1
|
Chen J, Cheng Z, Chen J, Qian L, Wang H, Liu Y. Advances in human norovirus research: Vaccines, genotype distribution and antiviral strategies. Virus Res 2024; 350:199486. [PMID: 39428038 DOI: 10.1016/j.virusres.2024.199486] [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: 05/03/2024] [Revised: 10/01/2024] [Accepted: 10/17/2024] [Indexed: 10/22/2024]
Abstract
Norovirus, belonging to the Caliciviridae family, is a non-enveloped, positive-sense single-stranded RNA virus. It is widely acknowledged as a significant etiological agent responsible for non-bacterial acute gastroenteritis and considered a major cause thereof. Norovirus is primarily tranmitted via fecal-oral route, but can also be transmitted via airborne routes. Clinical manifestations often include symptoms associated with acute gastroenteritis, like nausea, vomiting, watery diarrhea, stomach cramps, and others. Due to the specific pathogenic mechanism of the virus, and genomic diversity, there are currently no preventive vaccines or effective antiviral drugs available for treating norovirus-induced acute gastroenteritis infections. The management of such infections mainly relies on oral rehydration therapy while prevention necessitates adherence to personal hygiene measures. The present paper discusses the nature, transmission route, clinical manifestations, immune response mechanism, and vaccine research of Norovirus. The objective of this review manuscript is to systematically gather, analyze, and summarize recent research and investigations on norovirus in order to enhance our understanding of its characteristics and pathogenesis. This not only facilitates subsequent researchers in acquiring a more expedited and comprehensive grasp of the existing knowledge about norovirus but also provides clearer directions and goals for future studies.
Collapse
Affiliation(s)
- JunLi Chen
- Department of Laboratory Medicine, Department of Microbiology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212003,PR China
| | - ZhengChao Cheng
- Department of Laboratory Medicine, Department of Microbiology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212003,PR China
| | - Jing Chen
- Department of Laboratory Medicine, Department of Microbiology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212003,PR China
| | - Lingling Qian
- Central laboratory of Changshu Medicine Examination Institute, Changshu, Jiangsu 215500, PR China.
| | - Haoran Wang
- Department of Laboratory Medicine, Department of Microbiology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212003,PR China.
| | - YuWei Liu
- Department of Laboratory Medicine, Department of Microbiology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212003,PR China.
| |
Collapse
|
2
|
Alberer M, Moe CL, Hatz C, Kling K, Kirby AE, Lindsay L, Nothdurft HD, Riera-Montes M, Steffen R, Verstraeten T, Wu HM, DuPont HL. Norovirus acute gastroenteritis amongst US and European travellers to areas of moderate to high risk of travellers' diarrhoea: a prospective cohort study. J Travel Med 2024; 31:taad051. [PMID: 37074164 PMCID: PMC11500662 DOI: 10.1093/jtm/taad051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 03/21/2023] [Accepted: 04/04/2023] [Indexed: 04/20/2023]
Abstract
BACKGROUND Acute gastroenteritis (AGE) is a major medical condition for travellers worldwide, particularly travellers to low- and middle-income countries. Norovirus (NoV) is the most common cause of viral AGE in older children and adults, but data on prevalence and impact amongst travellers is limited. METHODS Prospective, multi-site, observational cohort study conducted 2015-2017, amongst adult international travellers from the US and Europe to areas of moderate to high risk of travel-acquired AGE. Participants provided self-collected pre-travel stool samples and self-reported AGE symptoms whilst travelling. Post-travel stool samples were requested from symptomatic subjects and a sample of asymptomatic travellers within 14 days of return. Samples were tested for NoV by RT-qPCR, genotyped if positive and tested for other common enteric pathogens by Luminex xTAG GPP. RESULTS Of the 1109 participants included, 437 (39.4%) developed AGE symptoms resulting in an overall AGE incidence of 24.7 per 100 person-weeks [95% confidence interval (CI): 22.4; 27.1]. In total, 20 NoV-positive AGE cases (5.2% of those tested) were identified at an incidence of 1.1 per 100 person-weeks (95% CI: 0.7; 1.7). NoV-positive samples belonged mostly to genogroup GII (18, 85.7%); None of the 13 samples sequenced belonged to genotype GII.4. Clinical severity of AGE was higher for NoV-positive than for NoV-negative cases (mean modified Vesikari Score 6.8 vs 4.9) with more cases classified as severe or moderate (25% vs 6.8%). In total, 80% of NoV-positive participants (vs 38.9% in NoV-negative) reported at least moderate impact on travel plans. CONCLUSIONS AGE is a prevalent disease amongst travellers with a small proportion associated with NoV. Post-travel stool sample collection timing might have influenced the low number of NoV cases detected; however, NoV infections resulted in high clinical severity and impact on travel plans. These results may contribute to targeted vaccine development and the design of future studies on NoV epidemiology.
Collapse
Affiliation(s)
- Martin Alberer
- Division of Infectious Diseases and Tropical Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Christine L Moe
- Hubert Department of Global Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Christoph Hatz
- Epidemiology, Biostatistics and Prevention Institute, WHO Collaborating Center for Travellers’ Health, University of Zurich, Zurich, Switzerland
| | - Kerstin Kling
- Epidemiology, Biostatistics and Prevention Institute, WHO Collaborating Center for Travellers’ Health, University of Zurich, Zurich, Switzerland
| | - Amy E Kirby
- Hubert Department of Global Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Lisa Lindsay
- P95 Pharmacovigilance and Epidemiology Services, Leuven, Belgium
| | - Hans D Nothdurft
- Division of Infectious Diseases and Tropical Medicine, University Hospital, LMU Munich, Munich, Germany
| | | | - Robert Steffen
- Epidemiology, Biostatistics and Prevention Institute, WHO Collaborating Center for Travellers’ Health, University of Zurich, Zurich, Switzerland
- Division of Epidemiology, Human Genetics & Environmental Sciences, University of Texas School of Public Health, Houston, TX, USA
| | | | - Henry M Wu
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Herbert L DuPont
- Kelsey Research Foundation, Houston, TX, USA
- School of Public Health, Center for Infectious Diseases, and McGovern Medical School, Department of Internal Medicine, University of Texas–Houston Health Science Center, Houston, TX, USA
| |
Collapse
|
3
|
Zhan X, Li Q, Tian P, Wang D. The attachment factors and attachment receptors of human noroviruses. Food Microbiol 2024; 123:104591. [PMID: 39038896 DOI: 10.1016/j.fm.2024.104591] [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: 04/14/2024] [Revised: 06/06/2024] [Accepted: 06/29/2024] [Indexed: 07/24/2024]
Abstract
Human noroviruses (HuNoVs) are the leading etiological agent causing the worldwide outbreaks of acute epidemic non-bacterial gastroenteritis. Histo-blood group antigens (HBGAs) are commonly acknowledged as cellular receptors or co-receptors for HuNoVs. However, certain genotypes of HuNoVs cannot bind with any HBGAs, suggesting potential additional co-factors and attachment receptors have not been identified yet. In addition, food items, such as oysters and lettuce, play an important role in the transmission of HuNoVs. In the past decade, a couple of attachment factors other than HBGAs have been identified and analyzed from foods and microbiomes. Attachment factors exhibit potential as inhibitors of viral binding to receptors on host cells. Therefore, it is imperative to further characterize the attachment factors for HuNoVs present in foods to effectively control the spread of HuNoVs within the food chain. This review summarizes the potential attachment factors/receptors of HuNoVs in humans, foods, and microbiome.
Collapse
Affiliation(s)
- Xiangjun Zhan
- Department of Food Science and Technology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Qianqian Li
- Department of Bioengineering, Shanghai Institute of Technology, Shanghai, 201418, China
| | - Peng Tian
- Produce Safety and Microbiology Research Unit, Western Regional Research Center, Agricultural Research Service-United States Department of Agriculture, Albany, CA, 94706, USA
| | - Dapeng Wang
- Department of Food Science and Technology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, 200240, China.
| |
Collapse
|
4
|
Nyblade C, Yuan L. Virus Shedding and Diarrhea: A Review of Human Norovirus Genogroup II Infection in Gnotobiotic Pigs. Viruses 2024; 16:1432. [PMID: 39339908 PMCID: PMC11437449 DOI: 10.3390/v16091432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 09/01/2024] [Accepted: 09/04/2024] [Indexed: 09/30/2024] Open
Abstract
For nearly twenty years, gnotobiotic (Gn) pigs have been used as a model of human norovirus (HuNoV) infection and disease. Unique in their ability to develop diarrhea and shed virus post oral challenge, Gn pigs have since been used to evaluate the infectivity of several genogroup II HuNoV strains. Nearly all major pandemic GII.4 variants have been tested in Gn pigs, with varying rates of infectivity. Some induce an asymptomatic state despite being shed in large quantities in stool, and others induce high incidence of both diarrhea and virus shedding. Non-GII.4 strains, including GII.12 and GII.6, have also been evaluated in Gn pigs. Again, rates of diarrhea and virus shedding tend to vary between studies. Several factors may influence these findings, including age, dosage, biological host factors, or bacterial presence. The impact of these factors is nuanced and requires further evaluation to elucidate the exact mechanisms behind increases or decreases in infection rates. Regardless, the value of Gn pig models in HuNoV research cannot be understated, and the model will surely continue to contribute to the field in years to come.
Collapse
Affiliation(s)
- Charlotte Nyblade
- Department of Biomedical Sciences and Pathobiology, Virginia Polytechnic and State University, Blacksburg, VA 24061, USA
| | - Lijuan Yuan
- Department of Biomedical Sciences and Pathobiology, Virginia Polytechnic and State University, Blacksburg, VA 24061, USA
| |
Collapse
|
5
|
Wang Y, Gao Z, Lu Q, Liu B, Jia L, Shen L, Tian Y, Li W, Yan H, Zhang D, Yang P, Fang L, Wang Q, Cui F. Transmissibility quantification of norovirus outbreaks in 2016-2021 in Beijing, China. J Med Virol 2023; 95:e29153. [PMID: 37804388 DOI: 10.1002/jmv.29153] [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: 07/11/2023] [Revised: 09/08/2023] [Accepted: 09/25/2023] [Indexed: 10/09/2023]
Abstract
The transmissibility is a crucial feature for norovirus, yet its quantitative estimation has been limited. Our objective was to estimate the basic reproduction number (R0 ) of norovirus and investigate its variation characteristics. Norovirus outbreaks reported from September 2016 to August 2021 in Beijing were analyzed. The susceptible-infected-removed compartment model was established to estimate R0 . Linear regression models and logistic regression models were used to explore the factors affecting the transmissibility of norovirus. The overall median R0 of norovirus was estimated as 2.1 (interquartile range [IQR] 1.8-2.5), with 650 norovirus outbreaks. The transmissibility of norovirus varied by year, outbreak setting and genotype. The R0 of norovirus during September 2019 to August 2020 (median 2.1, IQR 1.8-2.4) and September 2020 to August 2021 (median 2.0, IQR 1.7-2.3) was lower than that of September 2016 to August 2017 (median 2.3, IQR 1.8-2.7) (β = 0.94, p = 0.05; β = 0.93, p = 0.008). The R0 of norovirus for all other settings was lower than that for kindergarten (median 2.4, IQR 2.0-2.9) (primary school: median 2.0, IQR 1.7-2.4, β = 0.94, p = 0.001; secondary school: median 1.7, IQR 1.5-2.0, β = 0.87, p < 0.001; college: median 1.7, IQR 1.5-1.8, β = 0.89, p = 0.03; other closed settings: median 1.8, IQR 1.5-2.0, β = 0.90, p = 0.004). GⅡ.2[P16] outbreaks had a median R0 of 2.2 (IQR 1.8-2.7), which was higher than that for GⅡ.6[P7] outbreaks (median 1.8, IQR: 1.8-2.0, odds ratio = 0.19, p = 0.03; GⅡ.2[P16] as reference) and mixed-genotype outbreaks (median 1.7, IQR: 1.5-1.8, β = 0.92, p = 0.02; mixed-genotype as reference). In kindergartens and primary schools, norovirus shows increased transmissibility, emphasizing the vulnerable population and high-risk settings. Furthermore, the transmissibility of norovirus may change over time and with virus evolution, necessitating additional research to uncover the underlying mechanisms.
