1
|
Prasad BVV, Atmar RL, Ramani S, Palzkill T, Song Y, Crawford SE, Estes MK. Norovirus replication, host interactions and vaccine advances. Nat Rev Microbiol 2025; 23:385-401. [PMID: 39824927 DOI: 10.1038/s41579-024-01144-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/12/2024] [Indexed: 01/20/2025]
Abstract
Human noroviruses (HuNoVs) are the leading cause of acute gastroenteritis worldwide in all age groups and cause significant disease and economic burden globally. To date, no approved vaccines or antiviral therapies are available to treat or prevent HuNoV illness. Several candidate vaccines are in clinical trials, although potential barriers to successful development must be overcome. Recently, significant advances have been made in understanding HuNoV biology owing to breakthroughs in virus cultivation using human intestinal tissue-derived organoid (or enteroid) cultures, advances in structural biology technology combined with epitope mapping and increased metagenomic sequencing. New and unexpected strain-specific differences in pandemic versus non-pandemic virus structures, replication properties and virus-host interactions, including host factors required for susceptibility to infection and pathogenesis, are discussed.
Collapse
Affiliation(s)
- B V Venkataram Prasad
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Robert L Atmar
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Sasirekha Ramani
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Timothy Palzkill
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Yongcheng Song
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, TX, USA
| | - Sue E Crawford
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Mary K Estes
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA.
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
2
|
Farkas T. B cell lines fail to support efficient rhesus enteric calicivirus and human norovirus replication. J Virol 2025; 99:e0014325. [PMID: 40261012 PMCID: PMC12090725 DOI: 10.1128/jvi.00143-25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Accepted: 03/19/2025] [Indexed: 04/24/2025] Open
Abstract
Analyses of intestinal biopsies of infected individuals and/or nonhuman primates (NHP) suggested the possible immune cell tropism of human noroviruses (HuNoV) and rhesus enteric caliciviruses (ReCV). Subsequently, the first HuNoV cell culture system using human B cell lines was reported. However, reproducibility issues raised questions about the validity and suitability of B cell cultures for HuNoV research. Histo-blood group antigens (HBGA) are known HuNoV susceptibility factors, but the full range of HuNoV susceptibility determinants remains unknown. In contrast, strain-specific ReCV susceptibility determinants have been recently characterized. Here, we evaluated NHP B cell lines and the human BJAB cell line for susceptibility to ReCV-FT285 infection, which is controlled by the Coxsackie and adenovirus receptor (CAR) and the type A or B HBGA. NHP B cell lines lacked CAR and HBGA expression and resisted infection. Inconsistent, low-level virus replication was detectable in BJAB cells, and expression of CAR and HBGAs was evident by Western blots. However, <1% of live, but >80% of fixed and permeabilized BJAB cells were CAR+, suggesting that CAR is mostly internalized. Co-transfection of BJAB cells with hCAR and A enzyme expression vectors led to substantial surface CAR and type A HBGA expression but not to an increase in ReCV titers. dsRNA staining revealed initial ReCV and HuNoV infection in a few cells that most likely became abortive. Based on both the similarities between ReCV and HuNoV replication profiles and the results obtained in the present study, considering BJAB cells an efficient culture system for HuNoV research is not justified.IMPORTANCERecently, two human norovirus (HuNoV) cell culture systems have been developed-the B cell culture system and the enteroid culture system. While the enteroid cell culture system became widely used in HuNoV research, mainly due to reproducibility issues, the B cell culture system did not. Here, we used HuNoV and rhesus enteric caliciviruses (ReCV) to evaluate enteric calicivirus B cell infections, in correlation to cell surface molecular determinants that control the susceptibility to infection. These are fully characterized for ReCVs, but not for HuNoVs. We found that only few BJAB cells express the cell surface molecules necessary for ReCV infection and support low-level, initial ReCV and HuNoV infection, but virus replication is most likely abortive, with minimal progeny virus release. Our findings and the poor reproducibility indicate that the B cell culture system in its current form is unsuitable for ReCV or HuNoV research and does not represent an efficient valid cell culture system.
Collapse
Affiliation(s)
- Tibor Farkas
- Department of Veterinary Pathobiology, College of Veterinary Medicine & Biomedical Sciences, Texas A&M University, College Station, Texas, USA
| |
Collapse
|
3
|
Durkee-Shock J, Cohen A, Maghzian N, Pezzella G, Jensen-Wachspress M, Hostal A, Barton K, Gangler K, Dávila Saldaña BJ, Chaimongkol N, Bollard CM, Sosnovtsev SV, Cohen J, Nagata BM, Alves DA, Ghosh R, Seifert BA, Freeman A, Gonzalez C, Notarangelo LD, Green KY, Keller MD. Reconstitution of Norovirus-Specific T-Cell Responses Following Hematopoietic Stem Cell Transplantation in Patients With Inborn Errors of Immunity and Chronic Norovirus Infection. J Infect Dis 2025; 231:773-783. [PMID: 39140311 PMCID: PMC11911796 DOI: 10.1093/infdis/jiae398] [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: 02/20/2024] [Revised: 05/25/2024] [Accepted: 08/13/2024] [Indexed: 08/15/2024] Open
Abstract
BACKGROUND Chronic norovirus infection (CNI) causes significant morbidity in immunocompromised patients. No effective prevention or treatment currently exists. METHODS Two patients with inborn errors of immunity, X-linked severe combined immunodeficiency (X-SCID) and DOCK8 deficiency, were followed longitudinally for clinical course, immune reconstitution, norovirus-specific T-cell (NST) response, B-cell reconstitution, and norovirus-specific antibody production. Samples were obtained in the peri-hematopoietic stem cell transplant (HSCT) setting before and after CNI clearance. The norovirus strain causing CNI was followed longitudinally for norovirus stool viral loads and sequencing. RESULTS The noroviruses were identified as GII.4 Sydney[P4 New Orleans] in 1 patient and GII.17[P17] in the other. An exacerbation of diarrhea post-HSCT in the patient with X-SCID was consistent with norovirus infection but not with graft-versus-host disease on pathologic samples. Both patients recovered polyfunctional NSTs in the CD4 and CD8 T-cell compartments that recognized multiple norovirus structural and nonstructural viral antigens. T-cell responses were minimal during active CNI but detectable after resolution. Mapping of NST responses between the patient with DOCK8 deficiency and his matched sibling donor were nearly identical. B-cell reconstitution or new endogenous antibody production for immunoglobulin A or immunoglobulin G was not observed. CONCLUSIONS This report is the first to demonstrate reconstitution of NST immunity after HSCT closely temporally aligned with clearance of CNI, suggesting that cellular immunity is sufficient for norovirus clearance.
Collapse
Affiliation(s)
- Jessica Durkee-Shock
- Laboratory of Infectious Diseases, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland
| | - Ariella Cohen
- Center for Cancer and Immunology Research, Children’s National Hospital, Washington, District of Columbia
| | - Naseem Maghzian
- Center for Cancer and Immunology Research, Children’s National Hospital, Washington, District of Columbia
| | - Gloria Pezzella
- Center for Cancer and Immunology Research, Children’s National Hospital, Washington, District of Columbia
| | - Mariah Jensen-Wachspress
- Center for Cancer and Immunology Research, Children’s National Hospital, Washington, District of Columbia
| | - Anna Hostal
- Laboratory of Infectious Diseases, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland
| | - Karenna Barton
- Laboratory of Infectious Diseases, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland
| | - Krista Gangler
- Laboratory of Infectious Diseases, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland
| | - Blachy J Dávila Saldaña
- Center for Cancer and Immunology Research, Children’s National Hospital, Washington, District of Columbia
- Division of Blood and Marrow Transplantation, Children’s National Hospital, Washington, District of Columbia
| | - Natthawan Chaimongkol
- Laboratory of Infectious Diseases, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland
| | - Catherine M Bollard
- Center for Cancer and Immunology Research, Children’s National Hospital, Washington, District of Columbia
- Division of Blood and Marrow Transplantation, Children’s National Hospital, Washington, District of Columbia
| | - Stanislav V Sosnovtsev
- Laboratory of Infectious Diseases, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland
| | - Jeffrey Cohen
- Laboratory of Infectious Diseases, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland
| | - Bianca M Nagata
- Infectious Disease Pathogenesis Section, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, Rockville, Maryland
| | - Derron A Alves
- Infectious Disease Pathogenesis Section, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, Rockville, Maryland
| | - Rajarshi Ghosh
- NIAID Centralized Sequencing Program, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland
| | - Bryce A Seifert
- NIAID Centralized Sequencing Program, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland
| | - Alexandra Freeman
- Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland
| | - Corina Gonzalez
- Immune Deficiency Cellular Therapy Program, National Cancer Institute, Bethesda, Maryland
| | - Luigi D Notarangelo
- Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland
| | - Kim Y Green
- Laboratory of Infectious Diseases, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland
| | - Michael D Keller
- Center for Cancer and Immunology Research, Children’s National Hospital, Washington, District of Columbia
- Division of Allergy and Immunology, Children's National Hospital, Washington, District of Columbia
| |
Collapse
|
4
|
Colmenares MT, Matos ADO, Dantas PHS, Neto JRDC, Neves BJ, Gardinassi LGA, Silva-Sales M, Sales-Campos H. TREM-1 as a Potential Coreceptor in Norovirus Pathogenesis: Insights from Transcriptomic Analysis and Molecular Docking. ACS OMEGA 2025; 10:4881-4895. [PMID: 39959083 PMCID: PMC11822722 DOI: 10.1021/acsomega.4c10220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 01/14/2025] [Accepted: 01/22/2025] [Indexed: 02/18/2025]
Abstract
Norovirus (NoV) is a major cause of acute diarrheal disease in humans. However, due to complications in cultivating this virus, bioinformatics aids in elucidating the virus-host relationship. One of the molecules that has been associated with the burden of viral diseases is TREM-1, mainly due to its role in amplifying the inflammatory response. Thus, we hypothesized that TREM-1 may be involved in NoV infection. Analysis of public transcriptomic data sets showed an increased expression of Trem1 and Trem3 during murine NoV (MNoV) infection. Then, molecular docking was performed between murine TREM-1 and the P domain of the MNoV VP1 protein. The viral antigenic segment C'-D' was recognized by the murine TREM-1 CDR1 region. Subsequently, based on phylogenetic criteria, NoV VP1 proteins from the GII.4 genotype sequenced in different years (1987, 2010, 2012, 2014, 2016, and 2019) were modeled. Using docking and molecular dynamics simulations, a stable interaction was observed between the human TREM-1 Ig-like domain and the conserved S and P segments of the NoV VP1 protein. Notably, this interaction was conserved over the years and was mainly dictated by the TREM-1 CDR3 region. Also, coexpression between Trem1 with genes involved in apoptosis and pyroptosis pathways was surveyed during viral infection by MNoV. It was found that Trem1 is primarily expressed with genes from the pyroptosis pathway. These simulations strongly suggest the involvement of TREM-1 in NoV pathogenesis and its potential contribution as a coreceptor.
