega
- Departamento de Patología Clínica, Hospital Universitario Dr. José Eleuterio González - Facultad de Medicina, Universidad Autónoma de Nuevo León, 64460, Monterrey, N.L., Mexico
| | - Santos García
- Laboratorio de Bioquímica y Genética de Microorganismos, Departamento de Microbiología, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, 66455, San Nicolás de los Garza, N.L., México
| | - Jose Angel Merino-Mascorro
- Laboratorio de Bioquímica y Genética de Microorganismos, Departamento de Microbiología, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, 66455, San Nicolás de los Garza, N.L., México.
| |
Collapse
|
2
|
Purushotham JN, Lutz HL, Parker E, Andersen KG. Immunological drivers of zoonotic virus emergence, evolution, and endemicity. Immunity 2025; 58:784-796. [PMID: 40168990 PMCID: PMC11981831 DOI: 10.1016/j.immuni.2025.03.014] [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: 10/10/2024] [Revised: 03/11/2025] [Accepted: 03/12/2025] [Indexed: 04/03/2025]
Abstract
The disruption of natural ecosystems caused by climate change and human activity is amplifying the risk of zoonotic spillover, presenting a growing global health threat. In the past two decades, the emergence of multiple zoonotic viruses has exposed critical gaps in our ability to predict epidemic trajectories and implement effective interventions. RNA viruses, in particular, are challenging to control due to their high mutation rates and ability to adapt and evade immune defenses. To better prepare for future outbreaks, it is vital that we deepen our understanding of the factors driving viral emergence, transmission, and persistence in human populations. Specifically, deciphering the interactions between antibody-mediated immunity and viral evolution will be key. In this perspective, we explore these dynamic relationships and highlight research priorities that may guide the development of more effective strategies to mitigate the impact of emerging infectious diseases.
Collapse
Affiliation(s)
- Jyothi N Purushotham
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA; Scripps Research Translational Institute, La Jolla, CA, USA
| | - Holly L Lutz
- Denver Museum of Nature and Science, Denver, CO, USA
| | - Edyth Parker
- The Institute of Genomics and Global Health (IGH), Redeemer's University, Ede, Osun, Nigeria
| | - Kristian G Andersen
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA; Scripps Research Translational Institute, La Jolla, CA, USA.
| |
Collapse
|
3
|
Park J, Lindesmith LC, Olia AS, Costantini VP, Brewer-Jensen PD, Mallory ML, Kelley CE, Satterwhite E, Longo V, Tsybovsky Y, Stephens T, Marchioni J, Martins CA, Huang Y, Chaudhary R, Zweigart M, May SR, Reyes Y, Flitter B, Vinjé J, Tucker SN, Ippolito GC, Lavinder JJ, Snijder J, Kwong PD, Georgiou G, Baric RS. Broadly neutralizing antibodies targeting pandemic GII.4 variants or seven GII genotypes of human norovirus. Sci Transl Med 2025; 17:eads8214. [PMID: 40043137 DOI: 10.1126/scitranslmed.ads8214] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Accepted: 01/22/2025] [Indexed: 04/25/2025]
Abstract
Human norovirus causes more than 700 million illnesses annually. Extensive genetic diversity and a paucity of information on conserved neutralizing epitopes pose major obstacles to the design of broadly protective norovirus immunogens. Here, we used high-resolution liquid chromatography-tandem mass spectrometry (LC-MS/MS)-driven proteomics to quantitatively characterize the circulating serum IgG repertoire before and after immunization with an experimental monovalent norovirus GII.4 VP1 capsid-encoding adenoviral vaccine. Two participants were specifically selected on the basis of the breadth of serum neutralization responses either across GII.4 variants (participant A) or across GII genotypes (participant B). In participant A, vaccination back-boosted highly abundant serum antibody clonotypes targeting epitopes conserved among rapidly evolving GII.4 variants spanning from a strain identified in 1987 to a strain identified in 2019. In participant B, we identified a recall response consisting of broadly neutralizing monoclonal antibodies with remarkable cross-GII ligand-binding blockade (blocking ≥ seven GII genotypes) and virus neutralization breadth. The cocrystal structure of one of these antibodies, VX22, in complex with the VP1 capsid protruding (P) domain revealed a highly conserved epitope (residues 479 to 484 and 509 to 513) within two lateral loops of the P1 subdomain. Antibody evolutionary trajectory analysis further revealed that VX22 had originally evolved from an early heterologous infection, likely by a GII.12 strain. Together, our study demonstrates that norovirus human monoclonal antibodies with broad GII.4 potency and cross-GII breadth can be boosted in serum after immunization with an adenoviral vector-based vaccine, findings that may guide the design of immunogens for broadly protective norovirus vaccines.
Collapse
Affiliation(s)
- Juyeon Park
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
- Department of Chemical Engineering, University of Texas at Austin, Austin, TX 78712, USA
| | - Lisa C Lindesmith
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Adam S Olia
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Veronica P Costantini
- Division of Viral Diseases, Centers for Disease Control and Prevention, Atlanta, GA 30329, USA
| | - Paul D Brewer-Jensen
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Michael L Mallory
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Cynthia E Kelley
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute of Pharmaceutical Sciences, Utrecht University, 3584 CH, Utrecht, Netherlands
| | - Ed Satterwhite
- Department of Chemical Engineering, University of Texas at Austin, Austin, TX 78712, USA
| | - Victoria Longo
- Department of Chemical Engineering, University of Texas at Austin, Austin, TX 78712, USA
| | - Yaroslav Tsybovsky
- Electron Microscopy Laboratory, Cancer Research Technology Program, Leidos Biomedical Research, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Tyler Stephens
- Electron Microscopy Laboratory, Cancer Research Technology Program, Leidos Biomedical Research, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Jeffrey Marchioni
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX 78712, USA
| | - Christina A Martins
- Department of Chemical Engineering, University of Texas at Austin, Austin, TX 78712, USA
| | - Yimin Huang
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - Ridhi Chaudhary
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Mark Zweigart
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Samantha R May
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Yaoska Reyes
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC 27599, USA
| | | | - Jan Vinjé
- Division of Viral Diseases, Centers for Disease Control and Prevention, Atlanta, GA 30329, USA
| | | | - Gregory C Ippolito
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
- Department of Oncology, University of Texas at Austin, Austin, TX 78712, USA
| | - Jason J Lavinder
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
- Department of Chemical Engineering, University of Texas at Austin, Austin, TX 78712, USA
| | - Joost Snijder
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute of Pharmaceutical Sciences, Utrecht University, 3584 CH, Utrecht, Netherlands
| | - Peter D Kwong
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - George Georgiou
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
- Department of Chemical Engineering, University of Texas at Austin, Austin, TX 78712, USA
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX 78712, USA
- Department of Oncology, University of Texas at Austin, Austin, TX 78712, USA
| | - Ralph S Baric
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC 27599, USA
| |
Collapse
|
4
|
Prasad BVV, Atmar RL, Ramani S, Palzkill T, Song Y, Crawford SE, Estes MK. Norovirus replication, host interactions and vaccine advances. Nat Rev Microbiol 2025:10.1038/s41579-024-01144-9. [PMID: 39824927 DOI: 10.1038/s41579-024-01144-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/12/2024] [Indexed: 01/20/2025]
Abstract
Human noroviruses (HuNoVs) are the leading cause of acute gastroenteritis worldwide in all age groups and cause significant disease and economic burden globally. To date, no approved vaccines or antiviral therapies are available to treat or prevent HuNoV illness. Several candidate vaccines are in clinical trials, although potential barriers to successful development must be overcome. Recently, significant advances have been made in understanding HuNoV biology owing to breakthroughs in virus cultivation using human intestinal tissue-derived organoid (or enteroid) cultures, advances in structural biology technology combined with epitope mapping and increased metagenomic sequencing. New and unexpected strain-specific differences in pandemic versus non-pandemic virus structures, replication properties and virus-host interactions, including host factors required for susceptibility to infection and pathogenesis, are discussed.
Collapse
Affiliation(s)
- B V Venkataram Prasad
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Robert L Atmar
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Sasirekha Ramani
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Timothy Palzkill
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Yongcheng Song
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, TX, USA
| | - Sue E Crawford
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Mary K Estes
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA.
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
5
|
Lewis CB, Sherry L, Conley MJ, Nakashima M, Akbar S, Govindan N, Hosie MJ, Bhella D. Conformational Flexibility in Capsids Encoded by the Caliciviridae. Viruses 2024; 16:1835. [PMID: 39772145 PMCID: PMC11680396 DOI: 10.3390/v16121835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 11/06/2024] [Accepted: 11/22/2024] [Indexed: 01/11/2025] Open
Abstract
Caliciviruses are a diverse group of non-enveloped, positive-sense RNA viruses with a wide range of hosts and transmission routes. Norovirus is the most well-known member of the Caliciviridae; the acute gastroenteritis caused by human norovirus (HuNoV), for example, frequently results in closures of hospital wards and schools during the winter months. One area of calicivirus biology that has gained increasing attention over the past decade is the conformational flexibility exhibited by the protruding (P) domains of the major capsid protein VP1. This was observed in structure analyses of capsids encoded by many species and is often a consequence of environmental cues such as metal ions, changes to pH, or receptor/co-factor engagement. This review summarises the current understanding of P-domain flexibility, discussing the role this region plays in caliciviral infection and immune evasion, and highlighting potential avenues for further investigation.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - David Bhella
- MRC—University of Glasgow Centre for Virus Research, Garscube Campus, 464 Bearsden Road, Glasgow G61 1QH, UK; (C.B.L.); (L.S.); (M.J.C.); (M.N.); (S.A.); (M.J.H.)
| |
Collapse
|
6
|
Mackin SR, Sariol A, Diamond MS. Antibody-mediated control mechanisms of viral infections. Immunol Rev 2024; 328:205-220. [PMID: 39162394 PMCID: PMC11661935 DOI: 10.1111/imr.13383] [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] [Indexed: 08/21/2024]
Abstract
Antibodies generated after vaccination or natural pathogen exposure are essential mediators of protection against many infections. Most studies with viruses have focused on antibody neutralization, in which protection is conferred by the fragment antigen binding region (Fab) through targeting of different steps in the viral lifecycle including attachment, internalization, fusion, and egress. Beyond neutralization, the fragment crystallizable (Fc) region of antibodies can integrate innate and adaptive immune responses by engaging complement components and distinct Fc gamma receptors (FcγR) on different host immune cells. In this review, we discuss recent advances in our understanding of antibody neutralization and Fc effector functions, and the assays used to measure them. Additionally, we describe the contexts in which these mechanisms are associated with protection against viruses and highlight how Fc-FcγR interactions can improve the potency of antibody-based therapies.
Collapse
Affiliation(s)
- Samantha R. Mackin
- Department of Medicine, Washington University School of Medicine, MO 63110, USA
- Department of Pathology & Immunology and Center for Genome Sciences, Lab & Genomic Medicine, Washington University School of Medicine, MO 63110, USA
| | - Alan Sariol
- Department of Medicine, Washington University School of Medicine, MO 63110, USA
| | - Michael S. Diamond
- Department of Medicine, Washington University School of Medicine, MO 63110, USA
- Department of Pathology & Immunology and Center for Genome Sciences, Lab & Genomic Medicine, Washington University School of Medicine, MO 63110, USA
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO
- Andrew M. and Jane M. Bursky the Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO 63110, USA
- Center for Vaccines and Immunity to Microbial Pathogens, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
7
|
Atochina-Vasserman EN, Lindesmith LC, Mirabelli C, Ona NA, Reagan EK, Brewer-Jensen PD, Mercado-Lopez X, Shahnawaz H, Meshanni JA, Baboo I, Mallory ML, Zweigart MR, May SR, Mui BL, Tam YK, Wobus CE, Baric RS, Weissman D. Bivalent norovirus mRNA vaccine elicits cellular and humoral responses protecting human enteroids from GII.4 infection. NPJ Vaccines 2024; 9:182. [PMID: 39353926 PMCID: PMC11445234 DOI: 10.1038/s41541-024-00976-z] [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: 04/18/2024] [Accepted: 09/19/2024] [Indexed: 10/03/2024] Open
Abstract
Nucleoside-modified mRNA-LNP vaccines have revolutionized vaccine development against infectious pathogens due to their ability to elicit potent humoral and cellular immune responses. In this article, we present the results of the first norovirus vaccine candidate employing mRNA-LNP platform technology. The mRNA-LNP bivalent vaccine encoding the major capsid protein VP1 from GI.1 and GII.4 of human norovirus, generated high levels of neutralizing antibodies, robust cellular responses, and effectively protected human enteroids from infection by the most prevalent genotype (GII.4). These results serve as a proof of concept, demonstrating that a modified-nucleoside mRNA-LNP vaccine based on norovirus VP1 sequences can stimulate an immunogenic response in vivo and generates neutralizing antibodies capable of preventing viral infection in models of human gastrointestinal tract infection.
Collapse
Affiliation(s)
- Elena N Atochina-Vasserman
- Institue for RNA Innovation, the University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| | - Lisa C Lindesmith
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Carmen Mirabelli
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI, USA
| | - Nathan A Ona
- Institue for RNA Innovation, the University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Erin K Reagan
- Institue for RNA Innovation, the University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Paul D Brewer-Jensen
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Xiomara Mercado-Lopez
- Institue for RNA Innovation, the University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Hamna Shahnawaz
- Institue for RNA Innovation, the University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Jaclynn A Meshanni
- Institue for RNA Innovation, the University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Ishana Baboo
- Institue for RNA Innovation, the University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Michael L Mallory
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Mark R Zweigart
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Samantha R May
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | | - Ying K Tam
- Acuitas Therapeutics Inc, Vancouver, B.C., Canada
| | - Christiane E Wobus
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI, USA
| | - Ralph S Baric
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Drew Weissman
- Institue for RNA Innovation, the University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| |
Collapse
|
8
|
Ramani S, Javornik Cregeen SJ, Surathu A, Neill FH, Muzny DM, Doddapaneni H, Menon VK, Hoffman KL, Ross MC, Metcalf G, Opekun AR, Graham DY, Gibbs RA, Petrosino JF, Estes MK, Atmar RL. INTRA- AND INTER-HOST EVOLUTION OF HUMAN NOROVIRUS IN HEALTHY ADULTS. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.05.30.542907. [PMID: 39282326 PMCID: PMC11398385 DOI: 10.1101/2023.05.30.542907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/20/2024]
Abstract
Background Human noroviruses are a leading cause of acute and sporadic gastroenteritis worldwide. The evolution of human noroviruses in immunocompromised persons has been evaluated in many studies. Much less is known about the evolutionary dynamics of human norovirus in healthy adults. Methods We used sequential samples collected from a controlled human infection study with GI.1/Norwalk/US/68 virus to evaluate intra- and inter-host evolution of a human norovirus in healthy adults. Up to 12 samples from day 1 to day 56 post-challenge were sequenced using a norovirus-specific capture probe method. Results Complete genomes were assembled, even in samples that were below the limit of detection of standard RT-qPCR assays, up to 28 days post-challenge. Analysis of 123 complete genomes showed changes in the GI.1 genome in all persons, but there were no conserved changes across all persons. Single nucleotide variants resulting in non-synonymous amino acid changes were observed in all proteins, with the capsid VP1 and nonstructural protein NS3 having the largest numbers of changes. Conclusions These data highlight the potential of a new capture-based sequencing approach to assemble human norovirus genomes with high sensitivity and demonstrate limited conserved immune pressure-driven evolution of GI.1 virus in healthy adults.