Collapse
Affiliation(s)
- Yu Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
- Institute for Infectious Disease and Endemic Disease Control, Beijing Center for Disease Prevention and Control, Beijing, China
| | - Zhiyong Gao
- Institute for Infectious Disease and Endemic Disease Control, Beijing Center for Disease Prevention and Control, Beijing, China
| | - Qingbin Lu
- Department of Laboratory Science and Technology & Vaccine Research Center, School of Public Health, Peking University, Beijing, China
| | - Baiwei Liu
- Institute for Infectious Disease and Endemic Disease Control, Beijing Center for Disease Prevention and Control, Beijing, China
| | - Lei Jia
- Institute for Infectious Disease and Endemic Disease Control, Beijing Center for Disease Prevention and Control, Beijing, China
| | - Lingyu Shen
- Institute for Infectious Disease and Endemic Disease Control, Beijing Center for Disease Prevention and Control, Beijing, China
| | - Yi Tian
- Institute for Infectious Disease and Endemic Disease Control, Beijing Center for Disease Prevention and Control, Beijing, China
| | - Weihong Li
- Institute for Infectious Disease and Endemic Disease Control, Beijing Center for Disease Prevention and Control, Beijing, China
| | - Hanqiu Yan
- Institute for Infectious Disease and Endemic Disease Control, Beijing Center for Disease Prevention and Control, Beijing, China
| | - Daitao Zhang
- Institute for Infectious Disease and Endemic Disease Control, Beijing Center for Disease Prevention and Control, Beijing, China
| | - Peng Yang
- Institute for Infectious Disease and Endemic Disease Control, Beijing Center for Disease Prevention and Control, Beijing, China
| | - Liqun Fang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Quanyi Wang
- Institute for Infectious Disease and Endemic Disease Control, Beijing Center for Disease Prevention and Control, Beijing, China
| | - Fuqiang Cui
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
- Department of Laboratory Science and Technology & Vaccine Research Center, School of Public Health, Peking University, Beijing, China
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China
| |
Collapse
|
6
|
Masciopinto C. Extension of probability models of the risk of infections by human enteric viruses. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2023; 20:17499-17519. [PMID: 37920063 DOI: 10.3934/mbe.2023777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/04/2023]
Abstract
This study presents a novel approach for obtaining reliable models and coefficients to estimate the probability of infection caused by common human enteric viruses. The aim is to provide guidance for public health policies in disease prevention and control, by reducing uncertainty and management costs in health risk assessments. Conventional dose-response (DR) models, based on the theory elaborated by Furumoto and Mickey [1], exhibit limitations stemming from the heterogeneity of individual host susceptibilities to infection resulting from ingesting aggregate viruses. Moreover, the scarcity of well-designed viral challenge experiments contributes to significant uncertainty in these DR models. To address these issues, we conducted a review of infection models used in health risk analysis, focusing on Norovirus (NoV) GI.1, pooled Enterovirus group (EV), Poliovirus 1/SM, and Echo-12 virus via contaminated water or food. Using a mechanistic approach, we reevaluated the known DR models and coefficients for the probability of individual host infection in the mentioned viruses based on dose-infection challenge experiments. Specifically, we sought to establish a relationship between the minimum infectious dose (ID) and the ID having a 50% probability of initiating host infection in the same challenge experiment. Furthermore, we developed a new formula to estimate the degree of aggregation of GI.1 NoV at the mean infectious dose. The proposed models, based on "exact" beta-Poisson DR models, effectively predicted infection probabilities from ingestion of both disaggregated and aggregate NoV GI.1. Through a numerical evaluation, we compared the results with the maximum likelihood estimation (MLE) probability obtained from a controlled challenge trial with the NoV GI.1 virus described in the literature, demonstrating the accuracy of our approach. By addressing the indetermination of the unmeasured degree of NoV aggregation in each single infectious dose, our models reduce overestimations and uncertainties in microbial risk assessments. This improvement enhances the management of health risks associated with enteric virus infections.
Collapse
Affiliation(s)
- Costantino Masciopinto
- Consiglio Nazionale delle Ricerche, Istituto di Ricerca Sulle Acque, Bari viale F. De Blasio 5, 70132 Italia
| |
Collapse
|
7
|
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
|
8
|
Deb S, Mondal R, Lahiri D, Shome G, Roy AG, Sarkar V, Sarkar S, Benito-León J. Norovirus-associated neurological manifestations: summarizing the evidence. J Neurovirol 2023; 29:492-506. [PMID: 37477790 PMCID: PMC10501950 DOI: 10.1007/s13365-023-01152-0] [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: 02/18/2023] [Revised: 06/06/2023] [Accepted: 06/14/2023] [Indexed: 07/22/2023]
Abstract
Norovirus, a positive-stranded RNA virus, is one of the leading causes of acute gastroenteritis among all age groups worldwide. The neurological manifestations of norovirus are underrecognized, but several wide-spectrum neurological manifestations have been reported among infected individuals in the last few years. Our objective was to summarize the features of norovirus-associated neurological disorders based on the available literature. We used the existing PRISMA consensus statement. Data were collected from PubMed, EMBASE, Web of Science, and Scopus databases up to Jan 30, 2023, using pre-specified searching strategies. Twenty-one articles were selected for the qualitative synthesis. Among these, seven hundred and seventy-four patients with norovirus-associated neurological manifestations were reported. Most cases were seizure episodes, infection-induced encephalopathy, and immune-driven disorders. However, only a few studies have addressed the pathogenesis of norovirus-related neurological complications. The pathogenesis of these manifestations may be mediated by either neurotropism or aberrant immune-mediated injury, or both, depending on the affected system. Our review could help clinicians to recognize these neurological manifestations better and earlier while deepening the understanding of the pathogenesis of this viral infection.
Collapse
Affiliation(s)
- Shramana Deb
- Department of Neuroscience, S.N. Pradhan Centre for Neuroscience, Kolkata, India
| | - Ritwick Mondal
- Department of Clinical Pharmacology and Therapeutic Medicine, IPGMER and SSKM Hospital, Kolkata, India
| | - Durjoy Lahiri
- Department of Cognitive Neurology, Baycrest Health Sciences and Rotman Research Institute, University of Toronto, Ontario, Canada
| | - Gourav Shome
- Division of Molecular Medicine, Bose Institute, Kolkata, India
| | - Aakash Guha Roy
- Department of Internal Medicine, SSKM and IPGMER Hospital, Kolkata, India
| | - Vramanti Sarkar
- Department of Neuroscience, S.N. Pradhan Centre for Neuroscience, Kolkata, India
| | - Shramana Sarkar
- Department of Internal Medicine, SSKM and IPGMER Hospital, Kolkata, India
| | - Julián Benito-León
- Department of Neurology, University Hospital “12 de Octubre”, Madrid, Spain
- Research Institute (i+12), University Hospital “12 de Octubre”, Madrid, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
- Department of Medicine, Complutense University, Madrid, Spain
| |
Collapse
|
9
|
Yu Z, Shao Q, Xu Z, Chen C, Li M, Jiang Y, Cheng D. Immunogenicity and Blocking Efficacy of Norovirus GII.4 Recombinant P Protein Vaccine. Vaccines (Basel) 2023; 11:1053. [PMID: 37376442 DOI: 10.3390/vaccines11061053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 05/24/2023] [Accepted: 05/30/2023] [Indexed: 06/29/2023] Open
Abstract
Noroviruses (NoVs) are the main cause of acute gastroenteritis in all ages worldwide. The aim of this study was to produce the recombinant P protein of norovirus and to demonstrate its blocking effect. In this study, the engineered strains were induced to express the P protein of NoVs GII.4, which was identified using SDS-PAGE and ELISA as having the capacity to bind to histo-blood group antigens (HBGAs). Rabbits were immunized to obtain neutralizing antibodies. ELISA and ISC-RT-qPCR were used to determine the blocking efficacy of the neutralizing antibody to human norovirus (HuNoV) and murine norovirus (MNV). The recombinant P protein (35 KD) was obtained, and the neutralizing antibody was successfully prepared. The neutralizing antibody could block the binding of the P protein and HuNoV to HBGAs. Neutralizing antibodies can also block MNV invasion into host cells RAW264.7. The recombinant P protein expressed in E. coli can induce antibodies to block HuNoV and MNV. The recombinant P protein of NoVs GII.4 has the value of vaccine development.
Collapse
Affiliation(s)
- Zhendi Yu
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Qingyi Shao
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Zhangkai Xu
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Chenghao Chen
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Mingfan Li
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Yi Jiang
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Dongqing Cheng
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou 310053, China
| |
Collapse
|
10
|
Minimal Antigenic Evolution after a Decade of Norovirus GII.4 Sydney_2012 Circulation in Humans. J Virol 2023; 97:e0171622. [PMID: 36688654 PMCID: PMC9973034 DOI: 10.1128/jvi.01716-22] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Norovirus is a major human pathogen that can cause severe gastroenteritis in vulnerable populations. The extensive viral diversity presented by human noroviruses constitutes a major roadblock for the development of effective vaccines. In addition to the large number of genotypes, antigenically distinct variants of GII.4 noroviruses have chronologically emerged over the last 3 decades. The last variant to emerge, Sydney_2012, has been circulating at high incidence worldwide for over a decade. We analyzed 1449 capsid sequences from GII.4 Sydney_2012 viruses to determine genetic changes indicative of antigenic diversification. Phylogenetic analyses show that Sydney_2012 viruses scattered within the tree topology with no single cluster dominating during a given year or geographical location. Fourteen residues presented high variability, 7 of which mapped to 4 antigenic sites. Notably, ~52% of viruses presented mutations at 2 or more antigenic sites. Mutational patterns showed that residues 297 and 372, which map to antigenic site A, changed over time. Virus-like particles (VLPs) developed from wild-type Sydney_2012 viruses and engineered to display all mutations detected at antigenic sites were tested against polyclonal sera and monoclonal antibodies raised against Sydney_2012 and Farmington_Hills_2002 VLPs. Minimal changes in reactivity were detected with polyclonal sera and only 4 MAbs lost binding, with all mapping to antigenic site A. Notably, reversion of residues from Sydney_2012 reconstituted epitopes from ancestral GII.4 variants. Overall, this study demonstrates that, despite circulating for over a decade, Sydney_2012 viruses present minimal antigenic diversification and provides novel insights on the diversification of GII.4 noroviruses that could inform vaccine design. IMPORTANCE GII.4 noroviruses are the major cause of acute gastroenteritis in all age groups. This predominance has been attributed to the continued emergence of phylogenetically discrete variants that escape immune responses to previous infections. The last GII.4 variant to emerge, Sydney_2012, has been circulating at high incidence for over a decade, raising the question of whether this variant is undergoing antigenic diversification without presenting a major distinction at the phylogenetic level. Sequence analyses that include >1400 capsid sequences from GII.4 Sydney_2012 showed changes in 4 out of the 6 major antigenic sites. Notably, while changes were detected in one of the most immunodominant sites over time, these resulted in minimal changes in the antigenic profile of these viruses. This study provides new insights on the mechanism governing the antigenic diversification of GII.4 norovirus that could help in the development of cross-protective vaccines to human noroviruses.
Collapse
|
11
|
Functional and structural characterization of Norovirus GII.6 in recognizing histo-blood group antigens. Virol Sin 2023; 38:56-65. [PMID: 36216242 PMCID: PMC10006186 DOI: 10.1016/j.virs.2022.09.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 07/04/2022] [Indexed: 11/11/2022] Open
Abstract
Noroviruses (NoVs) are the primary cause of acute gastroenteritis worldwide. Histo-blood group antigens (HBGAs) are receptors or attachment factors that affect the prevalence and host susceptibility of NoVs. GII.6 NoV is one of the predominant genotypes in humans, which recognizes the type ABO secretor of HBGAs. However, the structural basis of GII.6 NoV's interaction with HBGAs receptors remains elusive. In this study, we investigated the binding features of the GII.6 strain to HBGAs using saliva- and glycan-ELISA assays and characterized the molecular basis of the GII.6 virus that recognizes H disaccharide. We showed that the GII.6 P domain recognized some A and O secretor's saliva samples, most B secretor's saliva samples, and H disaccharide antigen, but did not bind non-secretors' saliva. Further, we determined the crystal structures of GII.6 and its complex with H disaccharides at 1.7 Å, revealing that the P domain of GII.6 shares the conventional binding interface and mode of GII HBGAs. Single residue mutations at the GII.6-H binding sites could inhibit the binding of GII.6 to HBGAs, demonstrating that the interaction residues were crucial in maintaining NoV-glycan integrity. Finally, structural and sequence analyses showed that the major residues of the GII.6-H interaction were conserved among NoVs in the GII genogroup. Taken together, our study characterized the functional and structural features of GII.6 that allow it to interact with HBGAs, and shed light on NoV evolution, epidemiology, and anti-viral drug development.