Collapse
Affiliation(s)
- Mike Telemaco
Contreras Colmenares
- Laboratório
de Imunologia de Mucosas e Imunoinformática, Instituto de Patologia
Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia 74605-170, Brazil
| | - Amanda de Oliveira Matos
- Laboratório
de Imunologia de Mucosas e Imunoinformática, Instituto de Patologia
Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia 74605-170, Brazil
| | - Pedro Henrique
dos Santos Dantas
- Laboratório
de Imunologia de Mucosas e Imunoinformática, Instituto de Patologia
Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia 74605-170, Brazil
| | - José Rodrigues Do Carmo Neto
- Laboratório
de Imunologia de Mucosas e Imunoinformática, Instituto de Patologia
Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia 74605-170, Brazil
| | - Bruno Júnior Neves
- Laboratório
de Quimioinformática, Faculdade de Farmácia, Universidade Federal de Goiás, Goiânia, GO 74605-170, Brazil
| | | | - Marcelle Silva-Sales
- Laboratório
de Imunologia de Mucosas e Imunoinformática, Instituto de Patologia
Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia 74605-170, Brazil
- Laboratório
de Quimioinformática, Faculdade de Farmácia, Universidade Federal de Goiás, Goiânia, GO 74605-170, Brazil
| | - Helioswilton Sales-Campos
- Laboratório
de Imunologia de Mucosas e Imunoinformática, Instituto de Patologia
Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia 74605-170, Brazil
| |
Collapse
|
5
|
Chio CC, Chien JC, Chan HW, Huang HI. Overview of the Trending Enteric Viruses and Their Pathogenesis in Intestinal Epithelial Cell Infection. Biomedicines 2024; 12:2773. [PMID: 39767680 PMCID: PMC11672972 DOI: 10.3390/biomedicines12122773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 11/08/2024] [Accepted: 12/03/2024] [Indexed: 01/11/2025] Open
Abstract
Enteric virus infection is a major public health issue worldwide. Enteric viruses have become epidemic infectious diseases in several countries. Enteric viruses primarily infect the gastrointestinal tract and complete their life cycle in intestinal epithelial cells. These viruses are transmitted via the fecal-oral route through contaminated food, water, or person to person and cause similar common symptoms, including vomiting, abdominal pain, and diarrhea. Diarrheal disease is the third leading cause of death in children under five years of age, accounting for approximately 1.7 billion cases and 443,832 deaths annually in this age group. Additionally, some enteric viruses can invade other tissues, leading to severe conditions and even death. The pathogenic mechanisms of enteric viruses are also unclear. In this review, we organized the research on trending enteric virus infections, including rotavirus, norovirus, adenovirus, Enterovirus-A71, Coxsackievirus A6, and Echovirus 11. Furthermore, we discuss the gastrointestinal effects and pathogenic mechanisms of SARS-CoV-2 in intestinal epithelial cells, given the gastrointestinal symptoms observed during the COVID-19 pandemic. We conducted a literature review on their pathogenic mechanisms, which serves as a guide for formulating future treatment strategies for enteric virus infections.
Collapse
Affiliation(s)
- Chi-Chong Chio
- Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 33302, Taiwan; (C.-C.C.); (J.-C.C.); (H.-W.C.)
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 33302, Taiwan
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 33302, Taiwan
| | - Jou-Chun Chien
- Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 33302, Taiwan; (C.-C.C.); (J.-C.C.); (H.-W.C.)
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 33302, Taiwan
| | - Hio-Wai Chan
- Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 33302, Taiwan; (C.-C.C.); (J.-C.C.); (H.-W.C.)
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 33302, Taiwan
| | - Hsing-I Huang
- Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 33302, Taiwan; (C.-C.C.); (J.-C.C.); (H.-W.C.)
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 33302, Taiwan
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 33302, Taiwan
- Department of Pediatrics, Linkou Chang Gung Memorial Hospital, Kwei-Shan, Taoyuan 33305, Taiwan
| |
Collapse
|
6
|
Matsushima Y, Levenson EA, Chaimongkol N, Harris L, Zhao Y, Turan S, Otaizo-Carrasquero F, Ganesan S, Hornick KM, Martens C, Sosnovtsev SV, Green KY. Single-cell transcriptional analysis of murine norovirus infection in a human intestinal cell line. J Virol 2024; 98:e0161724. [PMID: 39475272 PMCID: PMC11575399 DOI: 10.1128/jvi.01617-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 09/18/2024] [Indexed: 11/20/2024] Open
Abstract
Noroviruses are a major agent of acute gastroenteritis in humans, but host cell requirements for efficient replication in vitro have not been established. We engineered a human intestinal cell line (designated mCD300lf-hCaco2) expressing the murine norovirus (MNV) receptor, mouse CD300lf to become fully permissive for MNV replication. To explore the replicative machinery and host response of these cells, we performed a single-cell RNA sequencing (scRNA-seq) transcriptomics analysis of an MNV infection over time. Marked similarities were observed between certain global features of MNV infection in human cells compared to those previously reported in mouse cells by whole population transcriptomics such as downregulation of ribosome biogenesis, mitochondrial dysfunction, and cell cycle preference for G1. Our scRNA-seq analysis allowed further resolution of an infected cell population into distinct clusters with varying levels of viral RNA and interferon-stimulated gene ISG15 transcripts. Cells with high viral replication displayed downregulated ribosomal protein small (RPS) and large (RPL) genes and mitochondrial complexes I, III, IV, and V genes during exponential viral propagation. Ferritin subunit genes FTL and FTH1 were also downregulated during active MNV replication, suggesting that inhibition of iron metabolism may increase replication efficiency. Consistent with this, transcriptional activation of these genes with ferric ammonium citrate and overexpression of FTL lowered virus yields. Comparative studies of cells that support varying levels of norovirus replication efficiency, as determined by scRNA-seq may lead to improved human cell-based culture systems and effective viral interventions.IMPORTANCEHuman noroviruses cause acute gastroenteritis in all age groups. Vaccines and antiviral drugs are not yet available, in part, because it is difficult to propagate the viruses causing human disease in standard laboratory cell culture systems. In contrast, a norovirus found in mice [murine norovirus (MNV)] replicates efficiently in murine-based cell culture and has served as a model system. In this study, we established a new human intestinal cell line that was genetically modified to express the murine norovirus receptor so that the human cells became permissive to murine norovirus infection. We then defined the host response to MNV infection in the engineered human cell line at a single-cell resolution and identified cellular genes associated with the highest levels of MNV replication. This study may lead to the improvement of the current human norovirus cell culture systems and help to identify norovirus-host interactions that could be targeted for antiviral drugs.
Collapse
Affiliation(s)
- Yuki Matsushima
- Caliciviruses Section, Laboratory of Infectious Diseases, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Eric A. Levenson
- Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Natthawan Chaimongkol
- Caliciviruses Section, Laboratory of Infectious Diseases, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Loyall Harris
- Caliciviruses Section, Laboratory of Infectious Diseases, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Yongmei Zhao
- Sequencing Facility Bioinformatics Group, Bioinformatics and Computational Science Directorate, Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Sevilay Turan
- Leidos Biomedical Sciences, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Francisco Otaizo-Carrasquero
- Genomics Research Section, Research Technologies Branch, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Sundar Ganesan
- Biological Imaging Section, Research Technologies Branch, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Katherine M. Hornick
- Collaborative Bioinformatics Resource, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Craig Martens
- Genomics Research Section, Research Technologies Branch, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Stanislav V. Sosnovtsev
- Caliciviruses Section, Laboratory of Infectious Diseases, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Kim Y. Green
- Caliciviruses Section, Laboratory of Infectious Diseases, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
7
|
Cuvry A, Molineaux L, Gozalbo-Rovira R, Neyts J, de Witte P, Rodríguez-Díaz J, Rocha-Pereira J. Human norovirus disturbs intestinal motility and transit time through its capsid proteins. PLoS Pathog 2024; 20:e1012710. [PMID: 39602402 PMCID: PMC11602112 DOI: 10.1371/journal.ppat.1012710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 10/31/2024] [Indexed: 11/29/2024] Open
Abstract
Human norovirus (HuNoV) accounts for over 700 million cases of gastroenteritis annually. Episodes of HuNoV disease are characterized by vomiting and diarrhea as the two most prominent symptoms. Despite its prevalence, our understanding of the pathophysiological mechanisms triggered upon HuNoV infection is limited, mainly due to a lack of suitable animal models. Our aim was to use the recent HuNoV zebrafish larvae model to study the effect of HuNoV infection on intestinal motility and investigate whether one viral protein could act as an enterotoxin, as seen with rotavirus. We studied whether HuNoV infection affects the contraction frequency of the intestinal bulb and the posterior intestine as well as the transit time. Infection of larvae, following injection of a HuNoV GII.4-containing stool sample in the yolk, resulted in an increased contraction frequency in the intestinal bulb. A comparable effect was observed in serotonin-treated larvae, corresponding to the natural function of serotonin. The higher replication efficacy of HuNoV GII.4 likely explains why they have a more marked effect on gut motility, when compared to other genotypes. Additionally, transit time of fluorescent food was prolonged in HuNoV GII.4 infected larvae, suggesting a loss of coordination in bowel movements upon infection. To identify the proteins responsible for the effect, individual HuNoV non-structural proteins and virus-like particles (VLPs) were injected intraperitoneally (ip). VLPs carrying VP1/VP2, but not those with only VP1, induced increased contraction frequencies in the intestinal bulb in a dose-dependent manner. In conclusion, our findings suggest that the viral capsid and potentially the minor capsid protein VP2 play a crucial role in the aetiology of symptoms associated with HuNoV, potentially acting as a viral enterotoxin. This work contributes to the understanding of the pathophysiological mechanisms in HuNoV-induced disease and further attests zebrafish as a valuable HuNoV disease model.
Collapse
Affiliation(s)
- Arno Cuvry
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute, Virus-Host Interactions & Therapeutic Approaches (VITA) Research Group, Leuven, Belgium
- KU Leuven, Translationeel Onderzoek van Gastro-enterologische Aandoeningen (TARGID), Leuven, Belgium
| | - Lorane Molineaux
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute, Virus-Host Interactions & Therapeutic Approaches (VITA) Research Group, Leuven, Belgium
| | | | - Johan Neyts
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute, Virology, Antiviral Drug & Vaccine Research Group, Leuven, Belgium
| | - Peter de Witte
- KU Leuven, Laboratory for Molecular Biodiscovery, KU Leuven-Department of Pharmaceutical and Pharmacological Sciences, Leuven, Belgium
| | - Jesús Rodríguez-Díaz
- Department of Microbiology, School of Medicine, University of Valencia, Valencia, Spain
| | - Joana Rocha-Pereira
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute, Virus-Host Interactions & Therapeutic Approaches (VITA) Research Group, Leuven, Belgium
| |
Collapse
|
8
|
Ford-Siltz LA, Tohma K, Kihn K, Kendra JA, Deredge D, Wintrode P, Gao Y, Parra GI. Characterization of cross-reactive, non-neutralizing monoclonal antibodies against a pandemic GII.4 norovirus variant. Microbiol Spectr 2024; 12:e0114324. [PMID: 39470287 PMCID: PMC11619288 DOI: 10.1128/spectrum.01143-24] [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: 05/07/2024] [Accepted: 09/10/2024] [Indexed: 10/30/2024] Open
Abstract
Antibodies are thought to play a major role in protection against human norovirus infection. Mouse humoral responses closely mimic those of humans; thus, mouse models are used to characterize norovirus epitopes on the major viral capsid protein, VP1. We have developed a panel of mouse monoclonal antibodies (mAbs) produced against the last pandemic variant to emerge, Sydney 2012. While most mAbs (25/44) were mapped to variable antigenic sites on VP1, 19 of the mAbs were cross-reactive against multiple genotypes or GII.4 variants. Most (12/19) of the cross-reactive mAbs bound to the Shell domain and were cross-reactive with different GII noroviruses. Interestingly, mAb 30A11 exhibited cross-reactivity against all tested norovirus genotypes (GI, GII, GIV, and GIX). This mAb was mapped to a highly conserved region of the Shell domain (51PIDPWII57) using peptide ELISA and immunofluorescence. Of those mapping to the Protruding (P) domain, two (19C10 and 14B11) showed cross-reactivity with GII noroviruses. Using hydrogen-deuterium exchange mass spectrometry, we mapped 19C10 to a conserved region of the P domain near the P/Shell interface, which explains its cross-reactivity with different GII noroviruses and lack of histo-blood group antigen-blocking activity. Binding and mutational analyses showed that residues 518, 519, and 525 are important for 19C10 and 14B11 epitope recognition. While the antibodies described here are mostly non-neutralizing, they can be useful tools for research and diagnostics of noroviruses. The role of non-neutralizing, cross-reactive antibodies targeting different areas of the viral capsid merits further research to facilitate our understanding of immunity to norovirus infection and disease. IMPORTANCE To gain insights into the overall immune responses to human norovirus, we characterized non-neutralizing, cross-reactive monoclonal antibodies (mAbs) developed against a pandemic GII.4 norovirus. We determined the binding epitope of an antibody that exhibited cross-reactivity against all tested noroviruses, which makes it a useful tool for research and diagnostics. The epitope of two additional non-neutralizing mAbs was mapped to a less conserved region on the viral capsid protein, explaining their cross-reactivity patterns. Often overlooked, the role of non-neutralizing, cross-reactive mAbs merits further research to facilitate our understanding of immunity to norovirus infection and disease.