Collapse
Affiliation(s)
- Sasirekha Ramani
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Sara J. Javornik Cregeen
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, 77030, USA
- The Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Anil Surathu
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, 77030, USA
- The Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Frederick H. Neill
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Donna M. Muzny
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Harsha Doddapaneni
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Vipin K. Menon
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Kristi L. Hoffman
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, 77030, USA
- The Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Matthew C. Ross
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, 77030, USA
- The Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Ginger Metcalf
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Antone R. Opekun
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030
| | - David Y. Graham
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030
| | - Richard A. Gibbs
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Joseph F. Petrosino
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, 77030, USA
- The Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Mary K. Estes
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, 77030, USA
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030
| | - Robert L. Atmar
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, 77030, USA
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030
| |
Collapse
|
9
|
Hu C, Yang S, Li S, Liu X, Liu Y, Chen Z, Chen H, Li S, He N, Cui H, Deng Y. Viral aptamer screening and aptamer-based biosensors for virus detection: A review. Int J Biol Macromol 2024; 276:133935. [PMID: 39029851 DOI: 10.1016/j.ijbiomac.2024.133935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 07/14/2024] [Accepted: 07/15/2024] [Indexed: 07/21/2024]
Abstract
Virus-induced infectious diseases have a detrimental effect on public health and exert significant influence on the global economy. Therefore, the rapid and accurate detection of viruses is crucial for effectively preventing and diagnosing infections. Aptamer-based detection technologies have attracted researchers' attention as promising solutions. Aptamers, small single-stranded DNA or RNA screened via systematic evolution of ligands by exponential enrichment (SELEX), possess a high affinity towards their target molecules. Numerous aptamers targeting viral marker proteins or virions have been developed and widely employed in aptamer-based biosensors (aptasensor) for virus detection. This review introduces SELEX schemes for screening aptamers and discusses distinctive SELEX strategies designed explicitly for viral targets. Furthermore, recent advances in aptamer-based biosensing methods for detecting common viruses using different virus-specific aptamers are summarized. Finally, limitations and prospects associated with developing of aptamer-based biosensors are discussed.
Collapse
Affiliation(s)
- Changchun Hu
- Hunan Key Laboratory of Biomedical Nanomaterials and Devices, Hunan University of Technology, Zhuzhou, Hunan 412007, China; School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Shuting Yang
- Hunan Key Laboratory of Biomedical Nanomaterials and Devices, Hunan University of Technology, Zhuzhou, Hunan 412007, China
| | - Shuo Li
- Hunan Key Laboratory of Biomedical Nanomaterials and Devices, Hunan University of Technology, Zhuzhou, Hunan 412007, China
| | - Xueying Liu
- Hunan Key Laboratory of Biomedical Nanomaterials and Devices, Hunan University of Technology, Zhuzhou, Hunan 412007, China
| | - Yuan Liu
- Institute for Future Sciences, University of South China, Changsha, Hunan 410000, China; Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Zhu Chen
- Hunan Key Laboratory of Biomedical Nanomaterials and Devices, Hunan University of Technology, Zhuzhou, Hunan 412007, China
| | - Hui Chen
- Hunan Key Laboratory of Biomedical Nanomaterials and Devices, Hunan University of Technology, Zhuzhou, Hunan 412007, China
| | - Song Li
- Hunan Key Laboratory of Biomedical Nanomaterials and Devices, Hunan University of Technology, Zhuzhou, Hunan 412007, China
| | - Nongyue He
- Hunan Key Laboratory of Biomedical Nanomaterials and Devices, Hunan University of Technology, Zhuzhou, Hunan 412007, China
| | - Haipo Cui
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Yan Deng
- Hunan Key Laboratory of Biomedical Nanomaterials and Devices, Hunan University of Technology, Zhuzhou, Hunan 412007, China; Institute for Future Sciences, University of South China, Changsha, Hunan 410000, China; Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China.
| |
Collapse
|
10
|
Liu J, Wang Z, Ma J, Ji S, Huo Y. Identification of a norovirus GII-specific antigenic epitope. Arch Virol 2024; 169:131. [PMID: 38819530 DOI: 10.1007/s00705-024-06060-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 04/17/2024] [Indexed: 06/01/2024]
Abstract
Noroviruses (NoVs) are the chief cause of acute viral gastroenteritis worldwide. By employing the major capsid protein VP1 of a GII.6 NoV strain as an immunogen, we generated two monoclonal antibodies (mAbs) with wide-spectrum binding activities against NoV genogroup II (GII) VP1 proteins. One mAb (10G7) could bind to native and denatured GII-specific VP1 proteins. The other mAb (10F2) could bind to all tested native GII VP1 proteins, but not to denatured GII.3, GII.4, GII.7, or GII.17 VP1 proteins. Using GII.6/GII.4 fusion proteins, the mAb 10F2 binding region was confirmed to be located in the C-terminal P1 domain. An enzyme-linked immunosorbent assay based on peptides covering the P domain did not detect any binding. Using a panel of VP1 proteins with swapped regions, deletions, and mutations, the mAb 10F2 binding region was determined to be located between residues 496 and 513. However, the residue(s) responsible for its varied binding affinity for different denatured GII VP1 proteins remain to be identified. In summary, two NoV GII-specific cross-reactive mAbs were generated, and their binding regions were determined. Our results might facilitate the detection and immunogenic study of NoVs.
Collapse
Affiliation(s)
- Jinjin Liu
- Affiliated Infectious Diseases Hospital of Zhengzhou University, Henan Infectious Diseases Hospital, The Sixth People's Hospital of Zhengzhou, Center for Translational Medicine, Zhengzhou, China
| | - Zhanzheng Wang
- Zhengzhou Cell to Antibody & antigen Biotechnology, Zhengzhou, China
| | - Jie Ma
- Affiliated Infectious Diseases Hospital of Zhengzhou University, Henan Infectious Diseases Hospital, The Sixth People's Hospital of Zhengzhou, Center for Translational Medicine, Zhengzhou, China
| | - Shaoping Ji
- Medical college, Cell Signal Transduction Laboratory, Henan University, Henan, China
| | - Yuqi Huo
- Affiliated Infectious Diseases Hospital of Zhengzhou University, Henan Infectious Diseases Hospital, The Sixth People's Hospital of Zhengzhou, Center for Translational Medicine, Zhengzhou, China.
| |
Collapse
|
11
|
Carlson KB, Dilley A, O'Grady T, Johnson JA, Lopman B, Viscidi E. A narrative review of norovirus epidemiology, biology, and challenges to vaccine development. NPJ Vaccines 2024; 9:94. [PMID: 38811605 PMCID: PMC11137017 DOI: 10.1038/s41541-024-00884-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 05/07/2024] [Indexed: 05/31/2024] Open
Abstract
Norovirus is a leading cause of acute gastroenteritis (AGE) globally. AGE resulting from norovirus causes significant morbidity and mortality in countries of all income levels, particularly among young children and older adults. Prevention of norovirus AGE represents a unique challenge as the virus is genetically diverse with multiple genogroups and genotypes cocirculating globally and causing disease in humans. Variants of the GII.4 genotype are typically the most common genotype, and other genotypes cause varying amounts of disease year-to-year, with GII.2, GII.3, and GII.6 most prevalent in recent years. Noroviruses are primarily transmitted via the fecal-oral route and only a very small number of virions are required for infection, which makes outbreaks of norovirus extremely difficult to control when they occur. Settings like long-term care facilities, daycares, and hospitals are at high risk of outbreaks and can have very high attack rates resulting in substantial costs and disease burden. Severe cases of norovirus AGE are most common in vulnerable patient populations, such as infants, the elderly, and immunocompromised individuals, with available treatments limited to rehydration therapies and supportive care. To date, there are no FDA-approved norovirus vaccines; however, several candidates are currently in development. Given the substantial human and economic burden associated with norovirus AGE, a vaccine to prevent morbidity and mortality and protect vulnerable populations could have a significant impact on global public health.
Collapse
Affiliation(s)
| | - Anne Dilley
- Epidemiologic Research & Methods, LLC, Atlanta, GA, USA
| | | | - Jordan A Johnson
- Epidemiologic Research & Methods, LLC, Atlanta, GA, USA
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Ben Lopman
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | | |
Collapse
|
12
|
Tohma K, Landivar M, Ford-Siltz LA, Pilewski KA, Kendra JA, Niendorf S, Parra GI. Antigenic Characterization of Novel Human Norovirus GII.4 Variants San Francisco 2017 and Hong Kong 2019. Emerg Infect Dis 2024; 30:1026-1029. [PMID: 38666659 PMCID: PMC11060466 DOI: 10.3201/eid3005.231694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2024] Open
Abstract
Norovirus is a major cause of acute gastroenteritis; GII.4 is the predominant strain in humans. Recently, 2 new GII.4 variants, Hong Kong 2019 and San Francisco 2017, were reported. Characterization using GII.4 monoclonal antibodies and serum demonstrated different antigenic profiles for the new variants compared with historical variants.
Collapse
|
13
|
Wasielewski VV, Itani TM, Zakharova YA, Semenov AV. Current trends and new approaches for human norovirus replication in cell culture: a literature review. Arch Virol 2024; 169:71. [PMID: 38459228 DOI: 10.1007/s00705-024-05999-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 01/21/2024] [Indexed: 03/10/2024]
Abstract
Human norovirus (HuNoV) is one of the world's leading causes of acute gastroenteritis. At present, effective reproduction of the virus in cell cultures remains a challenge for virologists, as there is a lack of a permissive cell line that allows the entire viral life cycle to be reproduced. This is a barrier to the study of the HuNoV life cycle, its tropism, and virus-host interactions. It is also a major hurdle for the development of viral detection platforms, and ultimately for the development of therapeutics. The lack of an inexpensive, technically simple, and easily implemented cultivation method also negatively affects our ability to evaluate the efficacy of a variety of control measures (disinfectants, food processes) for human norovirus. In the process of monitoring this pathogen, it is necessary to detect infectious viral particles in water, food, and other environmental samples. Therefore, improvement of in vitro replication of HuNoV is still needed. In this review, we discuss current trends and new approaches to HuNoV replication in cell culture. We highlight ways in which previous research on HuNoV and other noroviruses has guided and influenced the development of new HuNoV culture systems and discuss the improvement of in vitro replication of HuNoV.
Collapse
Affiliation(s)
- Valentin V Wasielewski
- Federal Service for Surveillance on Consumer Rights Protection and Human Wellbeing, Federal Scientific Research Institute of Viral Infections «Virome», Ekaterinburg, 620030, Russian Federation
| | - Tarek M Itani
- Federal Service for Surveillance on Consumer Rights Protection and Human Wellbeing, Federal Scientific Research Institute of Viral Infections «Virome», Ekaterinburg, 620030, Russian Federation.
| | - Yuliya A Zakharova
- Institute of Disinfectology of the F.F. Erisman Federal Scientific Centre of Hygiene Rospotrebnadzor, Mosсow, Russian Federation
| | - Aleksandr V Semenov
- Federal Service for Surveillance on Consumer Rights Protection and Human Wellbeing, Federal Scientific Research Institute of Viral Infections «Virome», Ekaterinburg, 620030, Russian Federation
- Ural Federal University named after the First President of Russia B.N. Yeltsin, Ekaterinburg, Russian Federation
| |
Collapse
|
14
|
Bonura F, Filizzolo C, Pizzo M, Sanfilippo GL, Cacioppo F, Palazzotto E, Di Bernardo F, Collura A, Martella V, De Grazia S, Giammanco GM. Biological Specimen Banking as a Time Capsule to Explore the Temporal Dynamics of Norovirus Epidemiology. Viruses 2023; 15:2303. [PMID: 38140544 PMCID: PMC10747129 DOI: 10.3390/v15122303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 10/20/2023] [Accepted: 11/22/2023] [Indexed: 12/24/2023] Open
Abstract
Norovirus is recognised as a major cause of epidemic and sporadic acute gastroenteritis (AGE) in all age groups. Information on the genetic diversity of the noroviruses circulating in the 1980s and 1990s, before the development and adoption of dedicated molecular assays, is limited compared with the last decades. Between 1986 and 2020, uninterrupted viral surveillance was conducted in symptomatic children hospitalized with AGE in Palermo, Italy, providing a unique time capsule for exploring the epidemiological and evolutionary dynamics of enteric viruses. A total of 8433 stool samples were tested using real-time RT-PCR. All samples were stored at -20 or -80 °C until processing. In this 35-year long time span, noroviruses of genogroup II (GII) were detected in 15.6% of AGE requiring hospitalization, whilst GI noroviruses were detected in 1.4% of AGE. Overall, the predominant norovirus capsid (Cap) genotype was GII.4 (60.8%), followed by GII.3 (13.3%) and GII.2 (12.4%). Temporal replacement of the GII.4 Cap variants associated with different polymerase (Pol) types were observed over the study period. The chronology of emergence and circulation of the different GII.4 variants were consistent with data available in the literature. Also, for GII.3 and GII.2 NoVs, the circulation of different lineages/strains, differing in either the Cap or Pol genes or in both, was observed. This long-term study revealed the ability of noroviruses to continuously and rapidly modify their genomic makeup and highlights the importance of surveillance activities in vaccine design.