Collapse
|
12
|
Rouphael N, Beck A, Kirby AE, Liu P, Natrajan MS, Lai L, Phadke V, Winston J, Raabe V, Collins MH, Girmay T, Alvarez A, Beydoun N, Karmali V, Altieri-Rivera J, Lindesmith LC, Anderson EJ, Wang Y, El-Khorazaty J, Petrie C, Baric RS, Baqar S, Moe CL, Mulligan MJ. Dose-Response of a Norovirus GII.2 Controlled Human Challenge Model Inoculum. J Infect Dis 2022; 226:1771-1780. [PMID: 35137154 PMCID: PMC9650503 DOI: 10.1093/infdis/jiac045] [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: 10/02/2021] [Accepted: 02/03/2022] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Genogroup II noroviruses are the most common cause of acute infectious gastroenteritis. We evaluated the use of a new GII.2 inoculum in a human challenge. METHODS Forty-four healthy adults (36 secretor-positive and 8 secretor-negative for histo-blood group antigens) were challenged with ascending doses of a new safety-tested Snow Mountain virus (SMV) GII.2 norovirus inoculum (1.2 × 104 to 1.2 × 107 genome equivalent copies [GEC]; n = 38) or placebo (n = 6). Illness was defined as diarrhea and/or vomiting postchallenge in subjects with evidence of infection (defined as GII.2 norovirus RNA detection in stool and/or anti-SMV immunoglobulin G [IgG] seroconversion). RESULTS The highest dose was associated with SMV infection in 90%, and illness in 70% of subjects with 10 of 12 secretor-positive (83%) and 4 of 8 secretor-negative (50%) becoming ill. There was no association between prechallenge anti-SMV serum IgG concentration, carbohydrate-binding blockade antibody, or salivary immunoglobulin A and infection. The median infectious dose (ID50) was 5.1 × 105 GEC. CONCLUSIONS High rates of infection and illness were observed in both secretor-positive and secretor-negative subjects in this challenge study. However, a high dose will be required to achieve the target of 75% illness to make this an efficient model for evaluating potential norovirus vaccines and therapeutics. CLINICAL TRIALS REGISTRATION NCT02473224.
Collapse
Affiliation(s)
- Nadine Rouphael
- Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, School of Medicine, Emory University, Atlanta, Georgia, USA
| | - Allison Beck
- Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, School of Medicine, Emory University, Atlanta, Georgia, USA
| | - Amy E Kirby
- Rollins School of Public Health, Emory University, Atlanta, Georgia, USA
| | - Pengbo Liu
- Rollins School of Public Health, 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
| | - Lilin Lai
- Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, School of Medicine, Emory University, Atlanta, Georgia, USA
| | - Varun Phadke
- Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, School of Medicine, Emory University, Atlanta, Georgia, USA
| | - Juton Winston
- Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, School of Medicine, Emory University, Atlanta, Georgia, USA
| | - Vanessa Raabe
- New York University Grossman School of Medicine and New York University Vaccine Center, New York, New York, 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
| | - Tigisty Girmay
- Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, School of Medicine, Emory University, Atlanta, Georgia, USA
| | - Alicarmen Alvarez
- Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, School of Medicine, Emory University, Atlanta, Georgia, USA
| | - Nour Beydoun
- Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, School of Medicine, Emory University, Atlanta, Georgia, USA
| | - Vinit Karmali
- Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, School of Medicine, Emory University, Atlanta, Georgia, USA
| | - Joanne Altieri-Rivera
- Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, School of Medicine, Emory University, Atlanta, Georgia, USA
| | - Lisa C Lindesmith
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Evan J Anderson
- Division of Infectious Diseases, Department of Pediatrics, School of Medicine, Emory University, Atlanta, Georgia, USA
| | - Yuke Wang
- Rollins School of Public Health, Emory University, Atlanta, Georgia, USA
| | | | - Carey Petrie
- The Emmes Company, LLC, Rockville, Maryland, USA
| | - Ralph S Baric
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Shahida Baqar
- Division of Microbiology and Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Christine L Moe
- Rollins School of Public Health, Emory University, Atlanta, Georgia, USA
| | - Mark J Mulligan
- New York University Grossman School of Medicine and New York University Vaccine Center, New York, New York, USA
| |
Collapse
|
13
|
Saikia K, Saharia N, Singh CS, Borah PP, Namsa ND. Association of histo-blood group antigens and predisposition to gastrointestinal diseases. J Med Virol 2022; 94:5149-5162. [PMID: 35882942 DOI: 10.1002/jmv.28028] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 06/26/2022] [Accepted: 07/23/2022] [Indexed: 12/15/2022]
Abstract
Infectious gastroenteritis is a common illness afflicting people worldwide. The two most common etiological agents of viral gastroenteritis, rotavirus and norovirus are known to recognize histo-blood group antigens (HBGAs) as attachment receptors. ABO, Lewis, and secretor HBGAs are distributed abundantly on mucosal epithelia, red blood cell membranes, and also secreted in biological fluids, such as saliva, intestinal content, milk, and blood. HBGAs are fucosylated glycans that have been implicated in the attachment of some enteric pathogens such as bacteria, parasites, and viruses. Single nucleotide polymorphisms in the genes encoding ABO (H), fucosyltransferase gene FUT2 (Secretor/Se), FUT3 (Lewis/Le) have been associated with changes in enzyme expression and HBGAs production. The highly polymorphic HBGAs among different populations and races influence genotype-specific susceptibility or resistance to enteric pathogens and its epidemiology, and vaccination seroconversion. Therefore, there is an urgent need to conduct population-based investigations to understand predisposition to enteric infections and gastrointestinal diseases. This review focuses on the relationship between HBGAs and predisposition to common human gastrointestinal illnesses caused by viral, bacterial, and parasitic agents.
Collapse
Affiliation(s)
- Kasturi Saikia
- Department of Molecular Biology and Biotechnology, Tezpur University, Napaam, Assam, India
| | - Niruprabha Saharia
- Department of Paediatrics, Tezpur Medical College and Hospital, Bihaguri, Tezpur, Assam, India
| | - Chongtham S Singh
- Department of Paediatrics, Regional Institute of Medical Sciences, Imphal, India
| | - Partha P Borah
- Department of Paediatrics and Neonatology, Pratiksha Hospital, Guwahati, Assam, India
| | - Nima D Namsa
- Department of Molecular Biology and Biotechnology, Tezpur University, Napaam, Assam, India.,Centre for Multi-disciplinary Research, Tezpur University, Napaam, Assam, India
| |
Collapse
|
14
|
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
|
15
|
Deere D, Ryan U. Current assumptions for quantitative microbial risk assessment (QMRA) of Norovirus contamination of drinking water catchments due to recreational activities: an update. JOURNAL OF WATER AND HEALTH 2022; 20:1543-1557. [PMID: 36308498 DOI: 10.2166/wh.2022.114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Contamination of drinking water from Norovirus (NoV) and other waterborne viruses is a major public health concern globally. Increasingly, quantitative microbial risk assessment (QMRA) is being used to assess the various risks from waterborne pathogens and evaluate control strategies. As urban populations grow and expand, there is increasing demand for recreational activities in drinking water catchments. QMRA relies on context-specific data to map out the pathways by which viruses can enter water and be transferred to drinking water consumers and identify risk factors and appropriate controls. This review examines the current evidence base and assumptions for QMRA analysis of NoV and other waterborne viral pathogens and recommends numerical values based on the most recent evidence to better understand the health risks associated with recreators in Australian drinking water sources; these are broadly applicable to all drinking water sources where recreational access is allowed. Key issues include the lack of an agreed upon data and dose-response models for human infectious NoV genotypes, faecal shedding by bathers, the extent of NoV infectivity and aggregation, resistance (secretor status) to NoV and the extent of secondary transmission.
Collapse
Affiliation(s)
- Dan Deere
- Water Futures and Water Research Australia, Sydney, Australia
| | - Una Ryan
- Harry Butler Institute, Murdoch University, Perth, Australia E-mail:
| |
Collapse
|
16
|
Choy RKM, Bourgeois AL, Ockenhouse CF, Walker RI, Sheets RL, Flores J. Controlled Human Infection Models To Accelerate Vaccine Development. Clin Microbiol Rev 2022; 35:e0000821. [PMID: 35862754 PMCID: PMC9491212 DOI: 10.1128/cmr.00008-21] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The timelines for developing vaccines against infectious diseases are lengthy, and often vaccines that reach the stage of large phase 3 field trials fail to provide the desired level of protective efficacy. The application of controlled human challenge models of infection and disease at the appropriate stages of development could accelerate development of candidate vaccines and, in fact, has done so successfully in some limited cases. Human challenge models could potentially be used to gather critical information on pathogenesis, inform strain selection for vaccines, explore cross-protective immunity, identify immune correlates of protection and mechanisms of protection induced by infection or evoked by candidate vaccines, guide decisions on appropriate trial endpoints, and evaluate vaccine efficacy. We prepared this report to motivate fellow scientists to exploit the potential capacity of controlled human challenge experiments to advance vaccine development. In this review, we considered available challenge models for 17 infectious diseases in the context of the public health importance of each disease, the diversity and pathogenesis of the causative organisms, the vaccine candidates under development, and each model's capacity to evaluate them and identify correlates of protective immunity. Our broad assessment indicated that human challenge models have not yet reached their full potential to support the development of vaccines against infectious diseases. On the basis of our review, however, we believe that describing an ideal challenge model is possible, as is further developing existing and future challenge models.
Collapse
Affiliation(s)
- Robert K. M. Choy
- PATH, Center for Vaccine Innovation and Access, Seattle, Washington, USA
| | - A. Louis Bourgeois
- PATH, Center for Vaccine Innovation and Access, Seattle, Washington, USA
| | | | - Richard I. Walker
- PATH, Center for Vaccine Innovation and Access, Seattle, Washington, USA
| | | | - Jorge Flores
- PATH, Center for Vaccine Innovation and Access, Seattle, Washington, USA
| |
Collapse
|
17
|
The Importance of Secretor-Status in Norovirus Infection Following Allogeneic Hematopoietic Stem Cell Transplantation. Viruses 2022; 14:v14071350. [PMID: 35891335 PMCID: PMC9318794 DOI: 10.3390/v14071350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 06/09/2022] [Accepted: 06/16/2022] [Indexed: 12/10/2022] Open
Abstract
Background. Human secretor-status is a strong susceptibility factor for norovirus infection in immunocompetent people. The predominant norovirus genotype GII.4 almost exclusively infects secretors and is also associated with more severe symptoms. However, it is not known to what extent this also applies to immunocompromised individuals. Our objective was to determine the importance of secretor-status and norovirus genotype for the susceptibility and/or the clinical course of norovirus infection in allogeneic hematopoietic stem cell transplant (HCT) patients. Methods: This was a retrospective study of 89 HCT patients diagnosed with norovirus infection. Secretor-status and norovirus genotype were determined using stored extracted DNA or blood (n = 89) and fecal samples (n = 22), respectively. Results: Seven of eighty-nine (8%) of the patients were secretor-negative, a small proportion compared to the expected rate of at least 20% non-secretors in the general Swedish population. Among the genotyped samples, norovirus genotype GII.4 was predominant (n = 12) and only detected in secretor-positive individuals. Patients with norovirus GII.4 had a median symptom duration of 36 (3–681) days compared to 15 (1–94) days in patients infected with other norovirus genotypes (n = 10, p = 0.1). Conclusions: The results suggest that secretor-status affects the susceptibility to norovirus infection even when the immune system is severely compromised. The norovirus genotype may also be a risk factor for chronic norovirus symptoms in immunocompromised patients.
Collapse
|
18
|
Han Q, Xue Z, Tan M, Wang L, Chen H, Zhang R. Bovine natural antibody IgM inhibits the binding of human norovirus protruding domain to its HBGA receptors. FEBS Open Bio 2022; 12:1489-1497. [PMID: 35674188 PMCID: PMC9340781 DOI: 10.1002/2211-5463.13450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 05/28/2022] [Accepted: 06/07/2022] [Indexed: 11/16/2022] Open
Abstract
Human norovirus (HuNoV) is the primary viral pathogen that causes acute gastroenteritis (AGE) in humans. The protruding (P) domain of HuNoV interacts with cell surface histo‐blood group antigens (HBGAs) to initiate infection. Owing to the lack of an effective in vitro culture method and a robust animal model, our understanding of HuNoVs is limited, and as a result, there are no commercial vaccines or antivirals available at present against the virus. In an attempt to develop a preventative measure, we previously identified that bovine colostrum (bCM) contains functional factors that inhibit the binding of HuNoV P domain to its HBGA receptors. In this study, a candidate functional factor in bCM was identified as immunoglobulin M (IgM) using mass spectrometry, followed by database comparison. The natural antibody IgM was further verified to be a functional protein that inhibited HuNoV P protein binding to HBGA receptors through receptor‐binding inhibition experiments using bCM, commercial IgM, and fetal bovine serum. Our findings provide a foundation for future development of natural IgM into an antiviral drug, which may help to prevent and/or treat HuNoV infection.