Collapse
Affiliation(s)
| | - Kentaro Tohma
- Division of Viral Products, CBER, FDA, Silver Spring, Maryland, USA
| | - Kyle Kihn
- University of Maryland School of Pharmacy, Baltimore, Maryland, USA
| | - Joseph A. Kendra
- Division of Viral Products, CBER, FDA, Silver Spring, Maryland, USA
| | - Daniel Deredge
- University of Maryland School of Pharmacy, Baltimore, Maryland, USA
| | - Patrick Wintrode
- University of Maryland School of Pharmacy, Baltimore, Maryland, USA
| | - Yamei Gao
- Division of Viral Products, CBER, FDA, Silver Spring, Maryland, USA
| | - Gabriel I. Parra
- Division of Viral Products, CBER, FDA, Silver Spring, Maryland, USA
| |
Collapse
|
9
|
Cheng C, Cai X, Li J, Zhang X, Xie Y, Zhang J. In Vitro Culture of Human Norovirus in the Last 20 Years. Biomedicines 2024; 12:2442. [PMID: 39595008 PMCID: PMC11592199 DOI: 10.3390/biomedicines12112442] [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: 09/14/2024] [Revised: 10/12/2024] [Accepted: 10/21/2024] [Indexed: 11/28/2024] Open
Abstract
Human noroviruses (HuNoVs) are the main pathogens that cause acute gastroenteritis and lead to huge economic losses annually. Due to the lack of suitable culture systems, the pathogenesis of HuNoVs and the development of vaccines and drugs have progressed slowly. Although researchers have employed various methods to culture HuNoVs in vitro in the last century, problems relating to the irreducibility, low viral titer, and non-infectiousness of the progeny virus should not be ignored. In 2016, researchers achieved the cultivation and successive passaging of some HuNoV genotypes using human intestinal enteroids, initially demonstrating the potential use of organoids in overcoming this challenge. This paper reviews the efforts made in the last 20 years to culture HuNoVs in vitro and discusses the superiority and limitations of employing human intestinal enteroids/organoids as an in vitro culture model for HuNoVs.
Collapse
Affiliation(s)
- Chao Cheng
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, Shanghai Institute of Infectious Disease and Biosecurity, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China; (C.C.); (J.L.); (X.Z.)
| | - Xia Cai
- Biosafety Level 3 Laboratory, Shanghai Medical College, Fudan University, Shanghai 200032, China;
| | - Jingjing Li
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, Shanghai Institute of Infectious Disease and Biosecurity, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China; (C.C.); (J.L.); (X.Z.)
| | - Xiaomeng Zhang
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, Shanghai Institute of Infectious Disease and Biosecurity, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China; (C.C.); (J.L.); (X.Z.)
| | - Youhua Xie
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, Shanghai Institute of Infectious Disease and Biosecurity, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China; (C.C.); (J.L.); (X.Z.)
| | - Junqi Zhang
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, Shanghai Institute of Infectious Disease and Biosecurity, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China; (C.C.); (J.L.); (X.Z.)
| |
Collapse
|
10
|
Barreto-Duran E, Synowiec A, Szczepański A, Gałuszka-Bulaga A, Węglarczyk K, Baj-Krzyworzeka M, Siedlar M, Bochenek M, Dufva M, Dogan AA, Lenart M, Pyrc K. Development of an intestinal mucosa ex vivo co-culture model to study viral infections. J Virol 2024; 98:e0098724. [PMID: 39212448 PMCID: PMC11495016 DOI: 10.1128/jvi.00987-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 07/17/2024] [Indexed: 09/04/2024] Open
Abstract
Studying viral infections necessitates well-designed cell culture models to deepen our understanding of diseases and develop effective treatments. In this study, we present a readily available ex vivo 3D co-culture model replicating the human intestinal mucosa. The model combines fully differentiated human intestinal epithelium (HIE) with human monocyte-derived macrophages (hMDMs) and faithfully mirrors the in vivo structural and organizational properties of intestinal mucosal tissues. Specifically, it mimics the lamina propria, basement membrane, and the air-exposed epithelial layer, enabling the pioneering observation of macrophage migration through the tissue to the site of viral infection. In this study, we applied the HIE-hMDMs model for the first time in viral infection studies, infecting the model with two globally significant viruses: severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and human norovirus GII.4. The results demonstrate the model's capability to support the replication of both viruses and show the antiviral role of macrophages, determined by their migration to the infection site and subsequent direct contact with infected epithelial cells. In addition, we evaluated the production of cytokines and chemokines in the intestinal niche, observing an increased interleukin-8 production during infection. A parallel comparison using a classical in vitro cell line model comprising Caco-2 and THP-1 cells for SARS-CoV-2 experiments confirmed the utility of the HIE-hMDMs model in viral infection studies. Our data show that the ex vivo tissue models hold important implications for advances in virology research.IMPORTANCEThe fabrication of intricate ex vivo tissue models holds important implications for advances in virology research. The co-culture model presented here provides distinct spatial and functional attributes not found in simplified models, enabling the evaluation of macrophage dynamics under severe acute respiratory syndrome coronavirus 2 and human norovirus (HuNoV) infections in the intestine. Moreover, these models, comprised solely of primary cells, facilitate the study of difficult-to-replicate viruses such as HuNoV, which cannot be studied in cell line models, and offer the opportunity for personalized treatment evaluations using patient cells. Similar co-cultures have been established for the study of bacterial infections and different characteristics of the intestinal tissue. However, to the best of our knowledge, a similar intestinal model for the study of viral infections has not been published before.
Collapse
Affiliation(s)
- Emilia Barreto-Duran
- Virogenetics Laboratory of Virology, Malopolska Centre of Biotechnology, Jagiellonian University, Krakow, Poland
| | - Aleksandra Synowiec
- Virogenetics Laboratory of Virology, Malopolska Centre of Biotechnology, Jagiellonian University, Krakow, Poland
- Doctoral School of Exact and Natural Sciences, Jagiellonian University, Kraków, Poland
| | - Artur Szczepański
- Virogenetics Laboratory of Virology, Malopolska Centre of Biotechnology, Jagiellonian University, Krakow, Poland
| | - Adrianna Gałuszka-Bulaga
- Department of Clinical Immunology, Institute of Pediatrics, Jagiellonian University Medical College, Krakow, Poland
| | - Kazimierz Węglarczyk
- Department of Clinical Immunology, Institute of Pediatrics, Jagiellonian University Medical College, Krakow, Poland
| | - Monika Baj-Krzyworzeka
- Department of Clinical Immunology, Institute of Pediatrics, Jagiellonian University Medical College, Krakow, Poland
| | - Maciej Siedlar
- Department of Clinical Immunology, Institute of Pediatrics, Jagiellonian University Medical College, Krakow, Poland
| | - Michał Bochenek
- Flow Cytometry Facility, Malopolska Centre of Biotechnology, Jagiellonian University, Krakow, Poland
| | - Martin Dufva
- Department of Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Asli Aybike Dogan
- Department of Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Marzena Lenart
- Virogenetics Laboratory of Virology, Malopolska Centre of Biotechnology, Jagiellonian University, Krakow, Poland
| | - Krzysztof Pyrc
- Virogenetics Laboratory of Virology, Malopolska Centre of Biotechnology, Jagiellonian University, Krakow, Poland
| |
Collapse
|
11
|
Lulla V, Sridhar A. Understanding neurotropic enteric viruses: routes of infection and mechanisms of attenuation. Cell Mol Life Sci 2024; 81:413. [PMID: 39365457 PMCID: PMC11452578 DOI: 10.1007/s00018-024-05450-6] [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: 07/10/2024] [Revised: 09/03/2024] [Accepted: 09/12/2024] [Indexed: 10/05/2024]
Abstract
The intricate connection between the gut and the brain involves multiple routes. Several viral families begin their infection cycle in the intestinal tract. However, amongst the long list of viral intestinal pathogens, picornaviruses, and astroviruses stand out for their ability to transition from the intestinal epithelia to central or peripheral nervous system cells. In immunocompromised, neonates and young children, these viral infections can manifest as severe diseases, such as encephalitis, meningitis, and acute flaccid paralysis. What confers this remarkable plasticity and makes them efficient in infecting cells of the gut and the brain axes? Here, we review the current understanding of the virus infection along the gut-brain axis for some enteric viruses and discuss the molecular mechanisms of their attenuation.
Collapse
Affiliation(s)
- Valeria Lulla
- Division of Virology, Department of Pathology, Addenbrooke's Hospital, University of Cambridge, Hills Road, Cambridge, CB2 0QQ, UK.
| | - Adithya Sridhar
- OrganoVIR Labs, Department of Pediatric Infectious Diseases, Amsterdam UMC, location Academic Medical Center, Amsterdam Institute for Reproduction and Development, University of Amsterdam, Meibergdreef 9, 1100 AZ, Amsterdam, The Netherlands
- OrganoVIR Labs, Department of Medical Microbiology, Amsterdam UMC, location Academic Medical Center, Amsterdam Institute for Infection and Immunity, University of Amsterdam, Meibergdreef 9, 1100 AZ, Amsterdam, The Netherlands
- Emma Center for Personalized Medicine, Amsterdam UMC, Amsterdam, The Netherlands
| |
Collapse
|
12
|
Tsukamoto B, Kurebayashi Y, Takahashi T, Abe Y, Ota R, Wakabayashi Y, Nishiie A, Minami A, Suzuki T, Takeuchi H. VP1 of human and murine noroviruses recognizes glycolipid sulfatide via the P domain. J Biochem 2024; 176:299-312. [PMID: 39012025 DOI: 10.1093/jb/mvae051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 06/19/2024] [Accepted: 07/12/2024] [Indexed: 07/17/2024] Open
Abstract
Noroviruses are a prevalent cause of human viral gastroenteritis, yet the precise mechanisms underlying their infection cycle, particularly their interactions with and entry into cells, remain poorly understood. Human norovirus (HuNoV) primarily targets human small intestinal epithelial cells, within which 3-O-sulfogalactosylceramide (sulfatide) ranks among the most abundant glycosphingolipids (GSLs). While sulfatide involvement in the binding and infection mechanism of several viruses has been documented, its interaction with noroviruses remains underexplored. This study investigated whether noroviruses interact with sulfatide. We found that the recombinant viral capsid protein VP1 of HuNoV (genogroups I and II) and murine norovirus (genogroup V) exhibited robust binding to sulfatide compared with other tested GSLs using enzyme-linked immunosorbent assay, thin-layer chromatography binding assay and real-time quantitative reverse transcription polymerase chain reaction binding assay. VP1 also bound 3-O-sulfated lactosylceramide, which shares the 3-O-sulfated galactose moiety with sulfatide. However, both VP1 and its P domain, identified as the sulfatide-binding domain, exhibited limited binding to structural analogues of sulfatide and other sulfated compounds. These findings suggest a specific recognition of the 3-O-sulfated galactose moiety. Notably, we found that sulfatide is a novel binding target for norovirus particles. Overall, our findings reveal a previously unknown norovirus-sulfatide interaction, proposing sulfatide as a potential candidate for norovirus infection receptors.