Collapse
Affiliation(s)
- Floriana Bonura
- Dipartimento di Scienze per la Promozione della Salute, Materno-Infantile, di Medicina Interna e Specialistica di Eccellenza “G. D’Alessandro”, Università di Palermo, Via del Vespro 133, 90127 Palermo, Italy; (C.F.); (M.P.); (G.L.S.); (F.C.); (E.P.); (S.D.G.); (G.M.G.)
| | - Chiara Filizzolo
- Dipartimento di Scienze per la Promozione della Salute, Materno-Infantile, di Medicina Interna e Specialistica di Eccellenza “G. D’Alessandro”, Università di Palermo, Via del Vespro 133, 90127 Palermo, Italy; (C.F.); (M.P.); (G.L.S.); (F.C.); (E.P.); (S.D.G.); (G.M.G.)
| | - Mariangela Pizzo
- Dipartimento di Scienze per la Promozione della Salute, Materno-Infantile, di Medicina Interna e Specialistica di Eccellenza “G. D’Alessandro”, Università di Palermo, Via del Vespro 133, 90127 Palermo, Italy; (C.F.); (M.P.); (G.L.S.); (F.C.); (E.P.); (S.D.G.); (G.M.G.)
| | - Giuseppa L. Sanfilippo
- Dipartimento di Scienze per la Promozione della Salute, Materno-Infantile, di Medicina Interna e Specialistica di Eccellenza “G. D’Alessandro”, Università di Palermo, Via del Vespro 133, 90127 Palermo, Italy; (C.F.); (M.P.); (G.L.S.); (F.C.); (E.P.); (S.D.G.); (G.M.G.)
| | - Federica Cacioppo
- Dipartimento di Scienze per la Promozione della Salute, Materno-Infantile, di Medicina Interna e Specialistica di Eccellenza “G. D’Alessandro”, Università di Palermo, Via del Vespro 133, 90127 Palermo, Italy; (C.F.); (M.P.); (G.L.S.); (F.C.); (E.P.); (S.D.G.); (G.M.G.)
| | - Emilia Palazzotto
- Dipartimento di Scienze per la Promozione della Salute, Materno-Infantile, di Medicina Interna e Specialistica di Eccellenza “G. D’Alessandro”, Università di Palermo, Via del Vespro 133, 90127 Palermo, Italy; (C.F.); (M.P.); (G.L.S.); (F.C.); (E.P.); (S.D.G.); (G.M.G.)
| | - Francesca Di Bernardo
- Unità Operativa di Microbiologia e Virologia, Ospedale Civico e di Cristina, ARNAS, 90129 Palermo, Italy; (F.D.B.); (A.C.)
| | - Antonina Collura
- Unità Operativa di Microbiologia e Virologia, Ospedale Civico e di Cristina, ARNAS, 90129 Palermo, Italy; (F.D.B.); (A.C.)
| | - Vito Martella
- Dipartimento di Sanità Pubblica e Zootecnia, Università Aldo Moro di Bari, 70010 Valenzano, Italy;
| | - Simona De Grazia
- Dipartimento di Scienze per la Promozione della Salute, Materno-Infantile, di Medicina Interna e Specialistica di Eccellenza “G. D’Alessandro”, Università di Palermo, Via del Vespro 133, 90127 Palermo, Italy; (C.F.); (M.P.); (G.L.S.); (F.C.); (E.P.); (S.D.G.); (G.M.G.)
| | - Giovanni M. Giammanco
- Dipartimento di Scienze per la Promozione della Salute, Materno-Infantile, di Medicina Interna e Specialistica di Eccellenza “G. D’Alessandro”, Università di Palermo, Via del Vespro 133, 90127 Palermo, Italy; (C.F.); (M.P.); (G.L.S.); (F.C.); (E.P.); (S.D.G.); (G.M.G.)
| |
Collapse
|
15
|
Ibaraki M, Lai L, Huerta C, Natrajan MS, Collins MH, Anderson EJ, Mulligan MJ, Rouphael N, Moe CL, Liu P. Blockade Antibody Responses in Human Subjects Challenged with a New Snow Mountain Virus Inoculum. ARCHIVES OF MICROBIOLOGY & IMMUNOLOGY 2023; 7:318-325. [PMID: 38707746 PMCID: PMC11067712 DOI: 10.26502/ami.936500129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2024]
Abstract
Background Noroviruses (NoVs) are a leading cause of non-bacterial gastroenteritis in young children and adults worldwide. Snow Mountain Virus (SMV) is the prototype of NoV GII genotype 2 (GII.2) that has been developed as a viral model for human challenge studies, an important tool for studying pathogenesis and immune response of NoV infections and for evaluating NoV vaccine candidates. Previous studies have identified blockade antibodies that block the binding of NoV virus-like particles (VLPs) to histo-blood group antigens (HBGAs) as a surrogate for neutralization in human Norwalk virus and GII.4 infections but little is known about SMV blockade antibodies. Methods In this secondary data analysis study, blockade antibodies were characterized in pre-challenge and post-challenge serum samples from human subjects challenged with a new SMV inoculum. The correlation between blockade antibody geometric mean antibody titers (GMTs) and SMV-specific serum IgG/IgA GMTs were examined after stratifying the subjects by infection status. A linear mixed model was applied to test the association between HBGA blockade antibody concentrations and post-challenge days accounting for covariates and random effects. Results Laboratory results from 33 SMV inoculated individuals were analyzed and 75.7% (25/33) participants became infected. Serum SMV-specific blockade antibodies, IgA, and IgG were all significantly different between infected and uninfected individuals beginning day 15 post-challenge. Within infected individuals, a significant correlation was observed between both IgG and IgA and blockade antibody concentration as early as day 6 post-challenge. Analysis of blockade antibody using the linear mixed model showed that infected individuals, when compared to uninfected individuals, had a statistically significant increase in blockade antibody concentrations across the post-challenge days. Among the post-challenge days, blockade antibody concentrations on days 15, 30, and 45 were significantly higher than those observed pre-challenge. The intraclass correlation coefficient (ICC) analysis indicated that the variability of blockade antibody titers is more observed between individuals rather than within subjects. Conclusions These results indicate that HBGA-blockade antibody GMTs are generated after SMV challenge and the blockade antibodies were still detectable at day 45 post-challenge. These data indicate that the second-generation of SMV inoculum is highly effective.
Collapse
Affiliation(s)
- Makoto Ibaraki
- Rollins School of Public Health, Emory University, Atlanta, Georgia, USA
| | - Lilin Lai
- Grossman School of Medicine and New York University Vaccine Center, New York University, New York, USA
| | - Christopher Huerta
- Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, School of Medicine, Emory University, Atlanta, Georgia, USA
| | - Muktha S Natrajan
- Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, School of Medicine, Emory University, Atlanta, Georgia, USA
| | - Matthew H Collins
- Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, School of Medicine, Emory University, Atlanta, Georgia, USA
| | - Evan J Anderson
- Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, School of Medicine, Emory University, Atlanta, Georgia, USA
- Division of Infectious Diseases, Department of Pediatrics, School of Medicine, Emory University, Atlanta, Georgia, USA
| | - Mark J Mulligan
- Grossman School of Medicine and New York University Vaccine Center, New York University, New York, USA
| | - Nadine Rouphael
- Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, School of Medicine, Emory University, Atlanta, Georgia, USA
| | - Christine L Moe
- Rollins School of Public Health, Emory University, Atlanta, Georgia, USA
| | - Pengbo Liu
- Rollins School of Public Health, Emory University, Atlanta, Georgia, USA
| |
Collapse
|
16
|
Bernard-Raichon L, Cadwell K. Immunomodulation by Enteric Viruses. Annu Rev Virol 2023; 10:477-502. [PMID: 37380186 DOI: 10.1146/annurev-virology-111821-112317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/30/2023]
Abstract
Enteric viruses display intricate adaptations to the host mucosal immune system to successfully reproduce in the gastrointestinal tract and cause maladies ranging from gastroenteritis to life-threatening disease upon extraintestinal dissemination. However, many viral infections are asymptomatic, and their presence in the gut is associated with an altered immune landscape that can be beneficial or adverse in certain contexts. Genetic variation in the host and environmental factors including the bacterial microbiota influence how the immune system responds to infections in a remarkably viral strain-specific manner. This immune response, in turn, determines whether a given virus establishes acute versus chronic infection, which may have long-lasting consequences such as susceptibility to inflammatory disease. In this review, we summarize our current understanding of the mechanisms involved in the interaction between enteric viruses and the immune system that underlie the impact of these ubiquitous infectious agents on our health.
Collapse
Affiliation(s)
- Lucie Bernard-Raichon
- Cell Biology Department, New York University Grossman School of Medicine, New York, NY, USA
| | - Ken Cadwell
- Division of Gastroenterology and Hepatology, Department of Medicine; Department of Systems Pharmacology and Translational Therapeutics; Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA;
| |
Collapse
|
17
|
Ibaraki M, Lai L, Huerta C, Natrajan MS, Collins MH, Anderson EJ, Mulligan MJ, Rouphael N, Moe CL, Liu P. Blockade Antibody Responses in Human Subjects Challenged with a New Snow Mountain Virus Inoculum. RESEARCH SQUARE 2023:rs.3.rs-3153900. [PMID: 37790500 PMCID: PMC10543019 DOI: 10.21203/rs.3.rs-3153900/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
Background Noroviruses (NoVs) are a leading cause of non-bacterial gastroenteritis in young children and adults worldwide. Snow Mountain Virus (SMV) is the prototype of NoV GII genotype 2 (GII.2) that has been developed as a viral model for human challenge models, an important tool for studying pathogenesis and immune response of NoV infections and for evaluating NoV vaccine candidates. Previous studies have identified blockade antibodies that block the binding of NoV virus-like particles (VLPs) to histo-blood group antigens (HBGAs) as a surrogate for neutralization in human Norwalk virus and GII.4 infections but little is known about SMV blockade antibodies. Methods In this secondary data analysis study, blockade antibodies were characterized in pre-challenge and post-challenge serum samples from human subjects challenged with a new SMV inoculum. The correlation between blockade antibody geometric mean antibody titers (GMTs) and SMV-specific serum IgG/IgA GMTs were examined after stratifying the subjects by infection status. A linear mixed model was applied to test the association between HBGA blockade antibody concentrations and post-challenge days accounting for covariates and random effects. Results Laboratory results from 33 SMV inoculated individuals were analyzed and 75.7% (25/33) participants became infected. Serum SMV-specific blockade antibodies, IgA, and IgG were all significantly different between infected and uninfected individuals beginning day 15 post-challenge. Within infected individuals, a significant correlation was observed between both IgG and IgA and blockade antibody concentration as early as day 6 post-challenge. Analysis of blockade antibody using the linear mixed model showed that infected individuals, when compared to uninfected individuals, had a statistically significant increase in blockade antibody concentrations across the post-challenge days. Among the post-challenge days, blockade antibody concentrations on days 15, 30, and 45 were significantly higher than those observed pre-challenge. The intraclass correlation coefficient (ICC) analysis indicated that the variability of blockade antibody titers is more observed between individuals rather than observations within subjects. Conclusions These results indicate that HBGA-blockade antibody GMTs are generated after SMV challenge and the blockade antibodies were still detectable at day 45 post-challenge. These data indicate that the second generation of SMV inoculum is highly effective.
Collapse
Affiliation(s)
| | - Lilin Lai
- New York University Vaccine Center, New York University
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Strother CA, Brewer-Jensen PD, Becker-Dreps S, Zepeda O, May S, Gonzalez F, Reyes Y, McElvany BD, Averill AM, Mallory ML, Montmayeur AM, Costantini VP, Vinjé J, Baric RS, Bucardo F, Lindesmith LC, Diehl SA. Infant antibody and B-cell responses following confirmed pediatric GII.17 norovirus infections functionally distinguish GII.17 genetic clusters. Front Immunol 2023; 14:1229724. [PMID: 37662930 PMCID: PMC10471973 DOI: 10.3389/fimmu.2023.1229724] [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: 05/26/2023] [Accepted: 07/25/2023] [Indexed: 09/05/2023] Open
Abstract
Genogroup II (GII) noroviruses are a major cause of diarrheal disease burden in children in both high- and low-income countries. GII.17 noroviruses are composed of distinct genetic clusters (I, II, IIIa, and IIIb) and have shown potential for replacing historically more prevalent GII.4 strains, but the serological basis for GII.17 antigenic diversity has not been studied in children. Utilizing samples from a birth cohort, we investigated antibody and B-cell responses to GII.17 cluster variants in confirmed GII.17 infections in young children as well as demonstrated that the distinct genetic clusters co-circulate. Polyclonal serum antibodies bound multiple clusters but showed cluster-specific blockade activity in a surrogate virus neutralization assay. Antibodies secreted by immortalized memory B cells (MBCs) from an infant GII.17 case were highly specific to GII.17 and exhibited blockade activity against this genotype. We isolated an MBC-derived GII.17-specific Immunoglobulin A (IgA) monoclonal antibody called NVA.1 that potently and selectively blocked GII.17 cluster IIIb and recognized an epitope targeted in serum from cluster IIIb-infected children. These data indicate that multiple antigenically distinct GII.17 variants co-circulate in young children, suggesting retention of cluster diversity alongside potential for immune escape given the existence of antibody-defined cluster-specific epitopes elicited during infection.
Collapse
Affiliation(s)
- Camilla A. Strother
- Department of Microbiology and Molecular Genetics, Larner College of Medicine, University of Vermont, Burlington, VT, United States
- Cellular, Molecular, and Biomedical Sciences Graduate Program, University of Vermont, Burlington, VT, United States
- Translational Global Infectious Disease Research Center, Larner College of Medicine, University of Vermont, Burlington, VT, United States
| | - Paul D. Brewer-Jensen
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Sylvia Becker-Dreps
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Family Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Omar Zepeda
- Department of Microbiology and Parasitology, Faculty of Medical Sciences, National Autonomous University of Nicaragua, León, Nicaragua
| | - Samantha May
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Fredman Gonzalez
- Department of Microbiology and Parasitology, Faculty of Medical Sciences, National Autonomous University of Nicaragua, León, Nicaragua
| | - Yaoska Reyes
- Department of Microbiology and Parasitology, Faculty of Medical Sciences, National Autonomous University of Nicaragua, León, Nicaragua
| | - Benjamin D. McElvany
- Department of Microbiology and Molecular Genetics, Larner College of Medicine, University of Vermont, Burlington, VT, United States
| | - April M. Averill
- Department of Microbiology and Molecular Genetics, Larner College of Medicine, University of Vermont, Burlington, VT, United States
| | - Michael L. Mallory
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Anna M. Montmayeur
- National Calicivirus Laboratory, Centers for Disease Control and Prevention, Atlanta, GA, United States
| | - Verónica P. Costantini
- National Calicivirus Laboratory, Centers for Disease Control and Prevention, Atlanta, GA, United States
| | - Jan Vinjé
- National Calicivirus Laboratory, Centers for Disease Control and Prevention, Atlanta, GA, United States
| | - Ralph S. Baric
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Filemon Bucardo
- Department of Microbiology and Parasitology, Faculty of Medical Sciences, National Autonomous University of Nicaragua, León, Nicaragua
| | - Lisa C. Lindesmith
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Sean A. Diehl
- Department of Microbiology and Molecular Genetics, Larner College of Medicine, University of Vermont, Burlington, VT, United States
- Cellular, Molecular, and Biomedical Sciences Graduate Program, University of Vermont, Burlington, VT, United States
- Translational Global Infectious Disease Research Center, Larner College of Medicine, University of Vermont, Burlington, VT, United States
| |
Collapse
|
19
|
Li J, Wang B, He X, Li Z, Sun L, Li W, Bai G. Epidemiological characteristics of norovirus infection in pediatric patients during the COVID-19 pandemic. J Med Virol 2023; 95:e28874. [PMID: 37322803 DOI: 10.1002/jmv.28874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 05/10/2023] [Accepted: 05/29/2023] [Indexed: 06/17/2023]
Abstract
To assess the epidemiological characteristics of norovirus infection. We included 5564 patients under the age of 18 years who visited the hospital in which the study took place from December 2020 to November 2022 with a primary diagnosis of acute diarrhea. Clinical information was extracted from the electronic health record system. We calculated the prevalence of norovirus infection by age, gender, season, year, and type of patients. A nonlinear association between age and prevalence rates was assessed using a restricted cubic spline regression model. A total of 5564 patients completed the test for human norovirus, among whom 1442 (25.9%) tested positive. The prevalence of norovirus infection was significantly lower in 2022 than in 2021 (35.9% vs. 53.7%, p < 0.001), and the highest prevalence was observed in winter (35.1%) and then followed by autumn (27.5%). Regarding the age pattern, the highest rate was seen in children aged 1-3 years (37.5%). Children at age 1.5 years may have the highest risk of having norovirus infection (Pnonlinear < 0.001). The prevalence of norovirus infection of norovirus during the COVID-19 pandemic was similar to that before the pandemic shown in literatures. A relatively high rate was observed in cool seasons and in younger children (i.e., 1-3 years).