Collapse
Affiliation(s)
- Qi Han
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Zhaolei Xue
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Ming Tan
- Division of Infectious Diseases, Cincinnati Children's Hospital Medical Center, OH, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, OH, USA
| | - Likai Wang
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Huiling Chen
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Ran Zhang
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| |
Collapse
|
19
|
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
|
20
|
Peña-Gil N, Santiso-Bellón C, Gozalbo-Rovira R, Buesa J, Monedero V, Rodríguez-Díaz J. The Role of Host Glycobiology and Gut Microbiota in Rotavirus and Norovirus Infection, an Update. Int J Mol Sci 2021; 22:13473. [PMID: 34948268 PMCID: PMC8704558 DOI: 10.3390/ijms222413473] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 12/13/2021] [Accepted: 12/13/2021] [Indexed: 12/12/2022] Open
Abstract
Rotavirus (RV) and norovirus (NoV) are the leading causes of acute gastroenteritis (AGE) worldwide. Several studies have demonstrated that histo-blood group antigens (HBGAs) have a role in NoV and RV infections since their presence on the gut epithelial surfaces is essential for the susceptibility to many NoV and RV genotypes. Polymorphisms in genes that code for enzymes required for HBGAs synthesis lead to secretor or non-secretor and Lewis positive or Lewis negative individuals. While secretor individuals appear to be more susceptible to RV infections, regarding NoVs infections, there are too many discrepancies that prevent the ability to draw conclusions. A second factor that influences enteric viral infections is the gut microbiota of the host. In vitro and animal studies have determined that the gut microbiota limits, but in some cases enhances enteric viral infection. The ways that microbiota can enhance NoV or RV infection include virion stabilization and promotion of virus attachment to host cells, whereas experiments with microbiota-depleted and germ-free animals point to immunoregulation as the mechanism by which the microbiota restrict infection. Human trials with live, attenuated RV vaccines and analysis of the microbiota in responder and non-responder individuals also allowed the identification of bacterial taxa linked to vaccine efficacy. As more information is gained on the complex relationships that are established between the host (glycobiology and immune system), the gut microbiota and intestinal viruses, new avenues will open for the development of novel anti-NoV and anti-RV therapies.
Collapse
Affiliation(s)
- Nazaret Peña-Gil
- Department of Microbiology, School of Medicine, University of Valencia, Avda. Blasco Ibáñez 17, 46010 Valencia, Spain; (N.P.-G.); (C.S.-B.); (R.G.-R.); (J.B.)
| | - Cristina Santiso-Bellón
- Department of Microbiology, School of Medicine, University of Valencia, Avda. Blasco Ibáñez 17, 46010 Valencia, Spain; (N.P.-G.); (C.S.-B.); (R.G.-R.); (J.B.)
| | - Roberto Gozalbo-Rovira
- Department of Microbiology, School of Medicine, University of Valencia, Avda. Blasco Ibáñez 17, 46010 Valencia, Spain; (N.P.-G.); (C.S.-B.); (R.G.-R.); (J.B.)
| | - Javier Buesa
- Department of Microbiology, School of Medicine, University of Valencia, Avda. Blasco Ibáñez 17, 46010 Valencia, Spain; (N.P.-G.); (C.S.-B.); (R.G.-R.); (J.B.)
| | - Vicente Monedero
- Department of Biotechnology, Institute of Agrochemistry and Food Technology (IATA-CSIC), 46980 Paterna, Spain;
| | - Jesús Rodríguez-Díaz
- Department of Microbiology, School of Medicine, University of Valencia, Avda. Blasco Ibáñez 17, 46010 Valencia, Spain; (N.P.-G.); (C.S.-B.); (R.G.-R.); (J.B.)
| |
Collapse
|
21
|
Hong X, Xue L, Liao Y, Wu A, Jiang Y, Kou X. Association of fucosyltransferase 2 gene with norovirus infection: A systematic review and meta-analysis. INFECTION, GENETICS AND EVOLUTION : JOURNAL OF MOLECULAR EPIDEMIOLOGY AND EVOLUTIONARY GENETICS IN INFECTIOUS DISEASES 2021; 96:105091. [PMID: 34610432 DOI: 10.1016/j.meegid.2021.105091] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 08/24/2021] [Accepted: 09/21/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND Norovirus is a leading cause of viral gastroenteritis outbreaks worldwide. Histo-blood group antigens (HBGAs) are important host attachment factors in susceptibility to norovirus. In this study, the association of FUT2 gene, which participates in the biosynthesis of HBGAs, with norovirus infection has been investigated. METHODS All relevant studies on the associations of FUT2 gene with norovirus were retrieved from PubMed, Web of Science, Embase, and Cochrane Library databases. Odds ratios (ORs) and 95% confidence interval (CI) were used to analyze the extracted data. I2 statistic, sensitivity analysis and publication bias analysis were used to confirm the findings. Subgroup analyses were performed for races, genotypes, development degree of the countries, publication years, age and setting when heterogeneity was recorded. RESULTS Twenty studies including 4066 participants were included for the meta-analysis. This analysis showed that there is a significant association between FUT2 gene and norovirus infection (OR = 3.02, 95%CI = 2.00-4.55, P < 0.001). Additionally, the ORs of norovirus infection among Chinese (OR = 4.49, 95%CI = 2.37-8.50, P < 0.001) were higher than those among Caucasian (OR = 3.23, 95%CI = 2.20-4.74, P < 0.001). CONCLUSIONS The meta-analysis suggested that FUT2 gene is associated with susceptibility to norovirus infection.
Collapse
Affiliation(s)
- Xiaojing Hong
- KingMed school of Laboratory Medicine of Guangzhou Medical University, Guangzhou, China
| | - Liang Xue
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Science, China.
| | - Yingyin Liao
- KingMed school of Laboratory Medicine of Guangzhou Medical University, Guangzhou, China
| | - Aiwu Wu
- KingMed school of Laboratory Medicine of Guangzhou Medical University, Guangzhou, China
| | - Yueting Jiang
- Department of Laboratory Medicine, First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| | - Xiaoxia Kou
- KingMed school of Laboratory Medicine of Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
22
|
van Loben Sels JM, Meredith LW, Sosnovtsev SV, de Graaf M, Koopmans MP, Lindesmith LC, Baric RS, Green KY, Goodfellow IG. A luciferase-based approach for measuring HBGA blockade antibody titers against human norovirus. J Virol Methods 2021; 297:114196. [PMID: 34019938 PMCID: PMC9924141 DOI: 10.1016/j.jviromet.2021.114196] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 05/13/2021] [Accepted: 05/17/2021] [Indexed: 01/14/2023]
Abstract
BACKGROUND Noroviruses are the most common cause of viral gastroenteritis worldwide, yet there is a deficit in the understanding of protective immunity. Surrogate neutralization assays have been widely used that measure the ability of antibodies to block virus-like particle (VLP) binding to histo-blood group antigens (HBGAs). However, screening large sample sets against multiple antigens using the traditional HBGA blocking assay requires significant investment in terms of time, equipment, and technical expertise, largely associated with the generation of purified VLPs. METHODS To address these issues, a luciferase immunoprecipitation system (LIPS) assay was modified to measure the norovirus-specific HBGA blockade activity of antibodies. The assay (designated LIPS-Blockade) was validated using a panel of well-characterized homotypic and heterotypic hyperimmune sera as well as strain-specific HBGA blocking monoclonal antibodies. RESULTS The LIPS-Blockade assay was comparable in specificity to a standard HBGA blocking protocol performed with VLPs. Using time-ordered patient sera, the luciferase-based approach was also able to detect changes in HBGA blocking titers following viral challenge and natural infection with norovirus. CONCLUSION In this study we developed a rapid, robust, and scalable surrogate neutralization assay for noroviruses that circumvented the need for purified VLPs. This LIPS-Blockade assay should streamline the process of large-scale immunological studies, ultimately aiding in the characterization of protective immunity to human noroviruses.
Collapse
Affiliation(s)
- Jessica M. van Loben Sels
- Division of Virology, Department of Pathology, University of Cambridge, Cambridge, CB2 2QQ UK,Caliciviruses Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, DHHS, Bethesda, MD, 20892 USA
| | - Luke W. Meredith
- Division of Virology, Department of Pathology, University of Cambridge, Cambridge, CB2 2QQ UK
| | - Stanislav V. Sosnovtsev
- Caliciviruses Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, DHHS, Bethesda, MD, 20892 USA
| | - Miranda de Graaf
- Department of Viroscience, Erasmus University Medical Center, 3015 CN, Rotterdam, the Netherlands.
| | - Marion P.G. Koopmans
- Department of Viroscience, Erasmus University Medical Center, 3015 CN Rotterdam, NL
| | - Lisa C. Lindesmith
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC, 27599 USA
| | - Ralph S. Baric
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC, 27599 USA
| | - Kim Y. Green
- Caliciviruses Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, DHHS, Bethesda, MD, 20892 USA,Corresponding author at: Dr. Kim Y. Green, Caliciviruses Section, LID/DIR/NIAID, National Institutes of Health (NIH), Building 50, Room 6318, 50 South Drive, Bethesda, MD 20892 USA –
| | - Ian G. Goodfellow
- Division of Virology, Department of Pathology, University of Cambridge, Cambridge, CB2 2QQ UK
| |
Collapse
|
23
|
Gao J, Xue L, Liang Y, Wang L, He F, Meng L, Cai W, Zhang J, Wang J, Ye Q, Wu S, Gu Q, Wu Q. Receptor profile and immunogenicity of the non-epidemic norovirus GII.8 variant. Virus Res 2021; 306:198603. [PMID: 34662679 DOI: 10.1016/j.virusres.2021.198603] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 09/22/2021] [Accepted: 10/07/2021] [Indexed: 11/26/2022]
Abstract
Noroviruses are causative agents of acute nonbacterial gastroenteritis epidemics worldwide. There are various genotypes, among which the non-epidemic genotype GII.8 can cause norovirus outbreaks. We previously demonstrated that the immunogenicity of GII.8 differed from that of epidemic variants. This study aimed to comprehensively compare the receptor profile and immunogenicity of the GII.8 variant with those of the epidemic variants. Using the baculovirus-insect cell expression system, we observed that recombinant capsid protein VP1 of the norovirus GII.8 GZ2017-L601 strain formed virus-like particles (VLPs) with a diameter of approximately 30 nm, as evidenced by transmission electron microscopy analysis. The GII.8 VLPs showed weak or moderate binding with all secretor histo-blood group antigens (HBGAs), but not with non-secretors, as evidenced by the HBGA-VLP binding test. The GII.8 VLP antiserum obtained from immunized BALB/c mice was tested for cross-reactivity with other norovirus genotypes (n = 28). The results showed that this antiserum demonstrated moderate cross-reactivity with GI.1, GII.3, and GII.15; however, no cross-reactivity with the epidemic variants of GII.2, GII.4, and GII.17 was observed. Additionally, the blocking-antibody activity of GII.8 antisera against GII.4 VLP-HBGAs and GII.17 VLP-HBGAs interactions and the cross-blocking of GII.8 VLP-HBGAs interactions by GI.1 and GII.4 antisera were evaluated using the HBGAs-VLP blocking test. However, no cross-blocking effect was observed. In summary, the characterization of norovirus GII.8 VLPs and derived antisera revealed that the GII.8 immunogenicity differed from that of epidemic variants.
Collapse
Affiliation(s)
- Junshan Gao
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Liang Xue
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China.
| | - Yanhui Liang
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Linping Wang
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Fenglan He
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Luobing Meng
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Weicheng Cai
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Jumei Zhang
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Juan Wang
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Qinghua Ye
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Shi Wu
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Qihui Gu
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Qingping Wu
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China.
| |
Collapse
|
24
|
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: 6.3] [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
|
25
|
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: 1.7] [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
|
26
|
Glud HA, George S, Skovgaard K, Larsen LE. Zoonotic and reverse zoonotic transmission of viruses between humans and pigs. APMIS 2021; 129:675-693. [PMID: 34586648 PMCID: PMC9297979 DOI: 10.1111/apm.13178] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 08/28/2021] [Indexed: 12/30/2022]
Abstract
Humans and pigs share a close contact relationship, similar biological traits, and one of the highest estimated number of viruses compared to other mammalian species. The contribution and directionality of viral exchange between humans and pigs remain unclear for some of these viruses, but their transmission routes are important to characterize in order to prevent outbreaks of disease in both host species. This review collects and assesses the evidence to determine the likely transmission route of 27 viruses between humans and pigs.
Collapse
Affiliation(s)
- Helena Aagaard Glud
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Sophie George
- Department of Veterinary and Animal Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Kerstin Skovgaard
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Lars Erik Larsen
- Department of Veterinary and Animal Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
27
|
Steele MK, Wikswo ME, Hall AJ, Koelle K, Handel A, Levy K, Waller LA, Lopman BA. Characterizing Norovirus Transmission from Outbreak Data, United States. Emerg Infect Dis 2021; 26:1818-1825. [PMID: 32687043 PMCID: PMC7392428 DOI: 10.3201/eid2608.191537] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Norovirus is the leading cause of acute gastroenteritis outbreaks in the United States. We estimated the basic (R0) and effective (Re) reproduction numbers for 7,094 norovirus outbreaks reported to the National Outbreak Reporting System (NORS) during 2009–2017 and used regression models to assess whether transmission varied by outbreak setting. The median R0 was 2.75 (interquartile range [IQR] 2.38–3.65), and median Re was 1.29 (IQR 1.12–1.74). Long-term care and assisted living facilities had an R0 of 3.35 (95% CI 3.26–3.45), but R0 did not differ substantially for outbreaks in other settings, except for outbreaks in schools, colleges, and universities, which had an R0 of 2.92 (95% CI 2.82–3.03). Seasonally, R0 was lowest (3.11 [95% CI 2.97–3.25]) in summer and peaked in fall and winter. Overall, we saw little variability in transmission across different outbreaks settings in the United States.