Collapse
Affiliation(s)
- Bunta Tsukamoto
- Department of Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Yuuki Kurebayashi
- Department of Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Tadanobu Takahashi
- Department of Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Yusuke Abe
- Department of Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Ryohei Ota
- Department of Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Yoshiki Wakabayashi
- Department of Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Anju Nishiie
- Department of Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Akira Minami
- Department of Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Takashi Suzuki
- Department of Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Hideyuki Takeuchi
- Department of Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| |
Collapse
|
13
|
Burton TD, Carrera Montoya J, Frota T, Mackenzie JM. Human norovirus cultivation models, immune response and vaccine landscape. Adv Virus Res 2024; 120:1-37. [PMID: 39455167 DOI: 10.1016/bs.aivir.2024.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2024]
Abstract
Norovirus infections are a leading cause of gastroenteritis worldwide. Despite the substantial global health burden and economic impact, there are currently no approved antiviral therapeutics or vaccines. Additionally, much of our knowledge of norovirus comes from experiments using surrogate viruses, such as murine norovirus and feline calicivirus. The challenge surrounding human norovirus research arises from a lack of robust cell culture systems and efficient animal models. In this review, we explore recent advances in the in vitro cultivation of human norovirus and reverse genetics systems and discuss commonly used in vivo models. We summarize the current understanding of both innate and adaptive immune responses to norovirus infection and provide an overview of vaccine strategies and the current clinical trial landscape, with a focus on the only vaccine candidate that has reached phase III clinical development stage.
Collapse
Affiliation(s)
- Thomas D Burton
- Department of Microbiology and Immunology, University of Melbourne, within the Peter Doherty Institute for Infection and Immunity, Parkville, Melbourne, VIC, Australia
| | - Julio Carrera Montoya
- Department of Microbiology and Immunology, University of Melbourne, within the Peter Doherty Institute for Infection and Immunity, Parkville, Melbourne, VIC, Australia
| | - Thalia Frota
- Department of Microbiology and Immunology, University of Melbourne, within the Peter Doherty Institute for Infection and Immunity, Parkville, Melbourne, VIC, Australia
| | - Jason M Mackenzie
- Department of Microbiology and Immunology, University of Melbourne, within the Peter Doherty Institute for Infection and Immunity, Parkville, Melbourne, VIC, Australia.
| |
Collapse
|
14
|
Hafner A, Meurs N, Garner A, Azar E, Kannan A, Passalacqua KD, Nagrath D, Wobus CE. Norovirus NS1/2 protein increases glutaminolysis for efficient viral replication. PLoS Pathog 2024; 20:e1011909. [PMID: 38976719 PMCID: PMC11257395 DOI: 10.1371/journal.ppat.1011909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 07/18/2024] [Accepted: 06/24/2024] [Indexed: 07/10/2024] Open
Abstract
Viruses are obligate intracellular parasites that rely on host cell metabolism for successful replication. Thus, viruses rewire host cell pathways involved in central carbon metabolism to increase the availability of building blocks for successful propagation. However, the underlying mechanisms of virus-induced alterations to host metabolism are largely unknown. Noroviruses (NoVs) are highly prevalent pathogens that cause sporadic and epidemic viral gastroenteritis. In the present study, we uncovered several strain-specific and shared host cell metabolic requirements of three murine norovirus (MNV) strains, MNV-1, CR3, and CR6. While all three strains required glycolysis, glutaminolysis, and the pentose phosphate pathway for optimal infection of macrophages, only MNV-1 relied on host oxidative phosphorylation. Furthermore, the first metabolic flux analysis of NoV-infected cells revealed that both glycolysis and glutaminolysis are upregulated during MNV-1 infection of macrophages. Glutamine deprivation affected the viral lifecycle at the stage of genome replication, resulting in decreased non-structural and structural protein synthesis, viral assembly, and egress. Mechanistic studies further showed that MNV infection and overexpression of the non-structural protein NS1/2 increased the enzymatic activity of the rate-limiting enzyme glutaminase. In conclusion, the inaugural investigation of NoV-induced alterations to host glutaminolysis identified NS1/2 as the first viral molecule for RNA viruses that regulates glutaminolysis either directly or indirectly. This increases our fundamental understanding of virus-induced metabolic alterations and may lead to improvements in the cultivation of human NoVs.
Collapse
Affiliation(s)
- Adam Hafner
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Noah Meurs
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Ari Garner
- Department of Microbiology, Immunology, and Inflammation, University of Illinois, Chicago, Illinois, United States of America
| | - Elaine Azar
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Aditya Kannan
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Karla D. Passalacqua
- Graduate Medical Education, Henry Ford Health, Detroit, Michigan, United States of America
| | - Deepak Nagrath
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Christiane E. Wobus
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan, United States of America
| |
Collapse
|
15
|
Omatola CA, Mshelbwala PP, Okolo MLO, Onoja AB, Abraham JO, Adaji DM, Samson SO, Okeme TO, Aminu RF, Akor ME, Ayeni G, Muhammed D, Akoh PQ, Ibrahim DS, Edegbo E, Yusuf L, Ocean HO, Akpala SN, Musa OA, Adamu AM. Noroviruses: Evolutionary Dynamics, Epidemiology, Pathogenesis, and Vaccine Advances-A Comprehensive Review. Vaccines (Basel) 2024; 12:590. [PMID: 38932319 PMCID: PMC11209302 DOI: 10.3390/vaccines12060590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 05/20/2024] [Accepted: 05/22/2024] [Indexed: 06/28/2024] Open
Abstract
Noroviruses constitute a significant aetiology of sporadic and epidemic gastroenteritis in human hosts worldwide, especially among young children, the elderly, and immunocompromised patients. The low infectious dose of the virus, protracted shedding in faeces, and the ability to persist in the environment promote viral transmission in different socioeconomic settings. Considering the substantial disease burden across healthcare and community settings and the difficulty in controlling the disease, we review aspects related to current knowledge about norovirus biology, mechanisms driving the evolutionary trends, epidemiology and molecular diversity, pathogenic mechanism, and immunity to viral infection. Additionally, we discuss the reservoir hosts, intra-inter host dynamics, and potential eco-evolutionary significance. Finally, we review norovirus vaccines in the development pipeline and further discuss the various host and pathogen factors that may complicate vaccine development.
Collapse
Affiliation(s)
- Cornelius Arome Omatola
- Department of Microbiology, Kogi State University, Anyigba 272102, Kogi State, Nigeria; (C.A.O.)
| | | | | | - Anyebe Bernard Onoja
- Department of Virology, University College Hospital, Ibadan 211101, Oyo State, Nigeria
| | - Joseph Oyiguh Abraham
- Department of Microbiology, Kogi State University, Anyigba 272102, Kogi State, Nigeria; (C.A.O.)
| | - David Moses Adaji
- Department of Biotechnology Science and Engineering, University of Alabama, Huntsville, AL 35899, USA
| | - Sunday Ocholi Samson
- Department of Molecular Biology, Biotechnology, and Biochemistry, Wrocław University of Science and Technology, Wybrzeże Wyspiańskiego 29, 50-370 Wrocław, Poland
| | - Therisa Ojomideju Okeme
- Department of Biological Sciences, Federal University Lokoja, Lokoja 260101, Kogi State, Nigeria
| | - Ruth Foluke Aminu
- Department of Microbiology, Kogi State University, Anyigba 272102, Kogi State, Nigeria; (C.A.O.)
| | - Monday Eneojo Akor
- Department of Microbiology, Kogi State University, Anyigba 272102, Kogi State, Nigeria; (C.A.O.)
| | - Gideon Ayeni
- Department of Biochemistry, Kogi State University, Anyigba 272102, Kogi State, Nigeria
| | - Danjuma Muhammed
- Epidemiology and Public Health Unit, Department of Biology, Universiti Putra, Seri Kembangan 43300, Malaysia
| | - Phoebe Queen Akoh
- Department of Microbiology, Kogi State University, Anyigba 272102, Kogi State, Nigeria; (C.A.O.)
| | | | - Emmanuel Edegbo
- Department of Microbiology, Kogi State University, Anyigba 272102, Kogi State, Nigeria; (C.A.O.)
| | - Lamidi Yusuf
- Department of Microbiology, Kogi State University, Anyigba 272102, Kogi State, Nigeria; (C.A.O.)
| | | | - Sumaila Ndah Akpala
- Department of Microbiology, Kogi State University, Anyigba 272102, Kogi State, Nigeria; (C.A.O.)
- Department of Biotechnology, Federal University Lokoja, Lokoja 260101, Kogi State, Nigeria
| | - Oiza Aishat Musa
- Department of Microbiology, Kogi State University, Anyigba 272102, Kogi State, Nigeria; (C.A.O.)
| | - Andrew Musa Adamu
- Australian Institute of Tropical Health and Medicine, James Cook University, Townsville 4811, QLD, Australia
- College of Public Health Medical and Veterinary Sciences, James Cook University, Townsville 4811, QLD, Australia
- Centre for Tropical Biosecurity, James Cook University, Townsville 4811, QLD, Australia
| |
Collapse
|
16
|
Chen Y, Jia Y, Zhu X, Xu L, Li H, Li H. Self-Powered Immunoassay of Norovirus in Human Stools by π-Electron-Rich Homojunction for Enhanced Charge Transfer. ACS Sens 2024; 9:2429-2439. [PMID: 38668680 DOI: 10.1021/acssensors.4c00108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Abstract
Norovirus (NoV) stands as a significant causative agent of nonbacterial acute gastroenteritis on a global scale, presenting a substantial threat to public health. Hence, the development of simple and rapid analytical techniques for NoV detection holds great importance in preventing and controlling the outbreak of the epidemic. In this work, a self-powered photoelectrochemical (PEC) immunosensor of NoV capsid protein (VP1) was proposed by the π-electron-rich carbon nitride homojunction (ER-CNH) as the photoanode. C4N2 ring derived from π-rich locust bean gum was introduced into the tri-s-triazine structure, creating a large π-delocalized conjugated carbon nitride homojunction. This strategy enhances the C/N atomic ratio, which widens light utilization, narrows the bandgap, and optimizes the electronic band structure of carbon nitride. By introduction of a π-rich conjugated structure, p-type domains were induced within n-type domains to build the internal electric field at the interface, thus forming a p-n homojunction to boost carrier separation and transfer. The ER-CNH photoanode exhibited excellent photoelectric performance and water oxidation capacity. Since VP1 inhibits the water oxidation of the ER-CNH photoanode, the open-circuit potential of the as-prepared PEC immunosensor system was reduced for detecting NoV VP1. The self-powered PEC immunosensor achieved a remarkably low detection limit (∼5 fg mL-1) and displayed high stability and applicability for actual stool samples. This research serves as a foundation concept for constructing immunosensors to detect other viruses and promotes the application of self-powered systems for life safety.