Collapse
Affiliation(s)
- Jiabin Li
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
- Department of Pharmacy, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Binghan Wang
- School of Public Health, Zhejiang University School of Medicine, Hangzhou, China
| | - Xinyu He
- Department of Child Health Care, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Ziqiao Li
- Department of Child Health Care, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Lidan Sun
- Department of Child Health Care, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Wei Li
- Department of Clinical Laboratory, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Guannan Bai
- Department of Child Health Care, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| |
Collapse
|
20
|
Lin SC, Bai GH, Lin PC, Chen CY, Hsu YH, Lee YC, Chen SY. Molecular and Genetics-Based Systems for Tracing the Evolution and Exploring the Mechanisms of Human Norovirus Infections. Int J Mol Sci 2023; 24:ijms24109093. [PMID: 37240438 DOI: 10.3390/ijms24109093] [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: 04/04/2023] [Revised: 05/09/2023] [Accepted: 05/16/2023] [Indexed: 05/28/2023] Open
Abstract
Human noroviruses (HuNoV) are major causes of acute gastroenteritis around the world. The high mutation rate and recombination potential of noroviruses are significant challenges in studying the genetic diversity and evolution pattern of novel strains. In this review, we describe recent advances in the development of technologies for not only the detection but also the analysis of complete genome sequences of noroviruses and the future prospects of detection methods for tracing the evolution and genetic diversity of human noroviruses. The mechanisms of HuNoV infection and the development of antiviral drugs have been hampered by failure to develop the infectious virus in a cell model. However, recent studies have demonstrated the potential of reverse genetics for the recovery and generation of infectious viral particles, suggesting the utility of this genetics-based system as an alternative for studying the mechanisms of viral infection, such as cell entry and replication.
Collapse
Affiliation(s)
- Sheng-Chieh Lin
- Department of Pediatrics, School of Medicine, College of Medicine, Taipei Medical University, Taipei City 11031, Taiwan
- Division of Allergy, Asthma, and Immunology, Department of Pediatrics, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan
| | - Geng-Hao Bai
- Department of Internal Medicine, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei City 10002, Taiwan
| | - Pei-Chun Lin
- Division of Pediatric Gastroenterology, Department of Pediatrics, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan
| | - Chung-Yung Chen
- Department of Bioscience Technology, Chung Yuan Christian University, Taoyuan City 32023, Taiwan
- Center for Nanotechnology, Institute of Biomedical Technology, Chung Yuan Christian University, Taoyuan City 32023, Taiwan
| | - Yi-Hsiang Hsu
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Yuan-Chang Lee
- Department of Infectious Diseases, School of Medicine, College of Medicine, Taipei Medical University, Taipei City 11031, Taiwan
- Department of Infectious Diseases, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan
| | - Shih-Yen Chen
- Department of Pediatrics, School of Medicine, College of Medicine, Taipei Medical University, Taipei City 11031, Taiwan
- Division of Pediatric Gastroenterology, Department of Pediatrics, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan
- TMU Research Center for Digestive Medicine, Taipei Medical University, Taipei City 11031, Taiwan
| |
Collapse
|
21
|
Kher G, Sabin C, Lun JH, Devant JM, Ruoff K, Koromyslova AD, von Itzstein M, Pancera M, Hansman GS. Direct Blockade of the Norovirus Histo-Blood Group Antigen Binding Pocket by Nanobodies. J Virol 2023; 97:e0183322. [PMID: 36971561 PMCID: PMC10134814 DOI: 10.1128/jvi.01833-22] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 12/27/2022] [Indexed: 03/29/2023] Open
Abstract
Noroviruses are the leading cause of outbreaks of acute gastroenteritis. These viruses usually interact with histo-blood group antigens (HBGAs), which are considered essential cofactors for norovirus infection. This study structurally characterizes nanobodies developed against the clinically important GII.4 and GII.17 noroviruses with a focus on the identification of novel nanobodies that efficiently block the HBGA binding site. Using X-ray crystallography, we have characterized nine different nanobodies that bound to the top, side, or bottom of the P domain. The eight nanobodies that bound to the top or side of the P domain were mainly genotype specific, while one nanobody that bound to the bottom cross-reacted against several genotypes and showed HBGA blocking potential. The four nanobodies that bound to the top of the P domain also inhibited HBGA binding, and structural analysis revealed that these nanobodies interacted with several GII.4 and GII.17 P domain residues that commonly engaged HBGAs. Moreover, these nanobody complementarity-determining regions (CDRs) extended completely into the cofactor pockets and would likely impede HBGA engagement. The atomic level information for these nanobodies and their corresponding binding sites provide a valuable template for the discovery of additional "designer" nanobodies. These next-generation nanobodies would be designed to target other important genotypes and variants, while maintaining cofactor interference. Finally, our results clearly demonstrate for the first time that nanobodies directly targeting the HBGA binding site can function as potent norovirus inhibitors. IMPORTANCE Human noroviruses are highly contagious and a major problem in closed institutions, such as schools, hospitals, and cruise ships. Reducing norovirus infections is challenging on multiple levels and includes the frequent emergence of antigenic variants, which complicates designing effective, broadly reactive capsid therapeutics. We successfully developed and characterized four norovirus nanobodies that bound at the HBGA pockets. Compared with previously developed norovirus nanobodies that inhibited HBGA through disrupted particle stability, these four novel nanobodies directly inhibited HBGA engagement and interacted with HBGA binding residues. Importantly, these new nanobodies specifically target two genotypes that have caused the majority of outbreaks worldwide and consequently would have an enormous benefit if they could be further developed as norovirus therapeutics. To date, we have structurally characterized 16 different GII nanobody complexes, a number of which block HBGA binding. These structural data could be used to design multivalent nanobody constructs with improved inhibition properties.
Collapse
Affiliation(s)
- Gargi Kher
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Charles Sabin
- Schaller Research Group, University of Heidelberg, DKFZ, Heidelberg, Germany
- Department of Infectious Diseases, Virology, University of Heidelberg, Heidelberg, Germany
| | - Jennifer H. Lun
- Schaller Research Group, University of Heidelberg, DKFZ, Heidelberg, Germany
- Department of Infectious Diseases, Virology, University of Heidelberg, Heidelberg, Germany
| | - Jessica M. Devant
- Schaller Research Group, University of Heidelberg, DKFZ, Heidelberg, Germany
- Department of Infectious Diseases, Virology, University of Heidelberg, Heidelberg, Germany
| | - Kerstin Ruoff
- Schaller Research Group, University of Heidelberg, DKFZ, Heidelberg, Germany
- Department of Infectious Diseases, Virology, University of Heidelberg, Heidelberg, Germany
| | - Anna D. Koromyslova
- Schaller Research Group, University of Heidelberg, DKFZ, Heidelberg, Germany
- Department of Infectious Diseases, Virology, University of Heidelberg, Heidelberg, Germany
| | - Mark von Itzstein
- Institute for Glycomics, Griffith University, Gold Coast Campus, Gold Coast, Queensland, Australia
| | - Marie Pancera
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Grant S. Hansman
- Schaller Research Group, University of Heidelberg, DKFZ, Heidelberg, Germany
- Department of Infectious Diseases, Virology, University of Heidelberg, Heidelberg, Germany
- Institute for Glycomics, Griffith University, Gold Coast Campus, Gold Coast, Queensland, Australia
| |
Collapse
|
22
|
Kumazaki M, Usuku S. Influence of herd immunity on norovirus: a long-term field study of repeated viral gastroenteritis outbreaks at the same facilities. BMC Infect Dis 2023; 23:265. [PMID: 37101126 PMCID: PMC10132420 DOI: 10.1186/s12879-023-08251-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 04/13/2023] [Indexed: 04/28/2023] Open
Abstract
BACKGROUND Viral acute gastroenteritis (AG) is detected worldwide annually. Outbreaks caused by viruses associated with gastroenteritis have been reported repeatedly at the same facilities in Yokohama, Japan over several years. We investigated the statuses of these repeated outbreaks to consider herd immunity at the facility level. METHODS Between September 2007 and August 2017, 1459 AG outbreaks were reported at 1099 facilities. Stool samples were collected for virological testing, and the norovirus gene was amplified and sequenced to determine the genotype using the N-terminal region of the capsid. RESULTS The outbreaks were caused by norovirus, sapovirus, rotavirus A, and rotavirus C. Norovirus was consistently predominant over the 10-year period. Of 1099 facilities, 227 reported multiple outbreaks, of which norovirus-only combinations accounted for 76.2%. More outbreaks were due to different genotype combinations than the same genotype combinations. For facilities that experienced two norovirus outbreaks, the average interval between outbreaks was longer for groups with the same combinations than for groups with different genogroup or genotype combinations, although no statistically significant differences were observed. At 44 facilities, outbreaks occurred repeatedly during the same AG season, and most exhibited combinations of different norovirus genotypes or viruses. Among 49 combinations with the same norovirus genotype at the same facilities over 10 years, the most prevalent genotypes were combinations of genogroup II genotype 4 (GII.4), followed by GII.2, GII.6, GII.3, GII.14, and GI.3. The mean interval between outbreaks was 31.2 ± 26.8 months for all combinations, and the mean intervals were longer for non-GII.4 genotype cases than for GII.4 cases, and statistically significant differences were observed (t-test, P < 0.05). Additionally, these average intervals were longer for kindergarten/nursery schools and primary schools than for nursing homes for older adults (t-test, P < 0.05). CONCLUSIONS Repeated AG outbreaks at the same facilities in Yokohama during the 10-year study period included mainly norovirus combinations. Herd immunity at the facility level was maintained for at least the same AG season. Norovirus genotype-specific herd immunity was maintained for an average of 31.2 months during the study period, and these intervals differed depending on genotype.
Collapse
Affiliation(s)
- Makoto Kumazaki
- Microbiological Testing and Research Division, Yokohama City Institute of Public Health, 2-7-1 Tomiokahigashi, Kanazawa-Ku, Yokohama, Kanagawa, 236-0051, Japan.
| | - Shuzo Usuku
- Microbiological Testing and Research Division, Yokohama City Institute of Public Health, 2-7-1 Tomiokahigashi, Kanazawa-Ku, Yokohama, Kanagawa, 236-0051, Japan
| |
Collapse
|
23
|
Lindesmith LC, Brewer-Jensen PD, Conrad H, O’Reilly KM, Mallory ML, Kelly D, Williams R, Edmunds WJ, Allen DJ, Breuer J, Baric RS. Emergent variant modeling of the serological repertoire to norovirus in young children. Cell Rep Med 2023; 4:100954. [PMID: 36854303 PMCID: PMC10040388 DOI: 10.1016/j.xcrm.2023.100954] [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: 09/14/2022] [Revised: 12/05/2022] [Accepted: 02/02/2023] [Indexed: 03/02/2023]
Abstract
Human norovirus is the leading cause of acute gastroenteritis. Young children and the elderly bear the greatest burden of disease, representing more than 200,000 deaths annually. Infection prevalence peaks at younger than 2 years and is driven by novel GII.4 variants that emerge and spread globally. Using a surrogate neutralization assay, we characterize the evolution of the serological neutralizing antibody (nAb) landscape in young children as they transition between sequential GII.4 pandemic variants. Following upsurge of the replacement variant, antigenic cartography illustrates remodeling of the nAb landscape to the new variant accompanied by improved nAb titer. However, nAb relative avidity remains focused on the preceding variant. These data support immune imprinting as a mechanism of immune evasion and GII.4 virus persistence across a population. Understanding the complexities of immunity to rapidly evolving and co-circulating viral variants, like those of norovirus, severe acute respiratory syndrome coronavirus 2 (SARS-CoV2), and dengue viruses, will fundamentally inform vaccine design for emerging pathogens.
Collapse
Affiliation(s)
- Lisa C. Lindesmith
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Paul D. Brewer-Jensen
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Helen Conrad
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Kathleen M. O’Reilly
- Centre for Mathematical Modelling of Infectious Diseases and Department of Infectious Disease Epidemiology, London School of Hygiene and Tropical Medicine, London WC1EW 7HT, UK
| | - Michael L. Mallory
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Daniel Kelly
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London WC1E 7HT, UK
| | - Rachel Williams
- Department of Infection, Immunity and Inflammation, UCL Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
- Department of Genetics & Genomic Medicine, UCL Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - W. John Edmunds
- Centre for Mathematical Modelling of Infectious Diseases and Department of Infectious Disease Epidemiology, London School of Hygiene and Tropical Medicine, London WC1EW 7HT, UK
| | - David J. Allen
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London WC1E 7HT, UK
| | - Judith Breuer
- Department of Infection, Immunity and Inflammation, UCL Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
- Department of Microbiology, Great Ormond Street Hospital for Children NHS Foundation Trust, London WC1N 3JH, UK
| | - Ralph S. Baric
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
24
|
Lipska AG, Sieradzan AK, Czaplewski C, Lipińska AD, Ocetkiewicz KM, Proficz J, Czarnul P, Krawczyk H, Liwo A. Long-time scale simulations of virus-like particles from three human-norovirus strains. J Comput Chem 2023; 44:1470-1483. [PMID: 36799410 DOI: 10.1002/jcc.27087] [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: 11/07/2022] [Revised: 12/22/2022] [Accepted: 01/29/2023] [Indexed: 02/18/2023]
Abstract
The dynamics of the virus like particles (VLPs) corresponding to the GII.4 Houston, GII.2 SMV, and GI.1 Norwalk strains of human noroviruses (HuNoV) that cause gastroenteritis was investigated by means of long-time (about 30 μs in the laboratory timescale) molecular dynamics simulations with the coarse-grained UNRES force field. The main motion of VLP units turned out to be the bending at the junction between the P1 subdomain (that sits in the VLP shell) and the P2 subdomain (that protrudes outside) of the major VP1 protein, this resulting in a correlated wagging motion of the P2 subdomains with respect to the VLP surface. The fluctuations of the P2 subdomain were found to be more pronounced and the P2 domain made a greater angle with the normal to the VLP surface for the GII.2 strain, which could explain the inability of this strain to bind the histo-blood group antigens (HBGAs).