Collapse
|
28
|
Reyes Y, González F, Gutierrez L, Blandon P, Centeno E, Zepeda O, Toval-Ruíz C, Lindesmith LC, Baric RS, Vielot N, Diez-Valcarce M, Vinjé J, Svensson L, Becker-Dreps S, Nordgren J, Bucardo F. Secretor status strongly influences the incidence of symptomatic norovirus infection in a genotype-dependent manner in a Nicaraguan birth cohort. J Infect Dis 2021; 225:105-115. [PMID: 34129046 DOI: 10.1093/infdis/jiab316] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 06/10/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND The role of histo-blood group on the burden and severity of norovirus gastroenteritis in young infants has not been well documented. METHODS Norovirus gastroenteritis was assessed in 443 Nicaraguan children followed from birth until 3 years of age. Stool samples were tested for norovirus by RT-qPCR and histo-blood group antigens (HBGA) were determined by phenotyping of saliva and blood. Hazards ratios (95% CI) and predictors of norovirus AGE outcome stratified by HBGA were estimated using Cox proportional hazards models. RESULTS Of 1,353 AGE episodes experienced by children, 229 (17%) tested positive for norovirus with an overall incidence of 21.9/100 child-years. Secretor children were infected as early as 2 months old and had a higher incidence of norovirus GII compared to non-secretor children (15.4 vs 4.1/100 child-years, P = 0.006). Furthermore, all GII.4 AGE episodes occurred in secretor children. Children infected with GI (adjusted OR=0.09, 95% CI 0.02-0.33) or non-GII.4 viruses (adjusted OR=0.2, 95% CI: 0.07-0.6) were less likely to have severe AGE compared to GII.4 infected children. CONCLUSION Secretor status in children strongly influences the incidence of symptomatic norovirus infection in a genogroup or genotype-dependent manner and provides evidence that clinical severity in children depends on norovirus genotypes.
Collapse
Affiliation(s)
- Yaoska Reyes
- Department of Microbiology and Parasitology, National Autonomous University of Nicaragua - León, León, Nicaragua.,Division of Molecular Medicine and Virology, Linköping University, Linköping, Sweden
| | - Fredman González
- Department of Microbiology and Parasitology, National Autonomous University of Nicaragua - León, León, Nicaragua
| | - Lester Gutierrez
- Department of Microbiology and Parasitology, National Autonomous University of Nicaragua - León, León, Nicaragua
| | - Patricia Blandon
- Department of Microbiology and Parasitology, National Autonomous University of Nicaragua - León, León, Nicaragua
| | - Edwing Centeno
- Department of Microbiology and Parasitology, National Autonomous University of Nicaragua - León, León, Nicaragua
| | - Omar Zepeda
- Department of Microbiology and Parasitology, National Autonomous University of Nicaragua - León, León, Nicaragua
| | - Christian Toval-Ruíz
- Department of Microbiology and Parasitology, National Autonomous University of Nicaragua - León, León, Nicaragua
| | - Lisa C Lindesmith
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Ralph S Baric
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Nadja Vielot
- Department of Family Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Marta Diez-Valcarce
- Centers for Disease Control and Prevention, Division of Viral Diseases, Atlanta, GA, United States of America.,Rollins School of Public Health, Emory University, Atlanta, Georgia, United States of America
| | - Jan Vinjé
- Centers for Disease Control and Prevention, Division of Viral Diseases, Atlanta, GA, United States of America
| | - Lennart Svensson
- Division of Molecular Medicine and Virology, Linköping University, Linköping, Sweden.,Division of Medicine, Karolinska Institute, Sweden
| | - Sylvia Becker-Dreps
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA.,Department of Family Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Johan Nordgren
- Division of Molecular Medicine and Virology, Linköping University, Linköping, Sweden
| | - Filemón Bucardo
- Department of Microbiology and Parasitology, National Autonomous University of Nicaragua - León, León, Nicaragua
| |
Collapse
|
29
|
Kondapi DS, Ramani S, Estes MK, Atmar RL, Okhuysen PC. Norovirus in Cancer Patients: A Review. Open Forum Infect Dis 2021; 8:ofab126. [PMID: 34189156 PMCID: PMC8232388 DOI: 10.1093/ofid/ofab126] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 03/10/2021] [Indexed: 12/15/2022] Open
Abstract
Norovirus (NoV) is the leading cause of viral-related diarrhea in cancer patients, in whom it can be chronic, contributing to decreased quality of life, interruption of cancer care, malnutrition, and altered mucosal barrier function. Immunosuppressed cancer patients shed NoV for longer periods of time than immunocompetent hosts, favoring quasispecies development and emergence of novel NoV variants. While nucleic acid amplification tests (NAATs) for NoV diagnosis have revolutionized our understanding of NoV burden of disease, not all NAATs provide information on viral load or infecting genotype. There is currently no effective antiviral or vaccine for chronic NoV infections. Screening for inhibitors of NoV replication in intestinal organoid culture models and creation of NoV-specific adoptive T cells are promising new strategies to develop treatments for chronic NoV in immunosuppressed patients. Herein we summarize data on the epidemiology, clinical manifestations, diagnostic challenges, and treatment of NoV infection in patients with cancer.
Collapse
Affiliation(s)
- Divya Samantha Kondapi
- Department of Infectious Diseases, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Section of Infectious Diseases, Baylor College of Medicine, Houston, Texas, USA
| | - Sasirekha Ramani
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Mary K Estes
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Robert L Atmar
- Section of Infectious Diseases, Baylor College of Medicine, Houston, Texas, USA
| | - Pablo C Okhuysen
- Infection Control and Employee Health, Division of Internal Medicine, Department of Infectious Diseases, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
30
|
Baay M, Neels P. Controlled Human Infection to Speed Up SARS-CoV-2 Vaccine Development. Front Immunol 2021; 12:658783. [PMID: 33777056 PMCID: PMC7994256 DOI: 10.3389/fimmu.2021.658783] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 02/25/2021] [Indexed: 12/20/2022] Open
Affiliation(s)
- Marc Baay
- P95 Epidemiology & Pharmacovigilance, Leuven, Belgium
| | - Pieter Neels
- International Alliance for Biological Standardization—IABS, Geneva, Switzerland
| |
Collapse
|
31
|
Rossouw E, Brauer M, Meyer P, du Plessis NM, Avenant T, Mans J. Virus Etiology, Diversity and Clinical Characteristics in South African Children Hospitalised with Gastroenteritis. Viruses 2021; 13:v13020215. [PMID: 33573340 PMCID: PMC7911269 DOI: 10.3390/v13020215] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/26/2021] [Accepted: 01/27/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Viral gastroenteritis remains a major cause of hospitalisation in young children. This study aimed to determine the distribution and diversity of enteric viruses in children ≤5 years, hospitalised with gastroenteritis at Kalafong Provincial Tertiary Hospital, Pretoria, South Africa, between July 2016 and December 2017. METHODS Stool specimens (n = 205) were screened for norovirus GI and GII, rotavirus, sapovirus, astrovirus and adenovirus by multiplex RT-PCR. HIV exposure and FUT2 secretor status were evaluated. Secretor status was determined by FUT2 genotyping. RESULTS At least one gastroenteritis virus was detected in 47% (96/205) of children. Rotavirus predominated (46/205), followed by norovirus (32/205), adenovirus (15/205), sapovirus (9/205) and astrovirus (3/205). Norovirus genotypes GI.3, GII.2, GII.3, GII.4, GII.7, GII.12, GII.21, and rotavirus strains G1P[8], G2P[4], G2P[6], G3P[4], G3P[8], G8P[4], G8P[6], G9P[6], G9P[8] and sapovirus genotypes GI.1, GI.2, GII.1, GII.4, GII.8 were detected; norovirus GII.4[P31] and rotavirus G3P[4] predominated. Asymptomatic norovirus infection (GI.3, GI.7, GII.4, GII.6, GII.13) was detected in 22% of 46 six-week follow up stools. HIV exposure (30%) was not associated with more frequent or severe viral gastroenteritis hospitalisations compared to unexposed children. Rotavirus preferentially infected secretor children (p = 0.143) and norovirus infected 78% secretors and 22% non-secretors. CONCLUSION Rotavirus was still the leading cause of gastroenteritis hospitalisations, but norovirus caused more severe symptoms.
Collapse
Affiliation(s)
- Esmari Rossouw
- Department of Medical Virology, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa;
| | - Marieke Brauer
- Immunology Laboratory, Ampath, Pretoria 0001, South Africa;
| | - Pieter Meyer
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa or
- National Health Laboratory Service, Tshwane Academic Division, Pretoria 0001, South Africa
| | - Nicolette M. du Plessis
- Department of Paediatrics, Kalafong Provincial Tertiary Hospital, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa; (N.M.d.P.); (T.A.)
| | - Theunis Avenant
- Department of Paediatrics, Kalafong Provincial Tertiary Hospital, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa; (N.M.d.P.); (T.A.)
| | - Janet Mans
- Department of Medical Virology, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa;
- Correspondence: ; Tel.: +27-12-319-2660
| |
Collapse
|
32
|
Bernstein DI, Atmar RL, Hoft DF. Influenza Challenge Models: Ready for Prime Time? Clin Infect Dis 2020; 71:3012-3013. [DOI: 10.1093/cid/ciaa278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- David I Bernstein
- Cincinnati Children’s Hospital, University of Cincinnati, Cincinnati, Ohio, USA
| | - Robert L Atmar
- Departments of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Daniel F Hoft
- Division of Infectious Diseases, Allergy and Immunology, Department of Internal Medicine, Saint Louis University, St. Louis, Missouri, USA
| |
Collapse
|
33
|
Detection of Norovirus in Saliva Samples from Acute Gastroenteritis Cases and Asymptomatic Subjects: Association with Age and Higher Shedding in Stool. Viruses 2020; 12:v12121369. [PMID: 33266188 PMCID: PMC7761458 DOI: 10.3390/v12121369] [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] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 11/26/2020] [Accepted: 11/28/2020] [Indexed: 12/11/2022] Open
Abstract
Norovirus infections are a leading cause of acute gastroenteritis outbreaks worldwide and across all age groups, with two main genogroups (GI and GII) infecting humans. The aim of our study was to investigate the occurrence of norovirus in saliva samples from individuals involved in outbreaks of acute gastroenteritis in closed and semiclosed institutions, and its relationship with the virus strain, virus shedding in stool, the occurrence of symptoms, age, and the secretor status of the individual. Epidemiological and clinical information was gathered from norovirus outbreaks occurring in Catalonia, Spain during 2017–2018, and stool and saliva samples were collected from affected and exposed resident individuals and workers. A total of 347 saliva specimens from 25 outbreaks were analyzed. Further, 84% of individuals also provided a paired stool sample. For GII infections, norovirus was detected in 17.9% of saliva samples from symptomatic cases and 5.2% of asymptomatic individuals. Positivity in saliva occurred in both secretors and nonsecretors. None of the individuals infected by norovirus GI was positive for the virus in saliva. Saliva positivity did not correlate with any of the studied symptoms but did correlate with age ≥ 65 years old. Individuals who were positive in saliva showed higher levels of virus shedding in stool. Mean viral load in positive saliva was 3.16 ± 1.08 log10 genome copies/mL, and the predominance of encapsidated genomes was confirmed by propidium monoazide (PMA)xx-viability RTqPCR assay. The detection of norovirus in saliva raises the possibility of oral-to-oral norovirus transmission during the symptomatic phase and, although to a lesser extent, even in cases of asymptomatic infections.
Collapse
|
34
|
Evaluation of the 50% Infectious Dose of Human Norovirus Cin-2 in Gnotobiotic Pigs: A Comparison of Classical and Contemporary Methods for Endpoint Estimation. Viruses 2020; 12:v12090955. [PMID: 32872283 PMCID: PMC7552045 DOI: 10.3390/v12090955] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 08/27/2020] [Indexed: 01/03/2023] Open
Abstract
Human noroviruses (HuNoVs) are the leading causative agents of epidemic and sporadic acute gastroenteritis that affect people of all ages worldwide. However, very few dose–response studies have been carried out to determine the median infectious dose of HuNoVs. In this study, we evaluated the median infectious dose (ID50) and diarrhea dose (DD50) of the GII.4/2003 variant of HuNoV (Cin-2) in the gnotobiotic pig model of HuNoV infection and disease. Using various mathematical approaches (Reed–Muench, Dragstedt–Behrens, Spearman–Karber, logistic regression, and exponential and approximate beta-Poisson dose–response models), we estimated the ID50 and DD50 to be between 2400–3400 RNA copies, and 21,000–38,000 RNA copies, respectively. Contemporary dose–response models offer greater flexibility and accuracy in estimating ID50. In contrast to classical methods of endpoint estimation, dose–response modelling allows seamless analyses of data that may include inconsistent dilution factors between doses or numbers of subjects per dose group, or small numbers of subjects. Although this investigation is consistent with state-of-the-art ID50 determinations and offers an advancement in clinical data analysis, it is important to underscore that such analyses remain confounded by pathogen aggregation. Regardless, challenging virus strain ID50 determination is crucial for identifying the true infectiousness of HuNoVs and for the accurate evaluation of protective efficacies in pre-clinical studies of therapeutics, vaccines and other prophylactics using this reliable animal model.