Collapse
Affiliation(s)
- Yun Chen
- School of Chemistry and Chemical Engineering, Institute for Energy Research, School of the Environment and Safety Engineering, Institute of Quantum and Sustainable Technology, Jiangsu University, Zhenjiang 212013, P. R. China
| | - Yunfan Jia
- School of Chemistry and Chemical Engineering, Institute for Energy Research, School of the Environment and Safety Engineering, Institute of Quantum and Sustainable Technology, Jiangsu University, Zhenjiang 212013, P. R. China
- Jiangsu Rugao Senior High School, Rugao 226500, P. R. China
| | - Xingwang Zhu
- School of Environmental Science and Engineering, College of Mechanical Engineering, Yangzhou University, Yangzhou 225002, P. R. China
| | - Li Xu
- School of Chemistry and Chemical Engineering, Institute for Energy Research, School of the Environment and Safety Engineering, Institute of Quantum and Sustainable Technology, Jiangsu University, Zhenjiang 212013, P. R. China
| | - Henan Li
- School of Chemistry and Chemical Engineering, Institute for Energy Research, School of the Environment and Safety Engineering, Institute of Quantum and Sustainable Technology, Jiangsu University, Zhenjiang 212013, P. R. China
| | - Huaming Li
- School of Chemistry and Chemical Engineering, Institute for Energy Research, School of the Environment and Safety Engineering, Institute of Quantum and Sustainable Technology, Jiangsu University, Zhenjiang 212013, P. R. China
| |
Collapse
|
17
|
Chandran S, Gibson KE. Improving the Detection and Understanding of Infectious Human Norovirus in Food and Water Matrices: A Review of Methods and Emerging Models. Viruses 2024; 16:776. [PMID: 38793656 PMCID: PMC11125872 DOI: 10.3390/v16050776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Revised: 05/02/2024] [Accepted: 05/10/2024] [Indexed: 05/26/2024] Open
Abstract
Human norovirus (HuNoV) is a leading global cause of viral gastroenteritis, contributing to numerous outbreaks and illnesses annually. However, conventional cell culture systems cannot support the cultivation of infectious HuNoV, making its detection and study in food and water matrices particularly challenging. Recent advancements in HuNoV research, including the emergence of models such as human intestinal enteroids (HIEs) and zebrafish larvae/embryo, have significantly enhanced our understanding of HuNoV pathogenesis. This review provides an overview of current methods employed for HuNoV detection in food and water, along with their associated limitations. Furthermore, it explores the potential applications of the HIE and zebrafish larvae/embryo models in detecting infectious HuNoV within food and water matrices. Finally, this review also highlights the need for further optimization and exploration of these models and detection methods to improve our understanding of HuNoV and its presence in different matrices, ultimately contributing to improved intervention strategies and public health outcomes.
Collapse
Affiliation(s)
| | - Kristen E. Gibson
- Department of Food Science, Center for Food Safety, University of Arkansas System Division of Agriculture, Fayetteville, AR 72704, USA;
| |
Collapse
|
18
|
Nilsson J, Rimkute I, Sihlbom C, Tenge VR, Lin SC, Atmar RL, Estes MK, Larson G. N-glycoproteomic analyses of human intestinal enteroids, varying in histo-blood group geno- and phenotypes, reveal a wide repertoire of fucosylated glycoproteins. Glycobiology 2024; 34:cwae029. [PMID: 38590172 PMCID: PMC11041853 DOI: 10.1093/glycob/cwae029] [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: 11/03/2023] [Revised: 03/12/2024] [Accepted: 04/03/2024] [Indexed: 04/10/2024] Open
Abstract
Human noroviruses, globally the main cause of viral gastroenteritis, show strain specific affinity for histo-blood group antigens (HBGA) and can successfully be propagated ex vivo in human intestinal enteroids (HIEs). HIEs established from jejunal stem cells of individuals with different ABO, Lewis and secretor geno- and phenotypes, show varying susceptibility to such infections. Using bottom-up glycoproteomic approaches we have defined and compared the N-linked glycans of glycoproteins of seven jejunal HIEs. Membrane proteins were extracted, trypsin digested, and glycopeptides enriched by hydrophilic interaction liquid chromatography and analyzed by nanoLC-MS/MS. The Byonic software was used for glycopeptide identification followed by hands-on verifications and interpretations. Glycan structures and attachment sites were identified from MS2 spectra obtained by higher-energy collision dissociation through analysis of diagnostic saccharide oxonium ions (B-ions), stepwise glycosidic fragmentation of the glycans (Y-ions), and peptide sequence ions (b- and y-ions). Altogether 694 unique glycopeptides from 93 glycoproteins were identified. The N-glycans encompassed pauci- and oligomannose, hybrid- and complex-type structures. Notably, polyfucosylated HBGA-containing glycopeptides of the four glycoproteins tetraspanin-8, carcinoembryonic antigen-related cell adhesion molecule 5, sucrose-isomaltase and aminopeptidase N were especially prominent and were characterized in detail and related to donor ABO, Lewis and secretor types of each HIE. Virtually no sialylated N-glycans were identified for these glycoproteins suggesting that terminal sialylation was infrequent compared to fucosylation and HBGA biosynthesis. This approach gives unique site-specific information on the structural complexity of N-linked glycans of glycoproteins of human HIEs and provides a platform for future studies on the role of host glycoproteins in gastrointestinal infectious diseases.
Collapse
Affiliation(s)
- Jonas Nilsson
- Department of Laboratory Medicine, Institute of Biomedicine, University of Gothenburg, Sahlgrenska University Hospital, Bruna Stråket 16, SE 413 45, Gothenburg, Sweden
- Department of Clinical Chemistry, Region Västra Götaland, Sahlgrenska University Hospital, Bruna Stråket 16, SE 413 45, Gothenburg, Sweden
- Proteomics Core Facilities, Sahlgrenska Academy, University of Gothenburg, Medicinaregatan 9E, SE 413 90, Gothenburg, Sweden
| | - Inga Rimkute
- Department of Laboratory Medicine, Institute of Biomedicine, University of Gothenburg, Sahlgrenska University Hospital, Bruna Stråket 16, SE 413 45, Gothenburg, Sweden
- Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Medicinaregatan 7A, SE 413 90, Gothenburg, Sweden
| | - Carina Sihlbom
- Proteomics Core Facilities, Sahlgrenska Academy, University of Gothenburg, Medicinaregatan 9E, SE 413 90, Gothenburg, Sweden
| | - Victoria R Tenge
- Department of Molecular Virology, Baylor College School of Medicine, One Baylor Plaza, Houston, TX 770 30, United States
| | - Shih-Ching Lin
- Department of Molecular Virology, Baylor College School of Medicine, One Baylor Plaza, Houston, TX 770 30, United States
- Present address: Department of Medicine, Washington University in St. Louis, St. Louis, MO, United States
| | - Robert L Atmar
- Department of Molecular Virology, Baylor College School of Medicine, One Baylor Plaza, Houston, TX 770 30, United States
- Department of Medicine, Baylor College of Medicine, One Baylor Plaza, Houston, TX 770 30, United States
| | - Mary K Estes
- Department of Molecular Virology, Baylor College School of Medicine, One Baylor Plaza, Houston, TX 770 30, United States
- Department of Medicine, Baylor College of Medicine, One Baylor Plaza, Houston, TX 770 30, United States
| | - Göran Larson
- Department of Laboratory Medicine, Institute of Biomedicine, University of Gothenburg, Sahlgrenska University Hospital, Bruna Stråket 16, SE 413 45, Gothenburg, Sweden
- Department of Clinical Chemistry, Region Västra Götaland, Sahlgrenska University Hospital, Bruna Stråket 16, SE 413 45, Gothenburg, Sweden
| |
Collapse
|
19
|
Hayashi T, Kobayashi S, Hirano J, Murakami K. Human norovirus cultivation systems and their use in antiviral research. J Virol 2024; 98:e0166323. [PMID: 38470106 PMCID: PMC11019851 DOI: 10.1128/jvi.01663-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/13/2024] Open
Abstract
Human norovirus (HuNoV) is a major cause of acute gastroenteritis and foodborne diseases, affecting all age groups. Despite its clinical needs, no approved antiviral therapies are available. Since the discovery of HuNoV in 1972, studies on anti-norovirals, mechanism of HuNoV infection, viral inactivation, etc., have been hampered by the lack of a robust laboratory-based cultivation system for HuNoV. A recent breakthrough in the development of HuNoV cultivation systems has opened opportunities for researchers to investigate HuNoV biology in the context of de novo HuNoV infections. A tissue stem cell-derived human intestinal organoid/enteroid (HIO) culture system is one of those that supports HuNoV replication reproducibly and, to our knowledge, is most widely distributed to laboratories worldwide to study HuNoV and develop therapeutic strategies. This review summarizes recently developed HuNoV cultivation systems, including HIO, and their use in antiviral studies.
Collapse
Affiliation(s)
- Tsuyoshi Hayashi
- Department of Virology II, National Institute of Infectious Diseases, Tokyo, Japan
| | - Sakura Kobayashi
- Department of Virology II, National Institute of Infectious Diseases, Tokyo, Japan
| | - Junki Hirano
- Laboratory of Virus Control, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Kosuke Murakami
- Department of Virology II, National Institute of Infectious Diseases, Tokyo, Japan
- Center for Emergency Preparedness and Response, National Institute of Infectious Diseases, Tokyo, Japan
| |
Collapse
|
20
|
Wasielewski VV, Itani TM, Zakharova YA, Semenov AV. Current trends and new approaches for human norovirus replication in cell culture: a literature review. Arch Virol 2024; 169:71. [PMID: 38459228 DOI: 10.1007/s00705-024-05999-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 01/21/2024] [Indexed: 03/10/2024]
Abstract
Human norovirus (HuNoV) is one of the world's leading causes of acute gastroenteritis. At present, effective reproduction of the virus in cell cultures remains a challenge for virologists, as there is a lack of a permissive cell line that allows the entire viral life cycle to be reproduced. This is a barrier to the study of the HuNoV life cycle, its tropism, and virus-host interactions. It is also a major hurdle for the development of viral detection platforms, and ultimately for the development of therapeutics. The lack of an inexpensive, technically simple, and easily implemented cultivation method also negatively affects our ability to evaluate the efficacy of a variety of control measures (disinfectants, food processes) for human norovirus. In the process of monitoring this pathogen, it is necessary to detect infectious viral particles in water, food, and other environmental samples. Therefore, improvement of in vitro replication of HuNoV is still needed. In this review, we discuss current trends and new approaches to HuNoV replication in cell culture. We highlight ways in which previous research on HuNoV and other noroviruses has guided and influenced the development of new HuNoV culture systems and discuss the improvement of in vitro replication of HuNoV.
Collapse
Affiliation(s)
- Valentin V Wasielewski
- Federal Service for Surveillance on Consumer Rights Protection and Human Wellbeing, Federal Scientific Research Institute of Viral Infections «Virome», Ekaterinburg, 620030, Russian Federation
| | - Tarek M Itani
- Federal Service for Surveillance on Consumer Rights Protection and Human Wellbeing, Federal Scientific Research Institute of Viral Infections «Virome», Ekaterinburg, 620030, Russian Federation.
| | - Yuliya A Zakharova
- Institute of Disinfectology of the F.F. Erisman Federal Scientific Centre of Hygiene Rospotrebnadzor, Mosсow, Russian Federation
| | - Aleksandr V Semenov
- Federal Service for Surveillance on Consumer Rights Protection and Human Wellbeing, Federal Scientific Research Institute of Viral Infections «Virome», Ekaterinburg, 620030, Russian Federation
- Ural Federal University named after the First President of Russia B.N. Yeltsin, Ekaterinburg, Russian Federation
| |
Collapse
|
21
|
Bemark M, Pitcher MJ, Dionisi C, Spencer J. Gut-associated lymphoid tissue: a microbiota-driven hub of B cell immunity. Trends Immunol 2024; 45:211-223. [PMID: 38402045 PMCID: PMC11227984 DOI: 10.1016/j.it.2024.01.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 01/29/2024] [Accepted: 01/30/2024] [Indexed: 02/26/2024]
Abstract
The diverse gut microbiota, which is associated with mucosal health and general wellbeing, maintains gut-associated lymphoid tissues (GALT) in a chronically activated state, including sustainment of germinal centers in a context of high antigenic load. This influences the rules for B cell engagement with antigen and the potential consequences. Recent data have highlighted differences between GALT and other lymphoid tissues. For example, GALT propagates IgA responses against glycans that show signs of having been generated in germinal centers. Other findings suggest that humans are among those species where GALT supports the diversification, propagation, and possibly selection of systemic B cells. Here, we review novel findings that identify GALT as distinctive, and able to support these processes.