Collapse
Affiliation(s)
- Agnieszka G Lipska
- Centre of Informatics Tri-city Academic Supercomputer and Network (CI TASK), Gdańsk University of Technology, Fahrenheit Union of Universities in Gdańsk, Gdańsk, Poland
| | - Adam K Sieradzan
- Centre of Informatics Tri-city Academic Supercomputer and Network (CI TASK), Gdańsk University of Technology, Fahrenheit Union of Universities in Gdańsk, Gdańsk, Poland.,Faculty of Chemistry, University of Gdańsk, Fahrenheit Union of Universities in Gdańsk, Gdańsk, Poland
| | - Cezary Czaplewski
- Centre of Informatics Tri-city Academic Supercomputer and Network (CI TASK), Gdańsk University of Technology, Fahrenheit Union of Universities in Gdańsk, Gdańsk, Poland.,Faculty of Chemistry, University of Gdańsk, Fahrenheit Union of Universities in Gdańsk, Gdańsk, Poland
| | - Andrea D Lipińska
- Laboratory of Virus Molecular Biology, Intercollegiate Faculty of Biotechnology, University of Gdańsk and Medical University of Gdańsk, Fahrenheit Union of Universities in Gdańsk, Gdańsk, Poland
| | - Krzysztof M Ocetkiewicz
- Centre of Informatics Tri-city Academic Supercomputer and Network (CI TASK), Gdańsk University of Technology, Fahrenheit Union of Universities in Gdańsk, Gdańsk, Poland
| | - Jerzy Proficz
- Centre of Informatics Tri-city Academic Supercomputer and Network (CI TASK), Gdańsk University of Technology, Fahrenheit Union of Universities in Gdańsk, Gdańsk, Poland
| | - Paweł Czarnul
- Faculty of Electronics, Telecommunications and Informatics, Gdańsk University of Technology, Fahrenheit Union of Universities in Gdańsk, Gdańsk, Poland
| | - Henryk Krawczyk
- Centre of Informatics Tri-city Academic Supercomputer and Network (CI TASK), Gdańsk University of Technology, Fahrenheit Union of Universities in Gdańsk, Gdańsk, Poland.,Faculty of Electronics, Telecommunications and Informatics, Gdańsk University of Technology, Fahrenheit Union of Universities in Gdańsk, Gdańsk, Poland
| | - Adam Liwo
- Centre of Informatics Tri-city Academic Supercomputer and Network (CI TASK), Gdańsk University of Technology, Fahrenheit Union of Universities in Gdańsk, Gdańsk, Poland.,Faculty of Chemistry, University of Gdańsk, Fahrenheit Union of Universities in Gdańsk, Gdańsk, Poland
| |
Collapse
|
25
|
Zhang Q, Zhu S, Zhang X, Su L, Ni J, Zhang Y, Fang L. Recent insights into reverse genetics of norovirus. Virus Res 2023; 325:199046. [PMID: 36657615 DOI: 10.1016/j.virusres.2023.199046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Revised: 12/23/2022] [Accepted: 01/15/2023] [Indexed: 01/18/2023]
Abstract
Norovirus is the leading cause of viral gastroenteritis globally, and poses substantial threats to public health. Despite substantial progress made in preventing norovirus diseases, the lack of a robust virus culture system has hampered biological research and effective strategies to combat this pathogen. Reverse genetic system is the technique to generate infectious viruses from cloned genetic constructs, which is a powerful tool for the investigation of viral pathogenesis and for the development of novel drugs and vaccines. The strategies of reverse genetics include bacterial artificial chromosomes, vaccinia virus vectors, and entirely plasmid-based systems. Since each strategy has its pros and cons, choosing appropriate approaches will greatly improve the efficiency of virus rescue. Reverse genetic systems that have been employed for norovirus greatly extend its life cycle and facilitate the development of medical countermeasures. In this review, we summarize the current knowledge on the structure, transmission, genetic evolution and clinical manifestations of norovirus, and describe recent advances in the studies of norovirus reverse genetics as well as its future prospects for therapeutics and vaccine development.
Collapse
Affiliation(s)
- Qinyi Zhang
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, China; Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, China
| | - Shuirong Zhu
- Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, China
| | | | - Lingxuan Su
- Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, China
| | - Jun Ni
- Department of Critical Care Medicine, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, China
| | - Yanjun Zhang
- Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, China.
| | - Lei Fang
- Department of Critical Care Medicine, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, China.
| |
Collapse
|
26
|
Lindesmith LC, Verardi R, Mallory ML, Edwards CE, Graham RL, Zweigart M, Brewer-Jensen PD, Debbink K, Kocher JF, Kwong PD, Baric RS. Norovirus. PLOTKIN'S VACCINES 2023:747-754.e5. [DOI: 10.1016/b978-0-323-79058-1.00043-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
27
|
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
|
28
|
Elbashir I, Aldoos NF, Mathew S, Al Thani AA, Emara MM, Yassine HM. Molecular epidemiology, genetic diversity, and vaccine availability of viral acute gastroenteritis in the middle East and North Africa (MENA) region. J Infect Public Health 2022; 15:1193-1211. [PMID: 36240530 DOI: 10.1016/j.jiph.2022.09.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 08/26/2022] [Accepted: 09/11/2022] [Indexed: 11/18/2022] Open
Abstract
Acute gastroenteritis is the cause of considerable mortality and morbidity worldwide, particularly among children under five years in underdeveloped countries. Most acute gastroenteritis (AGE) cases are attributed to viral etiologies, including rotavirus, norovirus, adenovirus, astrovirus, and sapovirus. This paper aimed to determine the prevalence rate of different viral etiologies of AGE in the Middle East and North Africa (MENA) region. Moreover, this paper explored rotavirus phylogenetic relatedness, compared VP7 and VP4 antigenic regions of rotavirus with vaccine strains, and explored the availability of vaccines in the MENA region. The literature search identified 160 studies from 18 countries from 1980 to 2019. The overall prevalence of rotavirus, norovirus, adenovirus, astrovirus, and sapovirus were 29.8 %, 13.9 %, 6.3 %, 3.5 %, and 3.2 % of tested samples, respectively. The most common rotavirus genotype combinations in the MENA region were G1P[8], G9P[9], and G2P[4], whereas GII.4 was the predominant norovirus genotype all of which were reported in almost all the studies with genotyping data. The comparison of VP7 and VP4 between circulating rotavirus in the MENA region and vaccine strains has revealed discrete divergent regions, including the neutralizing epitopes. Rotavirus vaccine was introduced to most of the countries of the MENA region; however, only a few studies have assessed the effectiveness of vaccine introduction. This paper provides a comprehensive update on the prevalence of the different viral agents of AGE in the MENA region.
Collapse
Affiliation(s)
- Israa Elbashir
- Biomedical Research Center, Qatar University, 2713 Doha, Qatar.
| | - Noor F Aldoos
- Department of Laboratory Medicine and Pathology, Hamad Medical Corporation, Doha, Qatar.
| | - Shilu Mathew
- Biomedical Research Center, Qatar University, 2713 Doha, Qatar.
| | - Asmaa A Al Thani
- Biomedical Research Center, Qatar University, 2713 Doha, Qatar; Department of Biomedical Sciences, College of Health Science-QU Health, Qatar University, Doha 2713, Qatar
| | - Mohamed M Emara
- Basic Medical Sciences Department, College of Medicine, QU Health, Qatar University, 2713 Doha, Qatar.
| | - Hadi M Yassine
- Biomedical Research Center, Qatar University, 2713 Doha, Qatar.
| |
Collapse
|
29
|
Lindesmith LC, Boshier FAT, Brewer-Jensen PD, Roy S, Costantini V, Mallory ML, Zweigart M, May SR, Conrad H, O’Reilly KM, Kelly D, Celma CC, Beard S, Williams R, Tutill HJ, Becker Dreps S, Bucardo F, Allen DJ, Vinjé J, Goldstein RA, Breuer J, Baric RS. Immune Imprinting Drives Human Norovirus Potential for Global Spread. mBio 2022; 13:e0186122. [PMID: 36102514 PMCID: PMC9600701 DOI: 10.1128/mbio.01861-22] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 08/25/2022] [Indexed: 01/11/2023] Open
Abstract
Understanding the complex interactions between virus and host that drive new strain evolution is key to predicting the emergence potential of variants and informing vaccine development. Under our hypothesis, future dominant human norovirus GII.4 variants with critical antigenic properties that allow them to spread are currently circulating undetected, having diverged years earlier. Through large-scale sequencing of GII.4 surveillance samples, we identified two variants with extensive divergence within domains that mediate neutralizing antibody binding. Subsequent serological characterization of these strains using temporally resolved adult and child sera suggests that neither candidate could spread globally in adults with multiple GII.4 exposures, yet young children with minimal GII.4 exposure appear susceptible. Antigenic cartography of surveillance and outbreak sera indicates that continued population exposure to GII.4 Sydney 2012 and antigenically related variants over a 6-year period resulted in a broadening of immunity to heterogeneous GII.4 variants, including those identified here. We show that the strongest antibody responses in adults exposed to GII.4 Sydney 2012 are directed to previously circulating GII.4 viruses. Our data suggest that the broadening of antibody responses compromises establishment of strong GII.4 Sydney 2012 immunity, thereby allowing the continued persistence of GII.4 Sydney 2012 and modulating the cycle of norovirus GII.4 variant replacement. Our results indicate a cycle of norovirus GII.4 variant replacement dependent upon population immunity. Young children are susceptible to divergent variants; therefore, emergence of these strains worldwide is driven proximally by changes in adult serological immunity and distally by viral evolution that confers fitness in the context of immunity. IMPORTANCE In our model, preepidemic human norovirus variants harbor genetic diversification that translates into novel antigenic features without compromising viral fitness. Through surveillance, we identified two viruses fitting this profile, forming long branches on a phylogenetic tree. Neither evades current adult immunity, yet young children are likely susceptible. By comparing serological responses, we demonstrate that population immunity varies by age/exposure, impacting predicted susceptibility to variants. Repeat exposure to antigenically similar variants broadens antibody responses, providing immunological coverage of diverse variants but compromising response to the infecting variant, allowing continued circulation. These data indicate norovirus GII.4 variant replacement is driven distally by virus evolution and proximally by immunity in adults.
Collapse
Affiliation(s)
- Lisa C. Lindesmith
- Department of Epidemiology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Florencia A. T. Boshier
- Department of Infection, Immunity and Inflammation, UCL Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Paul D. Brewer-Jensen
- Department of Epidemiology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Sunando Roy
- Department of Infection, Immunity and Inflammation, UCL Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Veronica Costantini
- Division of Viral Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Michael L. Mallory
- Department of Epidemiology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Mark Zweigart
- Department of Epidemiology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Samantha R. May
- Department of Epidemiology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Helen Conrad
- Department of Epidemiology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Kathleen M. O’Reilly
- Centre for Mathematical Modelling of Infectious Diseases, Department of Infectious Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Daniel Kelly
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Cristina C. Celma
- Enteric Virus Unit, The Virus Reference Department, UK Health Security Agency, London, United Kingdom
| | - Stuart Beard
- Enteric Virus Unit, The Virus Reference Department, UK Health Security Agency, London, United Kingdom
| | - Rachel Williams
- Department of Infection, Immunity and Inflammation, UCL Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
- Department of Genetics & Genomic Medicine, UCL Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Helena J. Tutill
- Department of Genetics & Genomic Medicine, UCL Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Sylvia Becker Dreps
- Department of Epidemiology, University of North Carolina, Chapel Hill, North Carolina, USA
- Department of Family Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Filemón Bucardo
- Department of Microbiology, National Autonomous University of Nicaragua, León, León, Nicaragua
| | - David J. Allen
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Jan Vinjé
- Division of Viral Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Richard A. Goldstein
- Division of Infection and Immunity, University College London, London, United Kingdom
| | - Judith Breuer
- Department of Infection, Immunity and Inflammation, UCL Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
- Department of Microbiology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, United Kingdom
| | - Ralph S. Baric
- Department of Epidemiology, University of North Carolina, Chapel Hill, North Carolina, USA
| |
Collapse
|
30
|
Ford-Siltz LA, Tohma K, Alvarado GS, Kendra JA, Pilewski KA, Crowe JE, Parra GI. Cross-reactive neutralizing human monoclonal antibodies mapping to variable antigenic sites on the norovirus major capsid protein. Front Immunol 2022; 13:1040836. [PMID: 36389818 PMCID: PMC9641292 DOI: 10.3389/fimmu.2022.1040836] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 09/30/2022] [Indexed: 04/14/2025] Open
Abstract
Human noroviruses are the major viral cause of acute gastroenteritis around the world. Although norovirus symptoms are in most cases mild and self-limited, severe and prolonged symptoms can occur in the elderly and in immunocompromised individuals. Thus, there is a great need for the development of specific therapeutics that can help mitigate infection. In this study, we sought to characterize a panel of human monoclonal antibodies (mAbs; NORO-123, -115, -273A, -263, -315B, and -250B) that showed carbohydrate blocking activity against the current pandemic variant, GII.4 Sydney 2012. All antibodies tested showed potent neutralization against GII.4 Sydney virus in human intestinal enteroid culture. While all mAbs recognized only GII.4 viruses, they exhibited differential binding patterns against a panel of virus-like particles (VLPs) representing major and minor GII.4 variants spanning twenty-five years. Using mutant VLPs, we mapped five of the mAbs to variable antigenic sites A (NORO-123, -263, -315B, and -250B) or C (NORO-115) on the major capsid protein. Those mapping to the antigenic site A showed blocking activity against multiple variants dating back to 1987, with one mAb (NORO-123) showing reactivity to all variants tested. NORO-115, which maps to antigenic site C, showed reactivity against multiple variants due to the low susceptibility for mutations presented by naturally-occurring variants at the proposed binding site. Notably, we show that cross-blocking and neutralizing antibodies can be elicited against variable antigenic sites. These data provide new insights into norovirus immunity and suggest potential for the development of cross-protective vaccines and therapeutics.