Collapse
|
35
|
Cantelli CP, Fumian TM, Malta FC, da Cunha DC, Brasil P, Nordgren J, Svensson L, Miagostovich MP, de Moraes MTB, Leite JPG. Norovirus infection and HBGA host genetic susceptibility in a birth community-cohort, Rio de Janeiro, Brazil. INFECTION, GENETICS AND EVOLUTION : JOURNAL OF MOLECULAR EPIDEMIOLOGY AND EVOLUTIONARY GENETICS IN INFECTIOUS DISEASES 2020; 82:104280. [PMID: 32165242 DOI: 10.1016/j.meegid.2020.104280] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Revised: 03/04/2020] [Accepted: 03/07/2020] [Indexed: 12/29/2022]
Abstract
Norovirus has emerged as an important viral agent of acute pediatric gastroenteritis, with a growing genetic diversity reported in the last decades. Histo-blood group antigens (HBGAs) present on the surface of enterocytes are susceptibility factors for norovirus infection and differ between populations which could affects the epidemiology and evolution of these viruses. This study investigated the frequency, incidence and genetic diversity of noroviruses in a cohort of rotavirus A vaccinated children in association to the host HBGA (Secretor/Lewis) genetic susceptibility profile. Norovirus genogroups I and II (GI/GII) were screened by RT-qPCR in 569 stool samples from 132 children followed-up from birth to 11 months of age during 2014--2018. Noroviruses were identified in 21.2% of children enrolled in this study, with a norovirus detection rate of 5.6% (32/569), in 17.1% and 4.7% of acute diarrheic episodes (ADE) and non-ADE, respectively. The norovirus incidence was 5.8 infections per 100 child-months. Partial nucleotide sequencing characterized six different norovirus genotypes, with GII.4 Sydney 2012 being detected in 50% associated with three different polymerase genotypes (GII·P31, GII·P16 and GII·P4 New Orleans 2009). FUT3 genotyping was yielded seven new mutations in this population. A significant association between symptomatic norovirus infection and secretor profile could be inferred.
Collapse
Affiliation(s)
- Carina Pacheco Cantelli
- Immunobiological Technology Institute/Bio-Manguinhos, Fiocruz, Avenida Brasil, 4365, Manguinhos, Rio de Janeiro, Brazil; Laboratory of Comparative and Environmental Virology, Oswaldo Cruz Institute, Fiocruz, Avenida Brasil, 4365, Manguinhos, Rio de Janeiro, Brazil.
| | - Tulio Machado Fumian
- Laboratory of Comparative and Environmental Virology, Oswaldo Cruz Institute, Fiocruz, Avenida Brasil, 4365, Manguinhos, Rio de Janeiro, Brazil
| | - Fábio Correia Malta
- Laboratory of Comparative and Environmental Virology, Oswaldo Cruz Institute, Fiocruz, Avenida Brasil, 4365, Manguinhos, Rio de Janeiro, Brazil
| | - Denise Cotrim da Cunha
- Sérgio Arouca Public Health National School, Fiocruz, Avenida Brasil, 4365, Manguinhos, Rio de Janeiro, Brazil
| | - Patricia Brasil
- Evandro Chagas National Institute of Infectious Diseases, Fiocruz, Avenida Brasil, 4365, Manguinhos, Rio de Janeiro, Brazil
| | - Johan Nordgren
- Division of Molecular Virology, Department of Clinical and Experimental Medicine, Linköping University, 581 85 Linköping, Sweden
| | - Lennart Svensson
- Division of Molecular Virology, Department of Clinical and Experimental Medicine, Linköping University, 581 85 Linköping, Sweden
| | - Marize Pereira Miagostovich
- Laboratory of Comparative and Environmental Virology, Oswaldo Cruz Institute, Fiocruz, Avenida Brasil, 4365, Manguinhos, Rio de Janeiro, Brazil
| | - Marcia Terezinha Baroni de Moraes
- Laboratory of Comparative and Environmental Virology, Oswaldo Cruz Institute, Fiocruz, Avenida Brasil, 4365, Manguinhos, Rio de Janeiro, Brazil
| | - José Paulo Gagliardi Leite
- Laboratory of Comparative and Environmental Virology, Oswaldo Cruz Institute, Fiocruz, Avenida Brasil, 4365, Manguinhos, Rio de Janeiro, Brazil
| |
Collapse
|
36
|
Abstract
PURPOSE OF REVIEW The majority of norovirus outbreaks in the United States occur in healthcare facilities. With the growing population of immunocompromised hosts who are in frequent contact with healthcare facilities, norovirus is not only a threat to hospitals and nursing homes but also to these individuals. This review summarizes the impact of norovirus infection on healthcare facilities and immunocompromised hosts. RECENT FINDINGS The natural history of norovirus infection in immunocompromised individuals remains poorly understood. Although host immune responses play a critical role in reducing duration of viral shedding and viral load in norovirus-infected individuals, why some immunocompromised patients spontaneously recover while others develop a chronic and protracted course of illness remains unclear. Norovirus outbreaks occur in healthcare facilities because the virus is highly contagious, resistant to disinfection and efficiently transmitted. The use of real-time metagenomic next-generation sequencing and phylogenetic analyses has provided valuable information on transmission patterns in complex hospital-associated norovirus outbreaks. The development of human intestinal enteroid cultures enables the determination of effectiveness of disinfectants against human noroviruses, circumventing the validity questions with surrogate virus models due to differences in susceptibility to inactivation and disinfectants. SUMMARY Metagenomics next-generation sequencing can enhance our understanding of norovirus transmission and lead to more timely mitigation strategies to curb norovirus outbreaks in healthcare facilities. With new in-vitro cultivation methods for human noroviruses, candidate vaccines and effective antivirals could be available in the near future.
Collapse
|
37
|
Noroviruses are highly infectious but there is strong variation in host susceptibility and virus pathogenicity. Epidemics 2020; 32:100401. [PMID: 32721875 DOI: 10.1016/j.epidem.2020.100401] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 06/18/2020] [Accepted: 07/14/2020] [Indexed: 02/06/2023] Open
Abstract
Noroviruses are a major public health concern: their high infectivity and environmental persistence have been documented in several studies. Genetic sequencing shows that noroviruses are highly variable, and exhibit rapid evolution. A few human challenge studies have been performed with norovirus, leading to estimates of their infectivity. However, such incidental estimates do not provide insight into the biological variation of the virus and the interaction with its human host. To study the variation in infectivity and pathogenicity of norovirus, multiple challenge studies must be analysed jointly, to compare their differences and describe how virus infectivity and host susceptibility vary. Since challenge studies can only provide a small sample of the diversity in the natural norovirus population, outbreaks should be exploited as an additional source of information. The present study shows how challenge studies and 'natural experiments' can be combined in a multilevel dose response framework. Infectivity and pathogenicity are analysed by secretor status as a host factor, and genogroup as a pathogen factor. Infectivity, characterized as the estimated mean infection risk when exposed to 1 genomic copy (qPCR unit)is 0.28 for GI norovirus, and 0.076 for GII virus, both in Se+ subjects. The corresponding risks of acute enteric illness are somewhat lower, about 0.2 (GI) and 0.035 (GII), in outbreaks. Se- subjects are protected, with substantially lower risks of infection (0.00007 and 0.015 at a dose of 1 GC of GI and GII virus, respectively). The present study shows there is considerable variability in risk of infection and especially risk of acute symptoms following infection with norovirus. These challenge and outbreak data consistently indicate high infectivity among secretor positives and protection in secretor negatives.
Collapse
|
38
|
Kozak S, Petterson S, McAlister T, Jennison I, Bagraith S, Roiko A. Utility of QMRA to compare health risks associated with alternative urban sewer overflow management strategies. JOURNAL OF ENVIRONMENTAL MANAGEMENT 2020; 262:110309. [PMID: 32250792 DOI: 10.1016/j.jenvman.2020.110309] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 01/10/2020] [Accepted: 02/18/2020] [Indexed: 06/11/2023]
Abstract
Wet weather sewer overflows pose potential short-term public health risks. With increasing populations, aging infrastructure and climate change, utilities are challenged with managing sewerage infrastructure to provide optimum outcomes. This study compared how modelled public health risk profiles could change under alternative sewer overflow management strategies during 12 and 24-month rainfall-runoff events. Specifically, existing conditions were compared with both a 'business-as-usual' (BAU) sewer upgrade and a more holistic 'effects-based planning' (EBP) approach based on pumped wet weather sewage overflows directed to a local receiving waterway. Options were compared based on their efficacy to reduce manhole overflows, recreational waterway guideline exceedances and downstream recreational waterway health risks estimated through a screening-level Quantitative Microbial Risk Assessment (QMRA). Results indicated that the two management strategies would be equally effective in reducing the frequency, duration and volume of manhole sewer overflows, eliminating them in the 12-month scenarios and reducing them from >5000 m3 for the 24-month baseline scenario, to 23 and 35 m3 for BAU and EBP, respectively. Baseline, BAU and EBP scenarios produced similar hours of enterococci guideline exceedances, ranging from 1 to 4 h difference. The QMRA produced similar health risk profiles for downstream recreational waterway users for all design events, suggesting that sewer overflows are not the primary driver of public health risks during and immediately following high rainfall events. As such, QMRA provided evidence that an EBP strategy may be used to manage wet weather sewer overflows in lieu of an expensive BAU upgrade, without exacerbating the public health of downstream waterway users. Further investigation of the broader environmental health impacts of implementing this type of innovative approach is warranted. Nonetheless, this work highlights the value of integrating QMRA with other modelling approaches to guide and inform sewer overflow management.
Collapse
Affiliation(s)
- Sonya Kozak
- School of Medicine, Griffith University, Parklands Drive, Gold Coast, Australia; Cities Research Institute, Griffith University, Parklands Drive, Gold Coast, Australia.
| | - Susan Petterson
- School of Medicine, Griffith University, Parklands Drive, Gold Coast, Australia; Water & Health Pty Ltd, P.O. 648, Salamander Bay, 2317, Australia.
| | - Tony McAlister
- School of Medicine, Griffith University, Parklands Drive, Gold Coast, Australia; Water Technology, Level 3, 43 Peel Street, South Brisbane, QLD, Australia.
| | - Ian Jennison
- Queensland Urban Utilities, 2/15 Green Square Close, Brisbane, Australia.
| | - Sam Bagraith
- Queensland Urban Utilities, 2/15 Green Square Close, Brisbane, Australia.
| | - Anne Roiko
- School of Medicine, Griffith University, Parklands Drive, Gold Coast, Australia; Cities Research Institute, Griffith University, Parklands Drive, Gold Coast, Australia.
| |
Collapse
|
39
|
Lindesmith LC, Brewer-Jensen PD, Mallory ML, Jensen K, Yount BL, Costantini V, Collins MH, Edwards CE, Sheahan TP, Vinjé J, Baric RS. Virus-Host Interactions Between Nonsecretors and Human Norovirus. Cell Mol Gastroenterol Hepatol 2020; 10:245-267. [PMID: 32289501 PMCID: PMC7301201 DOI: 10.1016/j.jcmgh.2020.03.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 03/26/2020] [Accepted: 03/27/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Human norovirus infection is the leading cause of acute gastroenteritis. Genetic polymorphisms, mediated by the FUT2 gene (secretor enzyme), define strain susceptibility. Secretors express a diverse set of fucosylated histoblood group antigen carbohydrates (HBGA) on mucosal cells; nonsecretors (FUT2-/-) express a limited array of HBGAs. Thus, nonsecretors have less diverse norovirus strain infections, including resistance to the epidemiologically dominant GII.4 strains. Because future human norovirus vaccines will comprise GII.4 antigen and because secretor phenotype impacts GII.4 infection and immunity, nonsecretors may mimic young children immunologically in response to GII.4 vaccination, providing a needed model to study cross-protection in the context of limited pre-exposure. METHODS By using specimens collected from the first characterized nonsecretor cohort naturally infected with GII.2 human norovirus, we evaluated the breadth of serologic immunity by surrogate neutralization assays, and cellular activation and cytokine production by flow cytometry. RESULTS GII.2 infection resulted in broad antibody and cellular immunity activation that persisted for at least 30 days for T cells, monocytes, and dendritic cells, and for 180 days for blocking antibody. Multiple cellular lineages expressing interferon-γ and tumor necrosis factor-α dominated the response. Both T-cell and B-cell responses were cross-reactive with other GII strains, but not GI strains. To promote entry mechanisms, inclusion of bile acids was essential for GII.2 binding to nonsecretor HBGAs. CONCLUSIONS These data support development of within-genogroup, cross-reactive antibody and T-cell immunity, key outcomes that may provide the foundation for eliciting broad immune responses after GII.4 vaccination in individuals with limited GII.4 immunity, including young children.