Collapse
Affiliation(s)
- Mats Bemark
- Department of Translational Medicine - Human Immunology, Lund University, J Waldenströms gata 35, Malmö, Sweden; Department of Clinical Immunology and Transfusion Medicine, Sahlgrenska University Hospital, Region Västra Götaland, Gothenburg, Sweden.
| | - Michael J Pitcher
- Peter Gorer Department of Immunobiology, King's College London, Guy's Hospital Campus, St Thomas' Street, London SE1 9RT, UK
| | - Chiara Dionisi
- Peter Gorer Department of Immunobiology, King's College London, Guy's Hospital Campus, St Thomas' Street, London SE1 9RT, UK
| | - Jo Spencer
- Peter Gorer Department of Immunobiology, King's College London, Guy's Hospital Campus, St Thomas' Street, London SE1 9RT, UK.
| |
Collapse
|
22
|
A rhesus macaque model of human norovirus infection. Nat Microbiol 2024; 9:593-594. [PMID: 38378852 DOI: 10.1038/s41564-023-01597-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2024]
|
23
|
Rimkute I, Chaimongkol N, Woods KD, Nagata BM, Darko S, Gudbole S, Henry AR, Sosnovtsev SV, Olia AS, Verardi R, Bok K, Todd JP, Woodward R, Kwong PD, Douek DC, Alves DA, Green KY, Roederer M. A non-human primate model for human norovirus infection. Nat Microbiol 2024; 9:776-786. [PMID: 38321182 DOI: 10.1038/s41564-023-01585-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 12/12/2023] [Indexed: 02/08/2024]
Abstract
Norovirus infection can cause gastrointestinal disease in humans. Development of therapies and vaccines against norovirus have been limited by the lack of a suitable and reliable animal model. Here we established rhesus macaques as an animal model for human norovirus infection. We show that rhesus macaques are susceptible to oral infection with human noroviruses from two different genogroups. Variation in duration of virus shedding (days to weeks) between animals, evolution of the virus over the time of infection, induction of virus-specific adaptive immune responses, susceptibility to reinfection and preferential replication of norovirus in the jejunum of rhesus macaques was similar to infection reported in humans. We found minor pathological signs and changes in epithelial cell surface glycosylation patterns in the small intestine during infection. Detection of viral protein and RNA in intestinal biopsies confirmed the presence of the virus in chromogranin A-expressing epithelial cells, as it does in humans. Thus, rhesus macaques are a promising non-human primate model to evaluate vaccines and therapeutics against norovirus disease.
Collapse
Affiliation(s)
- Inga Rimkute
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Natthawan Chaimongkol
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Kamron D Woods
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Bianca M Nagata
- Comparative Medicine Branch, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, MD, USA
| | - Samuel Darko
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Sucheta Gudbole
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Amy R Henry
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Stanislav V Sosnovtsev
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Adam S Olia
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Raffaello Verardi
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Karin Bok
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - John-Paul Todd
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Ruth Woodward
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Peter D Kwong
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Daniel C Douek
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Derron A Alves
- Comparative Medicine Branch, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, MD, USA
| | - Kim Y Green
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Mario Roederer
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA.
| |
Collapse
|
24
|
Shaffer M, Huynh K, Costantini V, Vinjé J, Bibby K. Heat inactivation of aqueous viable norovirus and MS2 bacteriophage. J Appl Microbiol 2024; 135:lxae033. [PMID: 38341278 PMCID: PMC11178036 DOI: 10.1093/jambio/lxae033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 02/05/2024] [Accepted: 02/08/2024] [Indexed: 02/12/2024]
Abstract
AIMS This study aimed to compare the heat inactivation kinetics of viable human norovirus with the surrogate, MS2 bacteriophage as well as assess the decay of the RNA signal. METHODS AND RESULTS Human intestinal enteroids were used to analyze the heat inactivation kinetics of viable human norovirus compared to the surrogate MS2 bacteriophage, which was cultured using a plaque assay. Norovirus decay rates were 0.22 min-1, 0.68 min-1, and 1.11 min-1 for 50°C, 60°C, and 70°C, respectively, and MS2 bacteriophage decay rates were 0.0065 min-1, 0.045 min-1, and 0.16 min-1 for 50°C, 60°C, and 70°C, respectively. Norovirus had significantly higher decay rates than MS2 bacteriophage at all tested temperatures (P = .002-.007). No decrease of RNA titers as measured by reverse transcription-PCR for both human norovirus and MS2 bacteriophage over time was observed, indicating molecular methods do not accurately depict viable human norovirus after heat inactivation and treatment efficiency is underestimated. CONCLUSIONS Overall, our data demonstrate that MS2 bacteriophage is a conservative surrogate to measure heat inactivation and potentially overestimates the infectious risk of norovirus. Furthermore, this study corroborates that measuring viral RNA titers, as evaluated by PCR methods, does not correlate with the persistence of viable norovirus under heat inactivation.
Collapse
Affiliation(s)
- Marlee Shaffer
- Department of Civil and Environmental Engineering and Earth Sciences, University of Notre Dame, IN 46556, United States
| | - Kimberly Huynh
- National Calicivirus Laboratory, Division of Viral Diseases, Centers for Disease Control and Prevention, Atlanta, GA, United States
| | - Verónica Costantini
- National Calicivirus Laboratory, Division of Viral Diseases, Centers for Disease Control and Prevention, Atlanta, GA, United States
| | - Jan Vinjé
- National Calicivirus Laboratory, Division of Viral Diseases, Centers for Disease Control and Prevention, Atlanta, GA, United States
| | - Kyle Bibby
- Department of Civil and Environmental Engineering and Earth Sciences, University of Notre Dame, IN 46556, United States
| |
Collapse
|
25
|
Huang L, Xie S, Zhang Y, Du W, Liang X, Pan W, Yang F, Niu R, Chen H, Geng L, Xiang L, Gong S, Xu W. The novel mechanism of human norovirus induced diarrhea: Activation of PKD2 caused by HuNoVs destroyed AQP3 expression through AP2γ in intestinal epithelial cells. Life Sci 2024; 337:122348. [PMID: 38103725 DOI: 10.1016/j.lfs.2023.122348] [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: 09/26/2023] [Revised: 11/30/2023] [Accepted: 12/09/2023] [Indexed: 12/19/2023]
Abstract
Our previous work has demonstrated protein kinase D2 (PKD2) played a critical influence in experimental colitis in animal. However, the role of PKD2 in human norovirus (HuNoVs)-induced diarrhea remained unknown. Aquaporin 3 (AQP3) expression, a critical protein mediating diarrhea, was assessed by western blot, qRT-PCR in intestinal epithelial cells (IECs). Luciferase, IF, IP and ChIP assay were used to explore the mechanism through which HuNoVs regulated AQP3. Herein, we found that AQP3 expression was drastically decreased in IECs in response to VP1 transfection, the major capsid protein of HuNoVs, or HuNoVs infection. Mechanistically, HuNoVs triggered phosphorylation of PKD2 through TLR2/MyD88/IRAK4, which further inhibited AP2γ activation and nuclear translocation, leading to suppress AQP3 transactivation in IECs. Most importantly, PKD2 interacted with MyD88/IRAK4, and VP1 overexpression enhanced this complex form, which, in turn, to increase PKD2 phosphorylation. In addition, endogenous PKD2 interacted with AP2γ, and this interaction was enhanced in response to HuNoVs treatment, and subsequently resulting in AP2γ phosphorylation inhibition. Moreover, inhibition of PKD2 activation could reverse the inhibitory effect of HuNoVs on AQP3 expression. In summary, we established a novel mechanism that HuNoV inhibited AQP3 expression through TLR2/MyD88/IRAK4/PKD2 signaling pathway, targeting PKD2 activity could be a promising strategy for prevention of HuNoVs-induced gastroenteritis.
Collapse
Affiliation(s)
- Ling Huang
- Department of Gastroenterology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China; Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou 510623, China
| | - Shuping Xie
- Department of Gastroenterology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China; Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou 510623, China
| | - Yuhua Zhang
- Department of Pediatrics, Putian Ninety-Five Hospital, Putian 351100, China
| | - Wenjun Du
- Department of Gastroenterology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China; Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou 510623, China
| | - Xinhua Liang
- Department of Gastroenterology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China; Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou 510623, China
| | - Wenxu Pan
- Department of Gastroenterology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China; Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou 510623, China
| | - Fangying Yang
- Department of Gastroenterology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China; Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou 510623, China
| | - Rongwei Niu
- Department of Gastroenterology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Huan Chen
- Department of Gastroenterology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China; Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou 510623, China
| | - Lanlan Geng
- Department of Gastroenterology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China; Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou 510623, China
| | - Li Xiang
- Department of Gastroenterology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China; Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou 510623, China.
| | - Sitang Gong
- Department of Gastroenterology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China; Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou 510623, China.
| | - Wanfu Xu
- Department of Gastroenterology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China; Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou 510623, China.
| |
Collapse
|
26
|
Roux E, Willms RJ, Van Dycke J, Cortes Calabuig Á, Van Espen L, Schoofs G, Matthijnssens J, Neyts J, de Witte P, Foley E, Rocha-Pereira J. Transcriptional profiling of zebrafish intestines identifies macrophages as host cells for human norovirus infection. Gut Microbes 2024; 16:2431167. [PMID: 39584740 PMCID: PMC11591593 DOI: 10.1080/19490976.2024.2431167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 10/25/2024] [Accepted: 11/11/2024] [Indexed: 11/26/2024] Open
Abstract
Human noroviruses (HuNoVs) are a major cause of diarrheal disease, yet critical aspects of their biology, including cellular tropism, remain unclear. Although research has traditionally focused on the intestinal epithelium, the hypothesis that HuNoV infects macrophages has been recurrently discussed and is investigated here using a zebrafish larval model. Through single-cell RNA sequencing of dissected zebrafish intestines, we unbiasedly identified macrophages as host cells for HuNoV replication, with all three open reading frames mapped to individual macrophages. Notably, HuNoV preferentially infects actively phagocytosing inflammatory macrophages. HuNoV capsid proteins and double-stranded RNA colocalized within intestinal macrophages of infected zebrafish larvae, and the negative-strand RNA intermediate was detected within FACS-sorted macrophages. Flow cytometry confirmed viral replication within these macrophages, constituting approximately 23% of HuNoV's host cells. Identifying macrophages as host cells prompts a reevaluation of their role in HuNoV pathogenesis, offering new directions for understanding and controlling this infection.