Collapse
Affiliation(s)
- Lauren A. Ford-Siltz
- Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, United States
| | - Kentaro Tohma
- Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, United States
| | - Gabriela S. Alvarado
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Joseph A. Kendra
- Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, United States
| | - Kelsey A. Pilewski
- Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, United States
| | - James E. Crowe
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States
- Department of Pediatrics and Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Gabriel I. Parra
- Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, United States
| |
Collapse
|
31
|
Intestinal Norovirus Binding Patterns in Nonsecretor Individuals. J Virol 2022; 96:e0086522. [PMID: 36121297 PMCID: PMC9555158 DOI: 10.1128/jvi.00865-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Human norovirus (HuNoV) infection is associated with an active FUT2 gene, which characterizes the secretor phenotype. However, nonsecretor individuals are also affected by HuNoV infection although in a lesser proportion. Here, we studied GII.3, GII.4, and GII.17 HuNoV interactions in nonsecretor individuals using virus-like particles (VLPs). Only GII.4 HuNoV specifically interacted with nonsecretor saliva. Competition experiments using histo-blood group antigen (HBGA)-specific monoclonal antibodies (MAbs) demonstrate that GII.4 VLPs recognized the Lewis a (Lea) antigen. We also analyzed HuNoV VLP interactions on duodenum tissue blocks from healthy nonsecretor individuals. VLP binding was observed for the three HuNoV genotypes in 10 of the 13 individuals, and competition experiments demonstrated that VLP recognition was driven by an interaction with the Lea antigen. In 3 individuals, binding was restricted to either GII.4 alone or GII.3 and GII.17. Finally, we performed a VLP binding assay on proximal and distal colon tissue blocks from a nonsecretor patient with Crohn's disease. VLP binding to inflammatory tissues was genotype specific since GII.4 and GII.17 VLPs were able to interact with regenerative mucosa, whereas GII.3 VLP was not. The binding of GII.4 and GII.17 HuNoV VLPs was linked to Lea in regenerative mucosae from the proximal and distal colon. Overall, our data clearly showed that Lea has a pivotal role in the recognition of HuNoV in nonsecretors. We also showed that Lea is expressed in inflammatory/regenerative tissues and interacts with HuNoV in a nonsecretor individual. The physiological and immunological consequences of such interactions in nonsecretors have yet to be elucidated. IMPORTANCE Human norovirus (HuNoV) is the main etiological agent of viral gastroenteritis in all age classes. HuNoV infection affects mainly secretor individuals where ABO(H) and Lewis histo-blood group antigens (HBGAs) are present in the small intestine. Nonsecretor individuals, who only express Lewis (Le) antigens, are less susceptible to HuNoV infection. Here, we studied the interaction of common HuNoV genotypes (GII.3, GII.4, and GII.17) in nonsecretor individuals using synthetic viral particles. Saliva binding assays showed that only GII.4 interacted with nonsecretor saliva via the Lewis a (Lea) antigen Surprisingly, the three genotypes interacted with nonsecretor enterocytes via the Lea antigen on duodenal tissue blocks, which were more relevant for HuNoV/HBGA studies. The Lea antigen also played a pivotal role in the recognition of GII.4 and GII.17 particles by inflammatory colon tissue from a nonsecretor Crohn's disease patient. The implications of HuNoV binding in nonsecretors remain to be elucidated in physiological and pathological conditions encountered in other intestinal diseases.
Collapse
|
32
|
Brewer-Jensen PD, Reyes Y, Becker-Dreps S, González F, Mallory ML, Gutiérrez L, Zepeda O, Centeno E, Vielot N, Diez-Valcarce M, Vinjé J, Baric R, Lindesmith LC, Bucardo F. Norovirus Infection in Young Nicaraguan Children Induces Durable and Genotype-Specific Antibody Immunity. Viruses 2022; 14:v14092053. [PMID: 36146859 PMCID: PMC9501366 DOI: 10.3390/v14092053] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 09/05/2022] [Accepted: 09/08/2022] [Indexed: 11/22/2022] Open
Abstract
There are significant challenges to the development of a pediatric norovirus vaccine, mainly due to the antigenic diversity among strains infecting young children. Characterizing human norovirus serotypes and understanding norovirus immunity in naïve children would provide key information for designing rational vaccine platforms. In this study, 26 Nicaraguan children experiencing their first norovirus acute gastroenteritis (AGE) episode during the first 18 months of life were investigated. We used a surrogate neutralization assay that measured antibodies blocking the binding of 13 different norovirus virus-like particles (VLPs) to histo-blood group antigens (HBGAs) in pre- and post-infection sera. To assess for asymptomatic norovirus infections, stools from asymptomatic children were collected monthly, screened for norovirus by RT-qPCR and genotyped by sequencing. Seroconversion of an HBGA-blocking antibody matched the infecting genotype in 25 (96%) of the 26 children. A subset of 13 (50%) and 4 (15%) of the 26 children experienced monotypic GII and GI seroconversion, respectively, strongly suggesting a type-specific response in naïve children, and 9 (35%) showed multitypic seroconversion. The most frequent pairing in multitypic seroconversion (8/12) were GII.4 Sydney and GII.12 noroviruses, both co-circulating at the time. Blocking antibody titers to these two genotypes did not correlate with each other, suggesting multiple exposure rather than cross-reactivity between genotypes. In addition, GII titers remained consistent for at least 19 months post-infection, demonstrating durable immunity. In conclusion, the first natural norovirus gastroenteritis episodes in these young children were dominated by a limited number of genotypes and induced responses of antibodies blocking binding of norovirus VLPs in a genotype-specific manner, suggesting that an effective pediatric norovirus vaccine likely needs to be multivalent and include globally dominant genotypes. The duration of protection from natural infections provides optimism for pediatric norovirus vaccines administered early in life.
Collapse
Affiliation(s)
- Paul D. Brewer-Jensen
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Yaoska Reyes
- Department of Microbiology, Faculty of Medical Sciences, National Autonomous University of Nicaragua, León 21000, Nicaragua
- Division of Molecular Medicine and Virology, Department of Clinical and Experimental Medicine, Linköping University, SE-581 83 Linköping, Sweden
| | - Sylvia Becker-Dreps
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Family Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Fredman González
- Department of Microbiology, Faculty of Medical Sciences, National Autonomous University of Nicaragua, León 21000, Nicaragua
| | - Michael L. Mallory
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Lester Gutiérrez
- Department of Microbiology, Faculty of Medical Sciences, National Autonomous University of Nicaragua, León 21000, Nicaragua
| | - Omar Zepeda
- Department of Microbiology, Faculty of Medical Sciences, National Autonomous University of Nicaragua, León 21000, Nicaragua
| | - Edwing Centeno
- Department of Microbiology, Faculty of Medical Sciences, National Autonomous University of Nicaragua, León 21000, Nicaragua
| | - Nadja Vielot
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Marta Diez-Valcarce
- Division of Viral Diseases, U.S. Centers for Disease Control and Prevention, Atlanta, GA 30329, USA
| | - Jan Vinjé
- Division of Viral Diseases, U.S. Centers for Disease Control and Prevention, Atlanta, GA 30329, USA
| | - Ralph Baric
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Lisa C. Lindesmith
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Filemon Bucardo
- Department of Microbiology, Faculty of Medical Sciences, National Autonomous University of Nicaragua, León 21000, Nicaragua
- Correspondence: ; Tel.: +505-89040938
| |
Collapse
|
33
|
Lindesmith LC, Brewer-Jensen PD, Mallory ML, Zweigart MR, May SR, Kelly D, Williams R, Becker-Dreps S, Bucardo F, Allen DJ, Breuer J, Baric RS. Antigenic Site Immunodominance Redirection Following Repeat Variant Exposure. Viruses 2022; 14:1293. [PMID: 35746763 PMCID: PMC9229260 DOI: 10.3390/v14061293] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 05/30/2022] [Accepted: 06/11/2022] [Indexed: 12/16/2022] Open
Abstract
Human norovirus is a leading cause of acute gastroenteritis, driven by antigenic variants within the GII.4 genotype. Antibody responses to GII.4 vaccination in adults are shaped by immune memory. How children without extensive immune memory will respond to GII.4 vaccination has not been reported. Here, we characterized the GII.4 neutralizing antibody (nAb) landscape following natural infection using a surrogate assay and antigenic site chimera virus-like particles. We demonstrate that the nAb landscape changes with age and virus exposure. Among sites A, C, and G, nAbs from first infections are focused on sites A and C. As immunity develops with age/exposure, site A is supplemented with antibodies that bridge site A to sites C and G. Cross-site nAbs continue to develop into adulthood, accompanied by an increase in nAb to site G. Continued exposure to GII.4 2012 Sydney correlated with a shift to co-dominance of sites A and G. Furthermore, site G nAbs correlated with the broadening of nAb titer across antigenically divergent variants. These data describe fundamental steps in the development of immunity to GII.4 over a lifetime, and illustrate how the antigenicity of one pandemic variant could influence the pandemic potential of another variant through the redirection of immunodominant epitopes.
Collapse
Affiliation(s)
- Lisa C. Lindesmith
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (P.D.B.-J.); (M.L.M.); (M.R.Z.); (S.R.M.); (S.B.-D.)
| | - Paul D. Brewer-Jensen
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (P.D.B.-J.); (M.L.M.); (M.R.Z.); (S.R.M.); (S.B.-D.)
| | - Michael L. Mallory
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (P.D.B.-J.); (M.L.M.); (M.R.Z.); (S.R.M.); (S.B.-D.)
| | - Mark R. Zweigart
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (P.D.B.-J.); (M.L.M.); (M.R.Z.); (S.R.M.); (S.B.-D.)
| | - Samantha R. May
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (P.D.B.-J.); (M.L.M.); (M.R.Z.); (S.R.M.); (S.B.-D.)
| | - Daniel Kelly
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London WC1E 7HT, UK; (D.K.); (D.J.A.)
| | - Rachel Williams
- Department of Infection, Immunity and Inflammation, UCL Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK; (R.W.); (J.B.)
- Department of Genetics & Genomic Medicine, UCL Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Sylvia Becker-Dreps
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (P.D.B.-J.); (M.L.M.); (M.R.Z.); (S.R.M.); (S.B.-D.)
- Department of Family Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Filemón Bucardo
- Department of Microbiology, National Autonomous University of Nicaragua-León (UNAN-León), León 21000, Nicaragua;
| | - David J. Allen
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London WC1E 7HT, UK; (D.K.); (D.J.A.)
| | - Judith Breuer
- Department of Infection, Immunity and Inflammation, UCL Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK; (R.W.); (J.B.)
- Department of Microbiology, Great Ormond Street Hospital for Children NHS Foundation Trust, London WC1N 3JH, UK
| | - Ralph S. Baric
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (P.D.B.-J.); (M.L.M.); (M.R.Z.); (S.R.M.); (S.B.-D.)
| |
Collapse
|
34
|
Bull JJ, Antia R. Which 'imperfect vaccines' encourage the evolution of higher virulence? Evol Med Public Health 2022; 10:202-213. [PMID: 35539897 PMCID: PMC9081871 DOI: 10.1093/emph/eoac015] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 04/06/2022] [Indexed: 12/27/2022] Open
Abstract
Background and objectives Theory suggests that some types of vaccines against infectious pathogens may lead to the evolution of variants that cause increased harm, particularly when they infect unvaccinated individuals. This theory was supported by the observation that the use of an imperfect vaccine to control Marek's disease virus in chickens resulted in the virus evolving to be more lethal to unvaccinated birds. This raises the concern that the use of some other vaccines may lead to similar pernicious outcomes. We examine that theory with a focus on considering the regimes in which such outcomes are expected. Methodology We evaluate the plausibility of assumptions in the original theory. The previous theory rested heavily on a particular form of transmission-mortality-recovery trade-off and invoked other assumptions about the pathways of evolution. We review alternatives to mortality in limiting transmission and consider evolutionary pathways that were omitted in the original theory. Results The regime where the pernicious evolutionary outcome occurs is narrowed by our analysis but remains possible in various scenarios. We propose a more nuanced consideration of alternative models for the within-host dynamics of infections and for factors that limit virulence. Our analysis suggests imperfect vaccines against many pathogens will not lead to the evolution of pathogens with increased virulence in unvaccinated individuals. Conclusions and implications Evolution of greater pathogen mortality driven by vaccination remains difficult to predict, but the scope for such outcomes appears limited. Incorporation of mechanistic details into the framework, especially regarding immunity, may be requisite for prediction accuracy. Lay Summary A virus of chickens appears to have evolved high mortality in response to a vaccine that merely prevented disease symptoms. Theory has predicted this type of evolution in response to a variety of vaccines and other interventions such as drug treatment. Under what circumstances is this pernicious result likely to occur? Analysis of the theory in light of recent changes in our understanding of viral biology raises doubts that medicine-driven, pernicious evolution is likely to be common. But we are far from a mechanistic understanding of the interaction between pathogen and host that can predict when vaccines and other medical interventions will lead to the unwanted evolution of more virulent pathogens. So, while the regime where a pernicious result obtains may be limited, caution remains warranted in designing many types of interventions.
Collapse
Affiliation(s)
- James J Bull
- Department of Biological Sciences, University of Idaho, Moscow, ID 83844-3051, USA
| | - Rustom Antia
- Department of Biology, Emory University, Atlanta, GA 30322, USA
| |
Collapse
|
35
|
Tohma K, Ford-Siltz LA, Kendra JA, Parra GI. Dynamic immunodominance hierarchy of neutralizing antibody responses to evolving GII.4 noroviruses. Cell Rep 2022; 39:110689. [PMID: 35417705 DOI: 10.1016/j.celrep.2022.110689] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 01/20/2022] [Accepted: 03/24/2022] [Indexed: 02/06/2023] Open
Abstract
A paradigm of RNA viruses is their ability to mutate and escape from herd immunity. Because antibody responses are a major effector for viral immunity, antigenic sites are usually under strong diversifying pressure. Here, we use norovirus as a model to study mechanisms of antigenic diversification of non-enveloped, fast-evolving RNA viruses. We comprehensively characterize all variable antigenic sites involved in virus neutralization and find that single neutralizing monoclonal antibodies (mAbs) map to multiple antigenic sites of GII.4 norovirus. Interactions of multiple epitopes on the viral capsid surface provide a broad mAb-binding repertoire with a remarkable difference in the mAb-binding profiles and immunodominance hierarchy for two distantly related GII.4 variants. Time-ordered mutant viruses confirm a progressive change of antibody immunodominance along with point mutations during the process of norovirus evolution. Thus, in addition to point mutations, switches in immunodominance that redirect immune responses could facilitate immune escape in RNA viruses.
Collapse
Affiliation(s)
- Kentaro Tohma
- Division of Viral Products, Center for Biologics Evaluation and Research, US Food and Drug Administration, 10903 New Hampshire Avenue, Building 52/72, Room 1309, Silver Spring, MD 20993, USA
| | - Lauren A Ford-Siltz
- Division of Viral Products, Center for Biologics Evaluation and Research, US Food and Drug Administration, 10903 New Hampshire Avenue, Building 52/72, Room 1309, Silver Spring, MD 20993, USA
| | - Joseph A Kendra
- Division of Viral Products, Center for Biologics Evaluation and Research, US Food and Drug Administration, 10903 New Hampshire Avenue, Building 52/72, Room 1309, Silver Spring, MD 20993, USA
| | - Gabriel I Parra
- Division of Viral Products, Center for Biologics Evaluation and Research, US Food and Drug Administration, 10903 New Hampshire Avenue, Building 52/72, Room 1309, Silver Spring, MD 20993, USA.
| |
Collapse
|
36
|
Huang Y, Zhou N, Zhang S, Yi Y, Han Y, Liu M, Han Y, Shi N, Yang L, Wang Q, Cui T, Jin H. Norovirus detection in wastewater and its correlation with human gastroenteritis: a systematic review and meta-analysis. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:22829-22842. [PMID: 35048346 PMCID: PMC8769679 DOI: 10.1007/s11356-021-18202-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 12/14/2021] [Indexed: 06/14/2023]
Abstract
Norovirus (NoV) is a major cause of sporadic cases and outbreaks of acute gastroenteritis (AGE), thereby imposing threat to health globally. It is unclear how quantitation of wastewater NoV reflects the incidence of human AGE infections; therefore, we conducted this systematic review and meta-analysis of published NoV wastewater surveillance studies. A literature search was performed, and all studies on NoV wastewater surveillance were identified. Quantitative results were evaluated. The results showed that the overall detection rate of NoV in wastewater was 82.10% (95% confidence interval [CI]: 74.22-89.92%); NoV concentration was statistically significant in terms of season (P < 0.001), with higher concentration in spring and winter. There were positive correlations between NoV GII concentration in wastewater and GII AGE cases (rs = 0.51, 95% CI: 0.18-0.74, I2 = 0%), total AGE cases (rs = 0.40, 95% CI: 0.15-0.61, I2 = 23%) and NoV outbreaks (rs = 0.47, 95% CI: 0.30-0.62, I2 = 0%). Results of cross-correlation analysis of partial data indicated that variations in GII concentration were consistent with or ahead of those in the number of AGE cases. The diversity of NoV genotypes in wastewater was elucidated, and the dominant strains in wastewater showed a consistent temporal distribution with those responsible for human AGE. Our study demonstrated the potential association of NoV detected in wastewater with AGE infections, and further studies are needed to confirm this conclusion.