Collapse
Affiliation(s)
- Lisa C Lindesmith
- Department of Epidemiology, University of North Carolina, Chapel Hill, North Carolina
| | - Paul D Brewer-Jensen
- Department of Epidemiology, University of North Carolina, Chapel Hill, North Carolina
| | - Michael L Mallory
- Department of Epidemiology, University of North Carolina, Chapel Hill, North Carolina
| | - Kara Jensen
- Department of Epidemiology, University of North Carolina, Chapel Hill, North Carolina
| | - Boyd L Yount
- Department of Epidemiology, University of North Carolina, Chapel Hill, North Carolina
| | - Veronica Costantini
- Division of Viral Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Matthew H Collins
- Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, School of Medicine, Emory University, Decatur, Georgia
| | - Caitlin E Edwards
- Department of Epidemiology, University of North Carolina, Chapel Hill, North Carolina
| | - Timothy P Sheahan
- Department of Epidemiology, University of North Carolina, Chapel Hill, North Carolina
| | - Jan Vinjé
- Division of Viral Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Ralph S Baric
- Department of Epidemiology, University of North Carolina, Chapel Hill, North Carolina.
| |
Collapse
|
40
|
Mateo R, Lindesmith LC, Garg SJ, Gottlieb K, Lin K, Said S, Leon JS, Sims AC, Weber DJ, Baric RS, Tucker SN, Taylor DN. Production and Clinical Evaluation of Norwalk GI.1 Virus Lot 001-09NV in Norovirus Vaccine Development. J Infect Dis 2020; 221:919-926. [PMID: 31628848 PMCID: PMC7050988 DOI: 10.1093/infdis/jiz540] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 10/12/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Human noroviruses (HuNoV) are the leading cause of gastroenteritis. No vaccine is currently available to prevent norovirus illness or infection. Safe, infectious challenge strains are needed to assess vaccine efficacy in the controlled human infection model (CHIM). METHODS A stock of HuNoV strain Norwalk virus ([NV] GI.1) was prepared. Healthy, genetically susceptible adults were inoculated with NV Lot 001-09NV and monitored for infection, gastroenteritis symptoms, and immune responses. RESULTS Lot 001-09NV induced gastroenteritis in 9 (56%) and infection in 11 (69%) of 16 genetically susceptible subjects. All infected subjects developed strong immune responses to GI.1 with a 30-fold (geometric mean titer) increase in blocking titers (BT50) and a 161-fold increase in GI.1-specific immunoglobulin (Ig)G titers when compared with baseline. GI.1-specific cellular responses in peripheral blood were observed 9 days postchallenge with an average of 3253 IgA and 1227 IgG antibody-secreting cells per million peripheral blood mononuclear cells. CONCLUSIONS GI.1 Lot 001-09NV appears to be similar in virulence to previous passages of NV strain 8fIIa. The safety profile, attack rate, and duration of illness make GI.1 Lot 001-09NV a useful challenge strain for future vaccine studies aimed at establishing immune correlates.
Collapse
Affiliation(s)
| | - Lisa C Lindesmith
- Department of Epidemiology, University of North Carolina, Chapel Hill, North Carolina, USA
| | | | | | - Karen Lin
- Vaxart, Inc., South San Francisco, California, USA
| | - Sara Said
- Vaxart, Inc., South San Francisco, California, USA
| | - Juan S Leon
- Hubert Department of Global Health, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA
| | - Amy C Sims
- Department of Epidemiology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - David J Weber
- Division of Infectious Diseases, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Ralph S Baric
- Department of Epidemiology, University of North Carolina, Chapel Hill, North Carolina, USA
| | | | | |
Collapse
|
41
|
Liao Y, Xue L, Gao J, Wu A, Kou X. ABO blood group-associated susceptibility to norovirus infection: A systematic review and meta-analysis. INFECTION GENETICS AND EVOLUTION 2020; 81:104245. [PMID: 32092482 DOI: 10.1016/j.meegid.2020.104245] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 02/09/2020] [Accepted: 02/14/2020] [Indexed: 12/21/2022]
Abstract
BACKGROUND Norovirus is responsible for the viral gastroenteritis burden worldwide. Histo-blood type antigens (HBGAs) are the only well-known factor regarding their effect on the pathogenesis of norovirus. Here, we performed the study to further investigate the association of the ABO blood group with norovirus susceptibility. METHODS All relevant studies were retrieved from PubMed, Embase, Web of Science, and Cochrane Library databases and the associations of ABO blood groups with norovirus were assessed. The pooled odds ratios (ORs) and 95% confidence interval (CI) were calculated from extracted data. I2 statistics, sensitivity analysis, and publication bias were used to confirm the findings. Subgroup analyses were performed for genotypes, publication years, development degree of the countries, and age if heterogeneity was recorded. RESULTS Seventeen articles covering 2304 participants were included. The overall analysis of the studies showed similar ORs of norovirus infection among individuals with blood type A (OR = 0.90, 95%CI = 0.71-1.14, P = 0.37) and blood type B (OR = 0.85, 95%CI = 0.66-1.12, P = 0.25) as compared to those controls. An increased odds of norovirus infection was found among individuals with blood type O (OR = 1.28, 95%CI = 1.03-1.59, P = 0.03), while the individuals with blood type AB (OR = 0.91, 95%CI = 0.60-1.39, P = 0.67) showed no correlation with norovirus infection. For blood type B and blood type AB, the results of subgroup analyses mirrored the observations above. CONCLUSIONS The meta-analysis suggested that the blood type A, B and AB might not affect susceptibility to norovirus infection. However, blood type O appeared to be more susceptible to norovirus infection.
Collapse
Affiliation(s)
- Yingyin Liao
- KingMed school of Laboratory Medicine of Guangzhou Medical University, Guangzhou, China
| | - Liang Xue
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Institute of Microbiology, NO.100, Xianlie Zhong Road, Guangzhou 510070, People's Republic of China; Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Guangzhou 510070, People's Republic of China
| | - Junshan Gao
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Institute of Microbiology, NO.100, Xianlie Zhong Road, Guangzhou 510070, People's Republic of China; Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Guangzhou 510070, People's Republic of China
| | - Aiwu Wu
- KingMed school of Laboratory Medicine of Guangzhou Medical University, Guangzhou, China
| | - Xiaoxia Kou
- KingMed school of Laboratory Medicine of Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
42
|
Abstract
PURPOSE OF REVIEW Noroviruses are a major cause of gastroenteritis. This review summarizes new information on noroviruses that may lead to the development of improved measures for limiting their human health impact. RECENT FINDINGS GII.4 strains remain the most common human noroviruses causing disease, although GII.2 and GII.17 strains have recently emerged as dominant strains in some populations. Histo-blood group antigen (HBGA) expression on the gut mucosa drives susceptibility to different norovirus strains. Antibodies that block virus binding to these glycans correlate with protection from infection and illness. Immunocompromised patients are significantly impacted by norovirus infection, and the increasing availability of molecular diagnostics has improved infection recognition. Human noroviruses can be propagated in human intestinal enteroid cultures containing enterocytes that are a significant primary target for initiating infection. Strain-specific requirements for replication exist with bile being essential for some strains. Several vaccine candidates are progressing through preclinical and clinical development and studies of potential antiviral interventions are underway. SUMMARY Norovirus epidemiology is complex and requires continued surveillance to track the emergence of new strains and recombinants, especially with the continued progress in vaccine development. Humans are the best model to study disease pathogenesis and prevention. New in-vitro cultivation methods should lead to better approaches for understanding virus-host interactions and ultimately to improved strategies for mitigation of human norovirus-associated disease.
Collapse
|
43
|
GII.13/21 Noroviruses Recognize Glycans with a Terminal β-Galactose via an Unconventional Glycan Binding Site. J Virol 2019; 93:JVI.00723-19. [PMID: 31118252 PMCID: PMC6639292 DOI: 10.1128/jvi.00723-19] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 05/01/2019] [Indexed: 01/17/2023] Open
Abstract
Evidence from both phenotypic binding assay and structural study support the observed interactions of human noroviruses (huNoVs) with histo-blood group antigens (HBGAs) as receptors or attachment factors, affecting their host susceptibility. GII.13 and GII.21 genotypes form a unique genetic lineage that differs from the mainstream GII huNoVs in their unconventional glycan binding site. Unlike the previous findings that GII.13/21 genotypes recognize only Lea antigen, we found in this study that they can interact with a group of glycans with a common terminal β-Gal, including Lec, lactose, and mucin core 2. However, this wide glycan binding spectrum in a unique binding mode of the GII.13/21 huNoVs appears not to increase their prevalence, probably due to the existence of decoy glycan receptors in human gastrointestinal tract limiting their infection. Our findings shed light on the host interaction and epidemiology of huNoVs, which would impact the strategy of huNoV control and prevention. Human noroviruses (huNoVs) recognize histo-blood group antigens (HBGAs) as host susceptibility factors. GII.13 and GII.21 huNoVs form a unique genetic lineage that emerged from mainstream GII NoVs via development of a new, nonconventional glycan binding site (GBS) that binds Lea antigen. This previous finding raised the question of whether the new GII.13/21 GBS really has such a narrow glycan binding spectrum. In this study, we provide solid phenotypic and structural evidence indicating that this new GBS recognizes a group of glycans with a common terminal β-galactose (β-Gal). First, we found that P domain proteins of GII.13/21 huNoVs circulating at different times bound three glycans sharing a common terminal β-Gal, including Lec, lactose, and mucin core 2. Second, we solved the crystal structures of the GII.13 P dimers in complex with Lec and mucin core 2, which showed that β-Gal is the major binding saccharide. Third, nonfat milk and lactose blocked the GII.13/21 P domain-glycan binding, which may explain the low prevalence of GII.13/21 viruses. Our data provide new insight into the host interactions and epidemiology of huNoVs, which would help in the control and prevention of NoV-associated diseases. IMPORTANCE Evidence from both phenotypic binding assay and structural study support the observed interactions of human noroviruses (huNoVs) with histo-blood group antigens (HBGAs) as receptors or attachment factors, affecting their host susceptibility. GII.13 and GII.21 genotypes form a unique genetic lineage that differs from the mainstream GII huNoVs in their unconventional glycan binding site. Unlike the previous findings that GII.13/21 genotypes recognize only Lea antigen, we found in this study that they can interact with a group of glycans with a common terminal β-Gal, including Lec, lactose, and mucin core 2. However, this wide glycan binding spectrum in a unique binding mode of the GII.13/21 huNoVs appears not to increase their prevalence, probably due to the existence of decoy glycan receptors in human gastrointestinal tract limiting their infection. Our findings shed light on the host interaction and epidemiology of huNoVs, which would impact the strategy of huNoV control and prevention.
Collapse
|
44
|
Emelko MB, Schmidt PJ, Borchardt MA. Confirming the need for virus disinfection in municipal subsurface drinking water supplies. WATER RESEARCH 2019; 157:356-364. [PMID: 30970285 DOI: 10.1016/j.watres.2019.03.057] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2018] [Revised: 03/08/2019] [Accepted: 03/26/2019] [Indexed: 06/09/2023]
Abstract
Enteric viruses pose the greatest acute human health risks associated with subsurface drinking water supplies, yet quantitative risk assessment tools have rarely been used to develop health-based targets for virus treatment in drinking water sourced from these supplies. Such efforts have previously been hampered by a lack of consensus concerning a suitable viral reference pathogen and dose-response model as well as difficulties in quantifying pathogenic viruses in water. A reverse quantitative microbial risk assessment (QMRA) framework and quantitative polymerase chain reaction data for norovirus genogroup I in subsurface drinking water supplies were used herein to evaluate treatment needs for such water supplies. Norovirus was not detected in over 90% of samples, which emphasizes the need to consider the spatially and/or temporally intermittent patterns of enteric pathogen contamination in subsurface water supplies. Collectively, this analysis reinforces existing recommendations that a minimum 4-log treatment goal is needed for enteric viruses in groundwater in absence of well-specific monitoring information. This result is sensitive to the virus dose-response model used as there is approximately a 3-log discrepancy among virus dose-response models in the existing literature. This emphasizes the need to address the uncertainties and lack of consensus related to various QMRA modelling approaches and the analytical limitations that preclude more accurate description of virus risks.
Collapse
Affiliation(s)
- M B Emelko
- Department of Civil and Environmental Engineering, University of Waterloo, 200 University Ave. W. Waterloo, Ontario, N2L 3G1, Canada.
| | - P J Schmidt
- Department of Civil and Environmental Engineering, University of Waterloo, 200 University Ave. W. Waterloo, Ontario, N2L 3G1, Canada
| | - M A Borchardt
- Agricultural Research Service, U.S. Department of Agriculture, Marshfield, WI, 54449, United States
| |
Collapse
|
45
|
The Antigenic Topology of Norovirus as Defined by B and T Cell Epitope Mapping: Implications for Universal Vaccines and Therapeutics. Viruses 2019; 11:v11050432. [PMID: 31083353 PMCID: PMC6563215 DOI: 10.3390/v11050432] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 05/07/2019] [Accepted: 05/08/2019] [Indexed: 12/11/2022] Open
Abstract
Human norovirus (HuNoV) is the leading cause of acute nonbacterial gastroenteritis. Vaccine design has been confounded by the antigenic diversity of these viruses and a limited understanding of protective immunity. We reviewed 77 articles published since 1988 describing the isolation, function, and mapping of 307 unique monoclonal antibodies directed against B cell epitopes of human and murine noroviruses representing diverse Genogroups (G). Of these antibodies, 91, 153, 21, and 42 were reported as GI-specific, GII-specific, MNV GV-specific, and G cross-reactive, respectively. Our goal was to reconstruct the antigenic topology of noroviruses in relationship to mapped epitopes with potential for therapeutic use or inclusion in universal vaccines. Furthermore, we reviewed seven published studies of norovirus T cell epitopes that identified 18 unique peptide sequences with CD4- or CD8-stimulating activity. Both the protruding (P) and shell (S) domains of the major capsid protein VP1 contained B and T cell epitopes, with the majority of neutralizing and HBGA-blocking B cell epitopes mapping in or proximal to the surface-exposed P2 region of the P domain. The majority of broadly reactive B and T cell epitopes mapped to the S and P1 arm of the P domain. Taken together, this atlas of mapped B and T cell epitopes offers insight into the promises and challenges of designing universal vaccines and immunotherapy for the noroviruses.