Collapse
Affiliation(s)
- Emma Roux
- Department of Microbiology, Immunology and Transplantation, Rega Institute, Virus-Host Interactions & Therapeutic Approaches (VITA) Research Group, KU Leuven, Leuven, Belgium
| | - Reegan J. Willms
- Department of Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Jana Van Dycke
- Department of Microbiology, Immunology and Transplantation, Rega Institute, Virus-Host Interactions & Therapeutic Approaches (VITA) Research Group, KU Leuven, Leuven, Belgium
| | | | - Lore Van Espen
- Department of Microbiology, Immunology and Transplantation, Rega Institute, Laboratory of Clinical and Epidemiological Virology, KU Leuven, Leuven, Belgium
| | - Geert Schoofs
- Department of Microbiology, Immunology and Transplantation, Rega Institute, Molecular Structural and Translational Virology Research Group, KU Leuven, Leuven, Belgium
| | - Jelle Matthijnssens
- Department of Microbiology, Immunology and Transplantation, Rega Institute, Laboratory of Clinical and Epidemiological Virology, KU Leuven, Leuven, Belgium
| | - Johan Neyts
- Department of Microbiology, Immunology and Transplantation, Rega Institute, Virology, Antiviral Drug & Vaccine Research Group, KU Leuven, Leuven, Belgium
| | - Peter de Witte
- Department of Pharmaceutical and Pharmacological Sciences, Laboratory for Molecular Biodiscovery, KU Leuven, Leuven, Belgium
| | - Edan Foley
- Department of Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Joana Rocha-Pereira
- Department of Microbiology, Immunology and Transplantation, Rega Institute, Virus-Host Interactions & Therapeutic Approaches (VITA) Research Group, KU Leuven, Leuven, Belgium
| |
Collapse
|
27
|
Mills JT, Minogue SC, Snowden JS, Arden WKC, Rowlands DJ, Stonehouse NJ, Wobus CE, Herod MR. Amino acid substitutions in norovirus VP1 dictate host dissemination via variations in cellular attachment. J Virol 2023; 97:e0171923. [PMID: 38032199 PMCID: PMC10734460 DOI: 10.1128/jvi.01719-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 11/03/2023] [Indexed: 12/01/2023] Open
Abstract
IMPORTANCE All viruses initiate infection by utilizing receptors to attach to target host cells. These virus-receptor interactions can therefore dictate viral replication and pathogenesis. Understanding the nature of virus-receptor interactions could also be important for the development of novel therapies. Noroviruses are non-enveloped icosahedral viruses of medical importance. They are a common cause of acute gastroenteritis with no approved vaccine or therapy and are a tractable model for studying fundamental virus biology. In this study, we utilized the murine norovirus model system to show that variation in a single amino acid of the major capsid protein alone can affect viral infectivity through improved attachment to suspension cells. Modulating plasma membrane mobility reduced infectivity, suggesting an importance of membrane mobility for receptor recruitment and/or receptor conformation. Furthermore, different substitutions at this site altered viral tissue distribution in a murine model, illustrating how in-host capsid evolution could influence viral infectivity and/or immune evasion.
Collapse
Affiliation(s)
- Jake T. Mills
- Astbury Centre for Structural Molecular Biology, School of Molecular & Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Susanna C. Minogue
- Astbury Centre for Structural Molecular Biology, School of Molecular & Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Joseph S. Snowden
- Astbury Centre for Structural Molecular Biology, School of Molecular & Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Wynter K. C. Arden
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - David J. Rowlands
- Astbury Centre for Structural Molecular Biology, School of Molecular & Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Nicola J. Stonehouse
- Astbury Centre for Structural Molecular Biology, School of Molecular & Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Christiane E. Wobus
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Morgan R. Herod
- Astbury Centre for Structural Molecular Biology, School of Molecular & Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| |
Collapse
|
28
|
Poydenot F, Lebreton A, Haiech J, Andreotti B. At the crossroads of epidemiology and biology: Bridging the gap between SARS-CoV-2 viral strain properties and epidemic wave characteristics. Biochimie 2023; 213:54-65. [PMID: 36931337 PMCID: PMC10017177 DOI: 10.1016/j.biochi.2023.03.006] [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: 09/29/2022] [Revised: 02/08/2023] [Accepted: 03/14/2023] [Indexed: 03/17/2023]
Abstract
The COVID-19 pandemic has given rise to numerous articles from different scientific fields (epidemiology, virology, immunology, airflow physics …) without any effort to link these different insights. In this review, we aim to establish relationships between epidemiological data and the characteristics of the virus strain responsible for the epidemic wave concerned. We have carried out this study on the Wuhan, Alpha, Delta and Omicron strains allowing us to illustrate the evolution of the relationships we have highlighted according to these different viral strains. We addressed the following questions. 1) How can the mean infectious dose (one quantum, by definition in epidemiology) be measured and expressed as an amount of viral RNA molecules (in genome units, GU) or as a number of replicative viral particles (in plaque-forming units, PFU)? 2) How many infectious quanta are exhaled by an infected person per unit of time? 3) How many infectious quanta are exhaled, on average, integrated over the whole contagious period? 4) How do these quantities relate to the epidemic reproduction rate R as measured in epidemiology, and to the viral load, as measured by molecular biological methods? 5) How has the infectious dose evolved with the different strains of SARS-CoV-2? We make use of state-of-the-art modelling, reviewed and explained in the appendix of the article (Supplemental Information, SI), to answer these questions using data from the literature in both epidemiology and virology. We have considered the modification of these relationships according to the vaccination status of the population.
Collapse
Affiliation(s)
- Florian Poydenot
- Laboratoire de Physique de l'Ecole Normale Supérieure (LPENS), CNRS UMR 8023, Ecole Normale Supérieure, Université PSL, Sorbonne Université, and Université de Paris, 75005, Paris, France
| | - Alice Lebreton
- Institut de Biologie de l'ENS (IBENS), École Normale Supérieure, CNRS, INSERM, Université PSL, 75005, Paris, France; INRAE, Micalis Institute, 78350, Jouy-en-Josas, France
| | - Jacques Haiech
- CNRS UMR7242 BSC ESBS, 300 Bd Sébastien Brant, CS 10413, 67412, Illkirch cedex, France.
| | - Bruno Andreotti
- Laboratoire de Physique de l'Ecole Normale Supérieure (LPENS), CNRS UMR 8023, Ecole Normale Supérieure, Université PSL, Sorbonne Université, and Université de Paris, 75005, Paris, France
| |
Collapse
|
29
|
Bernard-Raichon L, Cadwell K. Immunomodulation by Enteric Viruses. Annu Rev Virol 2023; 10:477-502. [PMID: 37380186 DOI: 10.1146/annurev-virology-111821-112317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/30/2023]
Abstract
Enteric viruses display intricate adaptations to the host mucosal immune system to successfully reproduce in the gastrointestinal tract and cause maladies ranging from gastroenteritis to life-threatening disease upon extraintestinal dissemination. However, many viral infections are asymptomatic, and their presence in the gut is associated with an altered immune landscape that can be beneficial or adverse in certain contexts. Genetic variation in the host and environmental factors including the bacterial microbiota influence how the immune system responds to infections in a remarkably viral strain-specific manner. This immune response, in turn, determines whether a given virus establishes acute versus chronic infection, which may have long-lasting consequences such as susceptibility to inflammatory disease. In this review, we summarize our current understanding of the mechanisms involved in the interaction between enteric viruses and the immune system that underlie the impact of these ubiquitous infectious agents on our health.
Collapse
Affiliation(s)
- Lucie Bernard-Raichon
- Cell Biology Department, New York University Grossman School of Medicine, New York, NY, USA
| | - Ken Cadwell
- Division of Gastroenterology and Hepatology, Department of Medicine; Department of Systems Pharmacology and Translational Therapeutics; Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA;
| |
Collapse
|
30
|
Ingle H, Makimaa H, Aggarwal S, Deng H, Foster L, Li Y, Kennedy EA, Peterson ST, Wilen CB, Lee S, Suthar MS, Baldridge MT. IFN-λ derived from nonsusceptible enterocytes acts on tuft cells to limit persistent norovirus. SCIENCE ADVANCES 2023; 9:eadi2562. [PMID: 37703370 PMCID: PMC10499323 DOI: 10.1126/sciadv.adi2562] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 08/10/2023] [Indexed: 09/15/2023]
Abstract
Norovirus is a leading cause of epidemic viral gastroenteritis, with no currently approved vaccines or antivirals. Murine norovirus (MNoV) is a well-characterized model of norovirus pathogenesis in vivo, and persistent strains exhibit lifelong intestinal infection. Interferon-λ (IFN-λ) is a potent antiviral that rapidly cures MNoV. We previously demonstrated that IFN-λ signaling in intestinal epithelial cells (IECs) controls persistent MNoV, and here demonstrate that IFN-λ acts on tuft cells, the exclusive site of MNoV persistence, to limit infection. While interrogating the source of IFN-λ to regulate MNoV, we confirmed that MDA5-MAVS signaling, required for IFN-λ induction to MNoV in vitro, controls persistent MNoV in vivo. We demonstrate that MAVS in IECs and not immune cells controls MNoV. MAVS in nonsusceptible enterocytes, but not in tuft cells, restricts MNoV, implicating noninfected cells as the IFN-λ source. Our findings indicate that host sensing of MNoV is distinct from cellular tropism, suggesting intercellular communication between IECs for antiviral signaling induction in uninfected bystander cells.
Collapse
Affiliation(s)
- Harshad Ingle
- Division of Infectious Diseases, Department of Medicine, Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Heyde Makimaa
- Division of Infectious Diseases, Department of Medicine, Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Somya Aggarwal
- Division of Infectious Diseases, Department of Medicine, Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Hongju Deng
- Division of Infectious Diseases, Department of Medicine, Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Lynne Foster
- Division of Infectious Diseases, Department of Medicine, Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Yuhao Li
- Division of Infectious Diseases, Department of Medicine, Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Elizabeth A. Kennedy
- Division of Infectious Diseases, Department of Medicine, Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Stefan T. Peterson
- Division of Infectious Diseases, Department of Medicine, Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Craig B. Wilen
- Departments of Laboratory Medicine and Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Sanghyun Lee
- Division of Biology and Medicine, Department of Molecular Microbiology and Immunology, Brown University, Providence, RI, USA
| | - Mehul S. Suthar
- Department of Pediatrics, Emory School of Medicine, Atlanta, GA, USA
| | - Megan T. Baldridge
- Division of Infectious Diseases, Department of Medicine, Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine, St. Louis, MO, USA
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
31
|
Feng Y, Pogan R, Thiede L, Müller-Guhl J, Uetrecht C, Roos WH. Fucose Binding Cancels out Mechanical Differences between Distinct Human Noroviruses. Viruses 2023; 15:1482. [PMID: 37515170 PMCID: PMC10383637 DOI: 10.3390/v15071482] [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: 04/17/2023] [Revised: 06/20/2023] [Accepted: 06/26/2023] [Indexed: 07/30/2023] Open
Abstract
The majority of nonbacterial gastroenteritis in humans and livestock is caused by noroviruses. Like most RNA viruses, frequent mutations result in various norovirus variants. The strain-dependent binding profiles of noroviruses to fucose are supposed to facilitate norovirus infection. It remains unclear, however, what the molecular mechanism behind strain-dependent functioning is. In this study, by applying atomic force microscopy (AFM) nanoindentation technology, we studied norovirus-like particles (noroVLPs) of three distinct human norovirus variants. We found differences in viral mechanical properties even between the norovirus variants from the same genogroup. The noroVLPs were then subjected to fucose treatment. Surprisingly, after fucose treatment, the previously found considerable differences in viral mechanical properties among these variants were diminished. We attribute a dynamic switch of the norovirus P domain upon fucose binding to the reduced differences in viral mechanical properties across the tested norovirus variants. These findings shed light on the mechanisms used by norovirus capsids to adapt to environmental changes and, possibly, increase cell infection. Hereby, a new step towards connecting viral mechanical properties to viral prevalence is taken.