Collapse
Affiliation(s)
- Yue Huang
- Department of Epidemiology and Health Statistics, School of Public Health, Southeast University, Nanjing, 210009, China
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Nan Zhou
- Department of Epidemiology and Health Statistics, School of Public Health, Southeast University, Nanjing, 210009, China
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Shihan Zhang
- Department of Epidemiology and Health Statistics, School of Public Health, Southeast University, Nanjing, 210009, China
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Youqin Yi
- Department of Epidemiology and Health Statistics, School of Public Health, Southeast University, Nanjing, 210009, China
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Ying Han
- Department of Epidemiology and Health Statistics, School of Public Health, Southeast University, Nanjing, 210009, China
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Minqi Liu
- Department of Epidemiology and Health Statistics, School of Public Health, Southeast University, Nanjing, 210009, China
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Yue Han
- Department of Epidemiology and Health Statistics, School of Public Health, Southeast University, Nanjing, 210009, China
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Naiyang Shi
- Department of Epidemiology and Health Statistics, School of Public Health, Southeast University, Nanjing, 210009, China
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Liuqing Yang
- Department of Epidemiology and Health Statistics, School of Public Health, Southeast University, Nanjing, 210009, China
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Qiang Wang
- Department of Epidemiology and Health Statistics, School of Public Health, Southeast University, Nanjing, 210009, China
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Tingting Cui
- Department of Epidemiology and Health Statistics, School of Public Health, Southeast University, Nanjing, 210009, China
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Hui Jin
- Department of Epidemiology and Health Statistics, School of Public Health, Southeast University, Nanjing, 210009, China.
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China.
| |
Collapse
|
37
|
Ogunsakin RE, Ebenezer O, Ginindza TG. A Bibliometric Analysis of the Literature on Norovirus Disease from 1991-2021. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:ijerph19052508. [PMID: 35270203 PMCID: PMC8909411 DOI: 10.3390/ijerph19052508] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/20/2022] [Accepted: 01/24/2022] [Indexed: 12/12/2022]
Abstract
Norovirus (NoV) is one of the oldest recognized diseases and the leading causal pathogen for acute gastroenteritis (AGE) worldwide. Though numerous studies have been reported on NoV disease, limited research has explored the publication trends in this area. As a result, the objective of this work was to fill the void by conducting a bibliometric study in publication trends on NoV studies as well as discovering the hotspots. The Web of Science central assemblage database was hunted for publications from 1991 to 2021 with “norovirus” in the heading. Microsoft Excel 2016, VOSviewer, R Bibliometrix, and Biblioshiny packages were deployed for the statistical analysis of published research articles. A total of 6021 published documents were identified in the Web of Science database for this thirty-year study period (1991–2021). The analyses disclosed that the Journal of Medical Virology was the leading journal in publications on norovirus studies with a total of 215 published articles, the Journal of Virology was the most cited document with 11,185 total citations. The United States of America (USA) has the most significant productivity in norovirus publications and is the leading country with the highest international collaboration. Analysis of top germane authors discovered that X. Jiang (135) and J. Vinje (119) were the two top relevant authors of norovirus publications. The commonly recognized funders were US and EU-based, with the US emerging as a top funder. This study reveals trends in scientific findings and academic collaborations and serves as a leading-edge model to reveal trends in global research in the field of norovirus research. This study points out the progress status and trends on NoV research. It can help researchers in the medical profession obtain a comprehensive understanding of the state of the art of NoV. It also has reference values for the research and application of the NoV visualization methods. Further, the research map on AGE obtained by our analysis is expected to help researchers efficiently and effectively explore the NoV field.
Collapse
Affiliation(s)
- Ropo E. Ogunsakin
- Discipline of Public Health Medicine, School of Nursing & Public Health, College of Health Sciences, University of KwaZulu-Natal, Private Bag X54001, Durban 4000, South Africa;
- Correspondence:
| | - Oluwakemi Ebenezer
- Department of Chemistry, Faculty of Natural Sciences, Mangosuthu University of Technology, Durban 4000, South Africa;
| | - Themba G. Ginindza
- Discipline of Public Health Medicine, School of Nursing & Public Health, College of Health Sciences, University of KwaZulu-Natal, Private Bag X54001, Durban 4000, South Africa;
- Cancer & Infectious Diseases Epidemiology Research Unit (CIDERU), College of Health Sciences, University of KwaZulu-Natal, Private Bag X54001, Durban 4000, South Africa
| |
Collapse
|
38
|
Abstract
Human noroviruses are the most common viral cause of acute gastroenteritis worldwide. Currently, there are no approved vaccines or specific therapeutics to treat the disease. Some obstacles delaying the development of a norovirus vaccine are: (i) the extreme diversity presented by noroviruses; (ii) our incomplete understanding of immunity to noroviruses; and (iii) the lack of a robust cell culture system or animal model for human noroviruses. Recent advances in in vitro cultivation of norovirus, novel approaches applied to viral genomics and immunity, and completion of vaccine trials and birth cohort studies have provided new information toward a better understanding of norovirus immunity. Here, we will discuss the complex relationship between norovirus diversity and correlates of protection for human noroviruses, and how this information could be used to guide the development of cross-protective vaccines.
Collapse
Affiliation(s)
- Lauren A. Ford-Siltz
- Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, United States
| | - Kentaro Tohma
- Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, United States
| | - Gabriel I. Parra
- Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, United States,CONTACT Gabriel I. Parra Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, 10903 New Hampshire Avenue, Building 52/72, Room 1308, Silver Spring, MD20993, United States
| |
Collapse
|
39
|
Tsai H, Yune P, Rao M. Norovirus disease among older adults. Ther Adv Infect Dis 2022; 9:20499361221136760. [DOI: 10.1177/20499361221136760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 10/17/2022] [Indexed: 11/16/2022] Open
Abstract
Norovirus, a leading cause of gastroenteritis outbreaks worldwide, results in substantial direct and indirect healthcare costs. Adults older than 65 years of age bear a significant proportion of the disease burden, and the disease course in this population is often more severe and protracted. In this narrative review, we discuss the epidemiology of norovirus infection, mechanisms of pathogenesis, and transmission pertinent to outbreaks along with infection prevention and control efforts. We also describe the clinical manifestations of norovirus disease with a focus on individuals older than 65 years of age, diagnosis and available treatment options, and the challenges and progress within vaccine development.
Collapse
Affiliation(s)
- Helen Tsai
- Montefiore Medical Center, Bronx, NY, USA
| | | | - Mana Rao
- Essen Medical Associates, Bronx, NY 10461, USA
- ArchCare, New York, NY, USA
| |
Collapse
|
40
|
Molecular epidemiology of norovirus variants detected in children under five years of age in Hyderabad, India. Indian J Med Microbiol 2021; 40:12-17. [PMID: 34856323 DOI: 10.1016/j.ijmmb.2021.11.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 11/23/2021] [Accepted: 11/26/2021] [Indexed: 11/20/2022]
Abstract
PURPOSE Noroviruses are common viral agents in acute diarrhea in all age groups worldwide. Norovirus has been classified into 10 genogroups, GI to GX with over 48 genotypes among them the GII.4 genotype has evolved over time with a clear pattern of periodic variant replacement. Immunity is strain or genotype specific with little or no protection conferred across genogroups. The present study was aimed to determine the epidemiology, prevalent genotypes of norovirus in children below five years of age in the Hyderabad region, India. METHODS The stool samples and clinical data were collected from 458 children below 5 years of age comprising of cases with acute gastroenteritis (n = 366) and a control group (n = 92) admitted to the pediatric ward. All the samples were tested for Norovirus by ELISA and RT-PCR. Sequencing was done for predominant strains. RESULTS 10.3% (n = 38) of cases and 3.2% (n = 3) of the control group were found to be Norovirus positive. Predominant genotypes were GII-82.5% followed by GI-12.5%. CONCLUSION Sequencing and Phylogenetic analyses of 20 GII.4 strains was done. All of the isolates are clustered away from published the GII.4 variants thus suggesting the appearance of a new variant.
Collapse
|
41
|
O'Reilly KM, Sandman F, Allen D, Jarvis CI, Gimma A, Douglas A, Larkin L, Wong KLM, Baguelin M, Baric RS, Lindesmith LC, Goldstein RA, Breuer J, Edmunds WJ. Predicted norovirus resurgence in 2021-2022 due to the relaxation of nonpharmaceutical interventions associated with COVID-19 restrictions in England: a mathematical modeling study. BMC Med 2021; 19:299. [PMID: 34753508 PMCID: PMC8577179 DOI: 10.1186/s12916-021-02153-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 10/04/2021] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND To reduce the coronavirus disease burden in England, along with many other countries, the government implemented a package of non-pharmaceutical interventions (NPIs) that have also impacted other transmissible infectious diseases such as norovirus. It is unclear what future norovirus disease incidence is likely to look like upon lifting these restrictions. METHODS Here we use a mathematical model of norovirus fitted to community incidence data in England to project forward expected incidence based on contact surveys that have been collected throughout 2020-2021. RESULTS We report that susceptibility to norovirus infection has likely increased between March 2020 and mid-2021. Depending upon assumptions of future contact patterns incidence of norovirus that is similar to pre-pandemic levels or an increase beyond what has been previously reported is likely to occur once restrictions are lifted. Should adult contact patterns return to 80% of pre-pandemic levels, the incidence of norovirus will be similar to previous years. If contact patterns return to pre-pandemic levels, there is a potential for the expected annual incidence to be up to 2-fold larger than in a typical year. The age-specific incidence is similar across all ages. CONCLUSIONS Continued national surveillance for endemic diseases such as norovirus will be essential after NPIs are lifted to allow healthcare services to adequately prepare for a potential increase in cases and hospital pressures beyond what is typically experienced.
Collapse
Affiliation(s)
- Kathleen M O'Reilly
- Centre for Mathematical Modelling of Infectious Diseases, Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, UK
| | - Frank Sandman
- Centre for Mathematical Modelling of Infectious Diseases, Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, UK.,Statistics, Modelling and Economics Department, National Infection Service, Public Health England, London, UK.,NIHR Health Protection Research Unit in Modelling and Health Economics, London School of Hygiene and Tropical Medicine, London, UK
| | - David Allen
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK
| | - Christopher I Jarvis
- Centre for Mathematical Modelling of Infectious Diseases, Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, UK
| | - Amy Gimma
- Centre for Mathematical Modelling of Infectious Diseases, Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, UK
| | - Amy Douglas
- Gastrointestinal Pathogens Unit, National Infection Service, Public Health England, London, UK
| | - Lesley Larkin
- Gastrointestinal Pathogens Unit, National Infection Service, Public Health England, London, UK
| | - Kerry L M Wong
- Centre for Mathematical Modelling of Infectious Diseases, Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, UK
| | - Marc Baguelin
- Centre for Mathematical Modelling of Infectious Diseases, Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, UK.,MRC Centre for Global Infectious Disease Analysis, J-IDEA, Department of Infectious Disease Epidemiology, Imperial College London, St Mary's Campus, London, UK
| | - Ralph S Baric
- Department of Epidemiology, University of North Carolina, Chapel Hill, USA
| | - Lisa C Lindesmith
- Department of Epidemiology, University of North Carolina, Chapel Hill, USA
| | | | - Judith Breuer
- Division of Infection and Immunity, University College London, London, UK.,Department of Microbiology, Virology and Infection Control, Great Ormond Street Hospital for Children, London, UK
| | - W John Edmunds
- Centre for Mathematical Modelling of Infectious Diseases, Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, UK
| |
Collapse
|
42
|
Zhao B, Hu L, Song Y, Patil K, Ramani S, Atmar RL, Estes MK, Prasad BVV. Norovirus Protease Structure and Antivirals Development. Viruses 2021; 13:v13102069. [PMID: 34696498 PMCID: PMC8537771 DOI: 10.3390/v13102069] [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: 09/16/2021] [Revised: 10/06/2021] [Accepted: 10/09/2021] [Indexed: 11/30/2022] Open
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 an excellent target for developing small-molecule inhibitors. The current strategy for developing HuNoV protease inhibitors is by targeting the enzyme’s active site and designing inhibitors that bind to the substrate-binding pockets located near the active site. However, subtle differential conformational flexibility in response to the different substrates in the polyprotein and structural differences in the active site and substrate-binding pockets across different genogroups, hamper the development of effective broad-spectrum inhibitors. A comparative analysis of the available HuNoV protease structures may provide valuable insight for identifying novel strategies for the design and development of such inhibitors. The goal of this review is to provide such analysis together with an overview of the current status of the design and development of HuNoV protease inhibitors.
Collapse
Affiliation(s)
- Boyang Zhao
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA; (B.Z.); (K.P.); (S.R.); (R.L.A.); (M.K.E.)
| | - Liya Hu
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX 77030, USA;
| | - Yongcheng Song
- Department of Pharmacology and Chemical Biology, Baylor College of Medicine, Houston, TX 77030, USA;
| | - Ketki Patil
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA; (B.Z.); (K.P.); (S.R.); (R.L.A.); (M.K.E.)
| | - Sasirekha Ramani
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA; (B.Z.); (K.P.); (S.R.); (R.L.A.); (M.K.E.)
| | - Robert L. Atmar
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA; (B.Z.); (K.P.); (S.R.); (R.L.A.); (M.K.E.)
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Mary K. Estes
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA; (B.Z.); (K.P.); (S.R.); (R.L.A.); (M.K.E.)
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - B. V. Venkataram Prasad
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA; (B.Z.); (K.P.); (S.R.); (R.L.A.); (M.K.E.)