Collapse
|
46
|
Owusu-Ansah EDGJ, Barnes B, Abaidoo R, Tine H, Dalsgaard A, Permin A, Schou TW. Probabilistic modeling for an integrated temporary acquired immunity with norovirus epidemiological data. Infect Dis Model 2019; 4:99-114. [PMID: 31080934 PMCID: PMC6503004 DOI: 10.1016/j.idm.2019.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 04/22/2019] [Indexed: 11/28/2022] Open
Abstract
Integration of acquired immunity into microbial risk assessment for illness incidence is of no doubt essential for the study of susceptibility to illness. In this study, a probabilistic model was set up as dose response for infection and a mathematical derivation was carried out by integrating immunity to obtain probability of illness models. Temporary acquire immunity from epidemiology studies which includes six different Norovirus transmission scenarios such as symptomatic individuals infectious, pre- and post-symptomatic infectiousness (low and high), innate genetic resistance, genogroup 2 type 4 and those with no immune boosting by asymptomatic infection were evaluated. Simulated results on illness inflation factor as a function of dose and exposure indicated that high frequency exposures had immense immunity build up even at high dose levels; hence minimized the probability of illness. Using Norovirus transmission dynamics data, results showed, and immunity included models had a reduction of 2-6 logs of magnitude difference in disease burden for both population and individual probable illness incidence. Additionally, the magnitude order of illness for each dose response remained largely the same for all transmission scenarios; symptomatic infectiousness and no immune boosting after asymptomatic infectiousness also remained the same throughout. With integration of epidemiological data on acquired immunity into the risk assessment, more realistic results were achieved signifying an overestimation of probable risk of illness when epidemiological immunity data are not included. This finding supported the call for rigorous integration of temporary acquired immunity in dose-response in all microbial risk assessments.
Collapse
Affiliation(s)
- Emmanuel de-Graft Johnson Owusu-Ansah
- Department of Mathematics, Faculty of Physical and Computational Science, College of Science, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana.,Laboratory of Integrated Statistical Applications (LISA), Faculty of Physical and Computational Sciences, College of Science, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana.,Regional Water and Environmental Sanitation Center-Kumasi, College of Engineering, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Benedict Barnes
- Department of Mathematics, Faculty of Physical and Computational Science, College of Science, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Robert Abaidoo
- Department of Theoretical and Applied Biology, Faculty of Bio Science, College of Science, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Hald Tine
- Research Group of Genomic Epidemiology, National Food Institute, Technical University of Denmark, Kgs Lyngby, Denmark
| | - Anders Dalsgaard
- Food Safety and Zoonoses, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Stigböjlen 4, 1870 Frederiksberg C, Denmark
| | - Anders Permin
- Office for Innovation and Sector Services, Administration, Technical University of Denmark, Anker Engelunds Vej, 2800 Kgs Lyngby, Denmark
| | | |
Collapse
|
47
|
Nordgren J, Svensson L. Genetic Susceptibility to Human Norovirus Infection: An Update. Viruses 2019; 11:E226. [PMID: 30845670 PMCID: PMC6466115 DOI: 10.3390/v11030226] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 02/28/2019] [Accepted: 03/02/2019] [Indexed: 12/15/2022] Open
Abstract
Noroviruses are the most common etiological agent of acute gastroenteritis worldwide. Despite their high infectivity, a subpopulation of individuals is resistant to infection and disease. This susceptibility is norovirus genotype-dependent and is largely mediated by the presence or absence of human histo-blood group antigens (HBGAs) on gut epithelial surfaces. The synthesis of these HBGAs is mediated by fucosyl- and glycosyltransferases under the genetic control of the FUT2 (secretor), FUT3 (Lewis) and ABO(H) genes. The so-called non-secretors, having an inactivated FUT2 enzyme, do not express blood group antigens and are resistant to several norovirus genotypes, including the predominant GII.4. Significant genotypic and phenotypic diversity of HBGA expression exists between different human populations. Here, we review previous in vivo studies on genetic susceptibility to norovirus infection. These are discussed in relation to population susceptibility, vaccines, norovirus epidemiology and the impact on public health.
Collapse
Affiliation(s)
- Johan Nordgren
- Division of Molecular Virology, Department of Clinical and Experimental Medicine, Linköping University, 58185 Linköping, Sweden.
| | - Lennart Svensson
- Division of Molecular Virology, Department of Clinical and Experimental Medicine, Linköping University, 58185 Linköping, Sweden.
- Division of Infectious Diseases, Department of Medicine, Karolinska Institute, 17177 Stockholm, Sweden.
| |
Collapse
|
48
|
Yang Y, Xia M, Wang L, Arumugam S, Wang Y, Ou X, Wang C, Jiang X, Tan M, Chen Y, Li X. Structural basis of host ligand specificity change of GII porcine noroviruses from their closely related GII human noroviruses. Emerg Microbes Infect 2019; 8:1642-1657. [PMID: 31711377 PMCID: PMC6853222 DOI: 10.1080/22221751.2019.1686335] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 10/20/2019] [Indexed: 02/07/2023]
Abstract
Diverse noroviruses infect humans and animals via the recognition of host-specific glycan ligands. Genogroup II (GII) noroviruses consist of human noroviruses (huNoVs) that generally bind histo-blood group antigens (HBGAs) as host factors and three porcine norovirus (porNoV) genotypes (GII.11/18/19) that form a genetic lineage lacking HBGA-binding ability. Thus, these GII porNoVs provide an excellent model to study norovirus evolution with host ligand specificity changes. Here we solved the crystal structures of a native GII.11 porNoV P protein and a closely-related GII.3 huNoV P protein complexed with an HBGA, focusing on the HBGA-binding sites (HBSs) compared with the previously known ones to understand the structural basis of the host ligand specificity change. We found that the GII.3 huNoV binds HBGAs via a conventional GII HBS that uses an arginine instead of the conserved aromatic residue for the required Van der Waals interaction, while the GII.11 porNoV HBS loses its HBGA-binding function because of two mutations (Q355/V451). A mutant that reversed the two mutated residues back to the conventional A355/Y451 restored the HBGA-binding function of the GII.11 porNoV P protein, which validated our observations. Similar mutations are also found in GII.19 porNoVs and a GII.19 P protein mutant with double reverse mutations restored the HBS function. This is the first reconstruction of a functional HBS based on one with new host specificity back to its parental one. These data shed light on the molecular basis of structural adaptation of the GII porNoVs to the pig hosts through mutations at their HBSs.
Collapse
Affiliation(s)
- Yang Yang
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, People’s Republic of China
- University of Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Ming Xia
- Division of Infectious Diseases, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Leyi Wang
- Division of Infectious Diseases, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Sahaana Arumugam
- Division of Infectious Diseases, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Yajing Wang
- College of Life Science, Nankai University, Tianjin, People’s Republic of China
| | - Xianjin Ou
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Chenlong Wang
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Xi Jiang
- Division of Infectious Diseases, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Ming Tan
- Division of Infectious Diseases, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Yutao Chen
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Xuemei Li
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, People’s Republic of China
| |
Collapse
|
49
|
Qian Y, Song M, Jiang X, Xia M, Meller J, Tan M, Chen Y, Li X, Rao Z. Structural Adaptations of Norovirus GII.17/13/21 Lineage through Two Distinct Evolutionary Paths. J Virol 2019; 93:e01655-18. [PMID: 30333166 PMCID: PMC6288326 DOI: 10.1128/jvi.01655-18] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 09/22/2018] [Indexed: 11/20/2022] Open
Abstract
Human noroviruses (huNoVs), which cause epidemic acute gastroenteritis, recognize histo-blood group antigens (HBGAs) as host attachment factors affecting host susceptibility. HuNoVs are genetically diverse, containing at least 31 genotypes in the two major genogroups (genogroup I [GI] and GII). Three GII genotypes, GII genotype 17 (GII.17), GII.13, and GII.21, form a unique genetic lineage, in which the GII.17 genotype retains the conventional GII HBGA binding site (HBS), while the GII.13/21 genotypes acquire a completely new HBS. To understand the molecular bases behind these evolutionary changes, we solved the crystal structures of the HBGA binding protruding domains of (i) an early GII.17 variant (the 1978 variant) that does not bind or binds weakly to HBGAs, (ii) the new GII.17 variant (the 2014/15 variant) that binds A/B/H antigens strongly via an optimized GII HBS, and (iii) a GII.13 variant (the 2010 variant) that binds the Lewis a (Lea) antigen via the new HBS. These serial, high-resolution structural data enable a comprehensive structural comparison to understand the evolutionary changes of the GII.17/13/21 lineage, including the emergence of the new HBS of the GII.13/21 sublineage and the possible HBS optimization of the recent GII.17 variant for an enhanced HBGA binding ability. Our study elucidates the structural adaptations of the GII.17/13/21 lineage through distinct evolutionary paths, which may allow a theory explaining huNoV adaptations and evolutions to be put forward.IMPORTANCE Our understanding of the molecular bases behind the interplays between human noroviruses and their host glycan ligands, as well as their evolutionary changes over time with alterations in their host ligand binding capability and host susceptibility, remains limited. By solving the crystal structures of the glycan ligand binding protruding (P) domains with or without glycan ligands of three representative noroviruses of the GII.17/13/21 genetic lineage, we elucidated the molecular bases of the human norovirus-glycan interactions of this special genetic lineage. We present solid evidence on how noroviruses of this genetic lineage evolved via different evolutionary paths to (i) optimize their glycan binding site for higher glycan binding function and (ii) acquire a completely new glycan binding site for new ligands. Our data shed light on the mechanism of the structural adaptations of human noroviruses through different evolutionary paths, facilitating our understanding of human norovirus adaptations, evolutions, and epidemiology.
Collapse
Affiliation(s)
- Ying Qian
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Mohan Song
- College of Life Science, Nankai University, Tianjin, China
| | - Xi Jiang
- Division of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Ming Xia
- Division of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Jarek Meller
- Division of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Environmental Health, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Ming Tan
- Division of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Yutao Chen
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Xuemei Li
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Zihe Rao
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
50
|
Bányai K, Estes MK, Martella V, Parashar UD. Viral gastroenteritis. Lancet 2018; 392:175-186. [PMID: 30025810 PMCID: PMC8883799 DOI: 10.1016/s0140-6736(18)31128-0] [Citation(s) in RCA: 260] [Impact Index Per Article: 43.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 05/09/2018] [Accepted: 05/15/2018] [Indexed: 12/20/2022]
Abstract
Enteric viruses, particularly rotaviruses and noroviruses, are a leading cause of gastroenteritis worldwide. Rotaviruses primarily affect young children, accounting for almost 40% of hospital admissions for diarrhoea and 200 000 deaths worldwide, with the majority of deaths occurring in developing countries. Two vaccines against rotavirus were licensed in 2006 and have been implemented in 95 countries as of April, 2018. Data from eight high-income and middle-income countries showed a 49-89% decline in rotavirus-associated hospital admissions and a 17-55% decline in all-cause gastroenteritis-associated hospital admissions among children younger than 5 years, within 2 years of vaccine introduction. Noroviruses affect people of all ages, and are a leading cause of foodborne disease and outbreaks of gastroenteritis worldwide. Prevention of norovirus infection relies on frequent hand hygiene, limiting contact with people who are infected with the virus, and disinfection of contaminated environmental surfaces. Norovirus vaccine candidates are in clinical trials; whether vaccines will provide durable protection against the range of genetically and antigenically diverse norovirus strains remains unknown. Treatment of viral gastroenteritis is based primarily on replacement of fluid and electrolytes.
Collapse
Affiliation(s)
- Krisztián Bányai
- Institute for Veterinary Medical Research, Centre for Agricultural Research, Hungarian Academy of Sciences, Budapest, Hungary.
| | - Mary K Estes
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Vito Martella
- Department of Veterinary Medicine, University Aldo Moro of Bari, Provincial Road to Casamassima, Valenzano, Italy
| | - Umesh D Parashar
- Viral Gastroenteritis Branch, Division of Viral Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| |
Collapse
|