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX 77030, USA;
- Correspondence: ; Tel.: +1-713-798-5686
| |
Collapse
|
43
|
Tenge VR, Hu L, Prasad BVV, Larson G, Atmar RL, Estes MK, Ramani S. Glycan Recognition in Human Norovirus Infections. Viruses 2021; 13:2066. [PMID: 34696500 PMCID: PMC8537403 DOI: 10.3390/v13102066] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 10/07/2021] [Accepted: 10/08/2021] [Indexed: 12/11/2022] Open
Abstract
Recognition of cell-surface glycans is an important step in the attachment of several viruses to susceptible host cells. The molecular basis of glycan interactions and their functional consequences are well studied for human norovirus (HuNoV), an important gastrointestinal pathogen. Histo-blood group antigens (HBGAs), a family of fucosylated carbohydrate structures that are present on the cell surface, are utilized by HuNoVs to initially bind to cells. In this review, we describe the discovery of HBGAs as genetic susceptibility factors for HuNoV infection and review biochemical and structural studies investigating HuNoV binding to different HBGA glycans. Recently, human intestinal enteroids (HIEs) were developed as a laboratory cultivation system for HuNoV. We review how the use of this novel culture system has confirmed that fucosylated HBGAs are necessary and sufficient for infection by several HuNoV strains, describe mechanisms of antibody-mediated neutralization of infection that involve blocking of HuNoV binding to HBGAs, and discuss the potential for using the HIE model to answer unresolved questions on viral interactions with HBGAs and other glycans.
Collapse
Affiliation(s)
- Victoria R. Tenge
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA; (V.R.T.); (B.V.V.P.); (R.L.A.); (M.K.E.)
| | - Liya Hu
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX 77030, USA;
| | - B. V. Venkataram Prasad
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA; (V.R.T.); (B.V.V.P.); (R.L.A.); (M.K.E.)
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX 77030, USA;
| | - Göran Larson
- Department of Laboratory Medicine, University of Gothenburg, SE 413 45 Gothenburg, Sweden;
| | - Robert L. Atmar
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA; (V.R.T.); (B.V.V.P.); (R.L.A.); (M.K.E.)
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Mary K. Estes
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA; (V.R.T.); (B.V.V.P.); (R.L.A.); (M.K.E.)
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Sasirekha Ramani
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA; (V.R.T.); (B.V.V.P.); (R.L.A.); (M.K.E.)
| |
Collapse
|
44
|
Zweigart MR, Becker-Dreps S, Bucardo F, González F, Baric RS, Lindesmith LC. Serological Humoral Immunity Following Natural Infection of Children with High Burden Gastrointestinal Viruses. Viruses 2021; 13:2033. [PMID: 34696463 PMCID: PMC8538683 DOI: 10.3390/v13102033] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 10/01/2021] [Accepted: 10/03/2021] [Indexed: 12/14/2022] Open
Abstract
Acute gastroenteritis (AGE) is a major cause of morbidity and mortality worldwide, resulting in an estimated 440,571 deaths of children under age 5 annually. Rotavirus, norovirus, and sapovirus are leading causes of childhood AGE. A successful rotavirus vaccine has reduced rotavirus hospitalizations by more than 50%. Using rotavirus as a guide, elucidating the determinants, breath, and duration of serological antibody immunity to AGE viruses, as well as host genetic factors that define susceptibility is essential for informing development of future vaccines and improving current vaccine candidates. Here, we summarize the current knowledge of disease burden and serological antibody immunity following natural infection to inform further vaccine development for these three high-burden viruses.
Collapse
Affiliation(s)
- Mark R. Zweigart
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC 27599, USA; (M.R.Z.); (S.B.-D.)
| | - Sylvia Becker-Dreps
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC 27599, USA; (M.R.Z.); (S.B.-D.)
- Department of Family Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Filemón Bucardo
- Department of Microbiology, National Autonomous University of Nicaragua, León 21000, Nicaragua; (F.B.); (F.G.)
| | - Fredman González
- Department of Microbiology, National Autonomous University of Nicaragua, León 21000, Nicaragua; (F.B.); (F.G.)
| | - Ralph S. Baric
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC 27599, USA; (M.R.Z.); (S.B.-D.)
| | - Lisa C. Lindesmith
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC 27599, USA; (M.R.Z.); (S.B.-D.)
| |
Collapse
|
45
|
Haddadin Z, Batarseh E, Hamdan L, Stewart LS, Piya B, Rahman H, Spieker AJ, Chappell J, Wikswo ME, Dunn JR, Payne DC, Vinjé J, Hall AJ, Halasa N. Characteristics of GII.4 Norovirus Versus Other Genotypes in Sporadic Pediatric Infections in Davidson County, Tennessee, USA. Clin Infect Dis 2021; 73:e1525-e1531. [PMID: 32667045 PMCID: PMC8492161 DOI: 10.1093/cid/ciaa1001] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 07/10/2020] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND Norovirus is a leading cause of epidemic acute gastroenteritis (AGE), with most outbreaks occurring during winter. The majority of outbreaks are caused by GII.4 noroviruses; however, data to support whether this is true for sporadic medically attended AGE are limited. Therefore, we sought to compare the clinical characteristics and seasonality of GII.4 vs non-GII.4 viruses. METHODS Children aged 15 days -17 years with AGE symptoms were recruited from the outpatient, emergency department, and inpatient settings at Vanderbilt Children's Hospital, Davidson County, Nashville, Tennessee, from December 2012 -November 2015. Stool specimens were tested using qRT-PCR for GI and GII noroviruses and subsequently genotyped by sequencing a partial region of the capsid gene. RESULTS A total of 3705 patients were enrolled, and stool specimens were collected and tested from 2885 (78%) enrollees. Overall, 636 (22%) samples were norovirus-positive, of which 567 (89%) were GII. Of the 460 (81%) genotyped GII-positive samples, 233 (51%) were typed as GII.4 and 227 (49%) as non-GII.4. Compared with children with non-GII.4 infections, children with GII.4 infections were younger, more likely to have diarrhea, and more likely to receive oral rehydration fluids. Norovirus was detected year-round and peaked during winter. CONCLUSIONS Approximately 40% of sporadic pediatric norovirus AGE cases were caused by GII.4 norovirus. Children infected with GII.4 had more severe symptoms that required more medical care. Seasonal variations were noticed among different genotypes. These data highlight the importance of continuous norovirus surveillance and provide important information on which strains pediatric norovirus vaccines should protect against.
Collapse
Affiliation(s)
- Zaid Haddadin
- Department of Pediatric Infectious Diseases, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Einas Batarseh
- Department of Pediatric Infectious Diseases, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Lubna Hamdan
- Department of Pediatric Infectious Diseases, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Laura S Stewart
- Department of Pediatric Infectious Diseases, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Bhinnata Piya
- Department of Pediatric Infectious Diseases, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Herdi Rahman
- Department of Pediatric Infectious Diseases, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Andrew J Spieker
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - James Chappell
- Department of Pediatric Infectious Diseases, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Mary E Wikswo
- Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - John R Dunn
- Tennessee Department of Health, Nashville, Tennessee, USA
| | - Daniel C Payne
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Jan Vinjé
- Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Aron J Hall
- Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Natasha Halasa
- Department of Pediatric Infectious Diseases, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
46
|
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
|
47
|
Noroviruses-The State of the Art, Nearly Fifty Years after Their Initial Discovery. Viruses 2021; 13:v13081541. [PMID: 34452406 PMCID: PMC8402810 DOI: 10.3390/v13081541] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 07/06/2021] [Accepted: 07/31/2021] [Indexed: 12/11/2022] Open
Abstract
Human noroviruses are recognised as the major global cause of viral gastroenteritis. Here, we provide an overview of notable advances in norovirus research and provide a short recap of the novel model systems to which much of the recent progress is owed. Significant advances include an updated classification system, the description of alternative virus-like protein morphologies and capsid dynamics, and the further elucidation of the functions and roles of various viral proteins. Important milestones include new insights into cell tropism, host and microbial attachment factors and receptors, interactions with the cellular translational apparatus, and viral egress from cells. Noroviruses have been detected in previously unrecognised hosts and detection itself is facilitated by improved analytical techniques. New potential transmission routes and/or viral reservoirs have been proposed. Recent in vivo and in vitro findings have added to the understanding of host immunity in response to norovirus infection, and vaccine development has progressed to preclinical and even clinical trial testing. Ongoing development of therapeutics includes promising direct-acting small molecules and host-factor drugs.
Collapse
|
48
|
Torén K, Schiöler L, Nenonen NP, Hannoun C, Roth A, Andersson LM, Westin J, Bergström T. Risk factors for norovirus infection in healthcare workers during nosocomial outbreaks: a cross-sectional study. Antimicrob Resist Infect Control 2021; 10:107. [PMID: 34294149 PMCID: PMC8299649 DOI: 10.1186/s13756-021-00979-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 07/08/2021] [Indexed: 12/03/2022] Open
Abstract
Background Norovirus outbreaks cause severe medico-socio-economic problems affecting healthcare workers and patients. The aim of the study was to investigate prevalence of norovirus infection and risk factors for infection in healthcare workers during nosocomial outbreaks. Methods A cross-sectional study of norovirus infections in healthcare workers was performed in seven outbreak wards in a large university hospital. Packs (swab for rectal sampling, and questionnaire) were posted to healthcare workers on notification of a ward outbreak. Rectal samples were examined with norovirus-specific real-time PCR. Replies from questionnaires were analysed using logistic regression models with norovirus genogroup (G)II positive findings as dependent variable. The results are expressed as odds ratios (OR) with 95% confidence intervals (CI). Sequencing and phylogenetic analyses (1040 nucleotides) were used to characterize norovirus strains from healthcare workers. Cluster analyses included norovirus GII.4 strains detected in ward patients during the ongoing outbreaks. Results Of 308 packs issued to healthcare workers, 129 (42%) were returned. norovirus GII was detected in 26 healthcare workers (20.2%). Work in cohort care (OR 4.8, 95% CI 1.4–16.3), work in wards for patients with dementia (OR 13.2, 95% CI 1.01–170.7), and having diarrhoea, loose stools or other gastrointestinal symptoms the last week (OR 7.7, 95% CI 2.5–27.2) were associated with increased norovirus prevalence in healthcare workers. Sequencing revealed norovirus GII.4 in healthcare workers samples, and strains detected in healthcare workers and ward patients during a given ward outbreak showed ≥ 99% similarity. Conclusion Norovirus positive findings in healthcare workers were strongly associated with symptomatic infection, close contact with sick patients, and dementia nursing. Supplementary Information The online version contains supplementary material available at 10.1186/s13756-021-00979-8.
Collapse
Affiliation(s)
- Kjell Torén
- School of Public Health and Community Medicine, Institute of Medicine, University of Gothenburg, Box 414, 405 30, Gothenburg, Sweden. .,Department of Occupational and Environmental Medicine, Sahlgrenska University Hospital, Gothenburg, Sweden.
| | - Linus Schiöler
- School of Public Health and Community Medicine, Institute of Medicine, University of Gothenburg, Box 414, 405 30, Gothenburg, Sweden
| | - Nancy P Nenonen
- Department of Infectious Diseases/Virology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Charles Hannoun
- Department of Infectious Diseases/Virology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Anette Roth
- Department of Infectious Diseases/Virology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Lars-Magnus Andersson
- Department of Infectious Diseases/Virology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Johan Westin
- Department of Infectious Diseases/Virology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Tomas Bergström
- Department of Infectious Diseases/Virology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
49
|
Alvarado G, Salmen W, Ettayebi K, Hu L, Sankaran B, Estes MK, Venkataram Prasad BV, Crowe JE. Broadly cross-reactive human antibodies that inhibit genogroup I and II noroviruses. Nat Commun 2021; 12:4320. [PMID: 34262046 PMCID: PMC8280134 DOI: 10.1038/s41467-021-24649-w] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 06/29/2021] [Indexed: 12/14/2022] Open
Abstract
The rational development of norovirus vaccine candidates requires a deep understanding of the antigenic diversity and mechanisms of neutralization of the virus. Here, we isolate and characterize a panel of broadly cross-reactive naturally occurring human monoclonal IgMs, IgAs and IgGs reactive with human norovirus (HuNoV) genogroup I or II (GI or GII). We note three binding patterns and identify monoclonal antibodies (mAbs) that neutralize at least one GI or GII HuNoV strain when using a histo-blood group antigen (HBGA) blocking assay. The HBGA blocking assay and a virus neutralization assay using human intestinal enteroids reveal that the GII-specific mAb NORO-320, mediates HBGA blocking and neutralization of multiple GII genotypes. The Fab form of NORO-320 neutralizes GII.4 infection more potently than the mAb, however, does not block HBGA binding. The crystal structure of NORO-320 Fab in complex with GII.4 P-domain shows that the antibody recognizes a highly conserved region in the P-domain distant from the HBGA binding site. Dynamic light scattering analysis of GII.4 virus-like particles with mAb NORO-320 shows severe aggregation, suggesting neutralization is by steric hindrance caused by multivalent cross-linking. Aggregation was not observed with the Fab form of NORO-320, suggesting that this clone also has additional inhibitory features.
Collapse
Affiliation(s)
- Gabriela Alvarado
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Wilhelm Salmen
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Khalil Ettayebi
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Liya Hu
- The Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Banumathi Sankaran
- Berkeley Center for Structural Biology, Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley Laboratory, Berkeley, CA, USA
| | - Mary K Estes
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
- Department of Medicine-Gastroenterology and Hepatology, Baylor College of Medicine, Houston, TX, USA
| | - B V Venkataram Prasad
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA.
- The Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX, USA.
| | - James E Crowe
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA.
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN, USA.
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
50
|
Misumi M, Nishiura H. Long-term dynamics of Norovirus transmission in Japan, 2005-2019. PeerJ 2021; 9:e11769. [PMID: 34306831 PMCID: PMC8280881 DOI: 10.7717/peerj.11769] [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: 12/10/2020] [Accepted: 06/22/2021] [Indexed: 11/20/2022] Open
Abstract
Norovirus continues to evolve, adjusting its pathogenesis and transmissibility. In the present study, we systematically collected datasets on Norovirus outbreaks in Japan from 2005 to 2019 and analyzed time-dependent changes in the asymptomatic ratio, the probability of virus detection, and the probability of infection given exposure. Reports of 1,728 outbreaks were published, and feces from all involved individuals, including those with asymptomatic infection, were tested for virus in 434 outbreaks. We found that the outbreak size did not markedly change over this period, but the variance in outbreak size increased during the winter (November–April). Assuming that natural history parameters did not vary over time, the asymptomatic ratio, the probability of virus detection, and the probability of infection given exposure were estimated to be 18.6%, 63.3% and 84.5%, respectively. However, a model with time-varying natural history parameters yielded better goodness-of-fit and suggested that the asymptomatic ratio varied by year. The asymptomatic ratio was as high as 25.8% for outbreaks caused by genotype GII.4 noroviruses. We conclude that Norovirus transmissibility has not changed markedly since 2005, and that yearly variation in the asymptomatic ratio could potentially be explained by the circulating dominant genotype.
Collapse
Affiliation(s)
- Megumi Misumi
- Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan.,Rumoi City Hospital, Rumoi, Hokkaido, Japan
| | - Hiroshi Nishiura
- Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan.,School of Public Health, Kyoto University, Kyoto, Japan
| |
Collapse
|