Collapse
Affiliation(s)
- Yuzhen Feng
- Moleculaire Biofysica, Zernike Instituut, Rijksuniversiteit Groningen, 9747AG Groningen, The Netherlands
| | - Ronja Pogan
- CSSB Centre for Structural Systems Biology, Deutsches Elektronen-Synchrotron (DESY) & Leibniz Institute of Virology (LIV), 22607 Hamburg, Germany
- Faculty V: School of Life Sciences, University of Siegen, 57076 Siegen, Germany
| | - Lars Thiede
- CSSB Centre for Structural Systems Biology, Deutsches Elektronen-Synchrotron (DESY) & Leibniz Institute of Virology (LIV), 22607 Hamburg, Germany
- Faculty V: School of Life Sciences, University of Siegen, 57076 Siegen, Germany
| | - Jürgen Müller-Guhl
- CSSB Centre for Structural Systems Biology, Deutsches Elektronen-Synchrotron (DESY) & Leibniz Institute of Virology (LIV), 22607 Hamburg, Germany
- Partner Site Hamburg-Lübeck-Borstel-Riems, Bernhard Nocht Institute for Tropical Medicine and German Center for Infection Research (DZIF), 20359 Hamburg, Germany
| | - Charlotte Uetrecht
- CSSB Centre for Structural Systems Biology, Deutsches Elektronen-Synchrotron (DESY) & Leibniz Institute of Virology (LIV), 22607 Hamburg, Germany
- Faculty V: School of Life Sciences, University of Siegen, 57076 Siegen, Germany
| | - Wouter H Roos
- Moleculaire Biofysica, Zernike Instituut, Rijksuniversiteit Groningen, 9747AG Groningen, The Netherlands
| |
Collapse
|
32
|
Peiper AM, Helm EW, Nguyen Q, Phillips M, Williams CG, Shah D, Tatum S, Iyer N, Grodzki M, Eurell LB, Nasir A, Baldridge MT, Karst SM. Infection of neonatal mice with the murine norovirus strain WU23 is a robust model to study norovirus pathogenesis. Lab Anim (NY) 2023; 52:119-129. [PMID: 37142696 PMCID: PMC10234811 DOI: 10.1038/s41684-023-01166-5] [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] [Received: 08/26/2022] [Accepted: 04/03/2023] [Indexed: 05/06/2023]
Abstract
Noroviruses are the leading cause of severe childhood diarrhea and foodborne disease worldwide. While they are a major cause of disease in all age groups, infections in the very young can be quite severe, with annual estimates of 50,000-200,000 fatalities in children under 5 years old. In spite of the remarkable disease burden associated with norovirus infections, very little is known about the pathogenic mechanisms underlying norovirus diarrhea, principally because of the lack of tractable small animal models. The development of the murine norovirus (MNV) model nearly two decades ago has facilitated progress in understanding host-norovirus interactions and norovirus strain variability. However, MNV strains tested thus far either do not cause intestinal disease or were isolated from extraintestinal tissue, raising concerns about translatability of research findings to human norovirus disease. Consequently, the field lacks a strong model of norovirus gastroenteritis. Here we provide a comprehensive characterization of a new small animal model system for the norovirus field that overcomes prior weaknesses. Specifically, we demonstrate that the WU23 MNV strain isolated from a mouse naturally presenting with diarrhea causes a transient reduction in weight gain and acute self-resolving diarrhea in neonatal mice of several inbred mouse lines. Moreover, our findings reveal that norovirus-induced diarrhea is associated with infection of subepithelial cells in the small intestine and systemic spread. Finally, type I interferons (IFNs) are critical to protect hosts from norovirus-induced intestinal disease whereas type III IFNs exacerbate diarrhea. This latter finding is consistent with other emerging data implicating type III IFNs in the exacerbation of some viral diseases. This new model system should enable a detailed investigation of norovirus disease mechanisms.
Collapse
Affiliation(s)
- Amy M Peiper
- Department of Molecular Genetics & Microbiology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Emily W Helm
- Department of Molecular Genetics & Microbiology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Quyen Nguyen
- Department of Molecular Genetics & Microbiology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Matthew Phillips
- Department of Molecular Genetics & Microbiology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Caroline G Williams
- Department of Molecular Genetics & Microbiology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Dhairya Shah
- Department of Molecular Genetics & Microbiology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Sarah Tatum
- Department of Molecular Genetics & Microbiology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Neha Iyer
- Department of Molecular Genetics & Microbiology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Marco Grodzki
- Department of Molecular Genetics & Microbiology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Laura B Eurell
- Office of Research, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Aqsa Nasir
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Megan T Baldridge
- Division of Infectious Diseases, Department of Medicine, Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Stephanie M Karst
- Department of Molecular Genetics & Microbiology, College of Medicine, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
33
|
Mills JT, Minogue SC, Snowden JS, Arden WKC, Rowlands DJ, Stonehouse NJ, Wobus CE, Herod MR. Amino acid substitutions in norovirus VP1 dictate cell tropism via an attachment process dependent on membrane mobility. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.17.528071. [PMID: 36824911 PMCID: PMC9949111 DOI: 10.1101/2023.02.17.528071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/19/2023]
Abstract
Viruses interact with receptors on the cell surface to initiate and co-ordinate infection. The distribution of receptors on host cells can be a key determinant of viral tropism and host infection. Unravelling the complex nature of virus-receptor interactions is, therefore, of fundamental importance to understanding viral pathogenesis. Noroviruses are non-enveloped, icosahedral, positive-sense RNA viruses of global importance to human health, with no approved vaccine or antiviral agent available. Here we use murine norovirus as a model for the study of molecular mechanisms of virus-receptor interactions. We show that variation at a single amino acid residue in the major viral capsid protein had a key impact on the interaction between virus and receptor. This variation did not affect virion production or virus growth kinetics, but a specific amino acid was rapidly selected through evolution experiments, and significantly improved cellular attachment when infecting immune cells in suspension. However, reducing plasma membrane mobility counteracted this phenotype, providing insight into for the role of membrane fluidity and receptor recruitment in norovirus cellular attachment. When the infectivity of a panel of recombinant viruses with single amino acid variations was compared in vivo, there were significant differences in the distribution of viruses in a murine model, demonstrating a role in cellular tropism in vivo. Overall, these results highlight the importance of lipid rafts and virus-induced receptor recruitment in viral infection, as well as how capsid evolution can greatly influence cellular tropism, within-host spread and pathogenicity.
Collapse
Affiliation(s)
- Jake T Mills
- Astbury Centre for Structural Molecular Biology, School of Molecular & Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Susanna C Minogue
- Astbury Centre for Structural Molecular Biology, School of Molecular & Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Joseph S Snowden
- Astbury Centre for Structural Molecular Biology, School of Molecular & Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Wynter K C Arden
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48130, USA
| | - David J Rowlands
- Astbury Centre for Structural Molecular Biology, School of Molecular & Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Nicola J Stonehouse
- Astbury Centre for Structural Molecular Biology, School of Molecular & Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Christiane E Wobus
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48130, USA
| | - Morgan R Herod
- Astbury Centre for Structural Molecular Biology, School of Molecular & Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| |
Collapse
|
34
|
Tohma K, Ushijima H. [Molecular epidemiology and evolution of human noroviruses]. Uirusu 2023; 73:17-32. [PMID: 39343517 DOI: 10.2222/jsv.73.17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Noroviruses are the most common viral cause of acute gastroenteritis after the introduction of rotavirus vaccines. Norovirus infection can cause severe symptoms in vulnerable populations including young children and the elderly. Thus, it is still a leading cause of death from diarrhea in children in developing countries. Recent advancement of genomics platforms facilitated understanding of the epidemiology of norovirus, while the whole picture of norovirus diversity is still undetermined. Currently, there are no approved vaccines for norovirus, but state-of-the-art norovirus cultivation systems could elucidate the antigenic diversity of this fast-evolving virus. In this review, we will summarize the historical and latest findings of norovirus epidemiology, diversity, and evolution.
Collapse
Affiliation(s)
- Kentaro Tohma
- Division of Viral Products, US Food and Drug Administration, Maryland, Unites States
| | - Hiroshi Ushijima
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo, Japan
| |
Collapse
|
35
|
Gebert JT, Scribano F, Engevik KA, Perry JL, Hyser JM. Gastrointestinal organoids in the study of viral infections. Am J Physiol Gastrointest Liver Physiol 2023; 324:G51-G59. [PMID: 36414538 PMCID: PMC9799139 DOI: 10.1152/ajpgi.00152.2022] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 11/04/2022] [Accepted: 11/14/2022] [Indexed: 11/24/2022]
Abstract
Viruses are among the most prevalent enteric pathogens. Although virologists historically relied on cell lines and animal models, human intestinal organoids (HIOs) continue to grow in popularity. HIOs are nontransformed, stem cell-derived, ex vivo cell cultures that maintain the cell type diversity of the intestinal epithelium. They offer higher throughput than standard animal models while more accurately mimicking the native tissue of infection than transformed cell lines. Here, we review recent literature that highlights virological advances facilitated by HIOs. We discuss the variations and limitations of HIOs, how HIOs have allowed for the cultivation of previously uncultivatable viruses, and how they have offered insight into tropism, entry, replication kinetics, and host-pathogen interactions. In each case, we discuss exemplary viruses and archetypal studies. We discuss how the speed and flexibility of HIO-based studies contributed to our knowledge of SARS-CoV-2 and antiviral therapeutics. Finally, we discuss the current limitations of HIOs and future directions to overcome these.
Collapse
Affiliation(s)
- J Thomas Gebert
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas
| | - Francesca Scribano
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas
| | - Kristen A Engevik
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas
| | - Jacob L Perry
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas
| | - Joseph M Hyser
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas
- Center for Metagenomics and Microbiome Research, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
36
|
Kim DH, Jeong M, Kim JH, Son JE, Lee JJY, Park SJ, Lee J, Kim M, Oh JW, Park MS, Byun S. Lactobacillus salivarius HHuMin-U Activates Innate Immune Defense against Norovirus Infection through TBK1-IRF3 and NF-κB Signaling Pathways. RESEARCH (WASHINGTON, D.C.) 2022; 2022:0007. [PMID: 39290965 PMCID: PMC11407524 DOI: 10.34133/research.0007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Accepted: 10/21/2022] [Indexed: 09/19/2024]
Abstract
The composition of commensal bacteria plays a critical role in controlling immune responses in the intestine. Studies have shown that specific bacterial strains may have the capacity to enhance host immune defense against gastrointestinal viral infections. While norovirus is known to be the most common cause of gastroenteritis, leading to an estimated 200,000 deaths every year, identification of bacterial strains with protective effects against norovirus infection remains elusive. Here, we discovered Lactobacillus salivarius HHuMin-U (HHuMin-U) as a potent antiviral strain against norovirus infection. HHuMin-U significantly suppressed murine norovirus replication and lowered viral RNA titers in macrophages. The transcriptome sequencing (RNA sequencing) analysis revealed that HHuMin-U markedly enhanced the expression level of antiviral interferon-stimulated genes compared to mock treatment. HHuMin-U treatment dose-dependently induced type I interferons (IFN-α and IFN-β) and tumor necrosis factor-α production in mouse and human macrophages, promoting antiviral innate responses against norovirus infection. Investigation on the molecular mechanism demonstrated that HHuMin-U can activate nuclear factor κB and TANK-binding kinase 1 (TBK1)-interferon regulatory factor 3 signaling pathways, leading to the phosphorylation of signal transducer and activator of transcription 1 and signal transducer and activator of transcription 2, the key mediators of interferon-stimulated genes. Finally, oral administration of HHuMin-U increased IFN-β levels in the ileum of mice and altered the gut microbiome profile. These results suggest the species/strain-specific importance of gut microbial composition for antiviral immune responses and the potential use of HHuMin-U as a probiotic agent.
Collapse
Affiliation(s)
- Da Hyun Kim
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Minju Jeong
- Department of Agricultural Biotechnology, Seoul National University, Seoul 08826, Republic of Korea
| | - Jae Hwan Kim
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Joe Eun Son
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - John J Y Lee
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Sang-Jun Park
- Research Center, BIFIDO Co. Ltd., Hanam 12930, Republic of Korea
| | - Juyeon Lee
- Research Center, BIFIDO Co. Ltd., Hanam 12930, Republic of Korea
| | - Minwoo Kim
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Jong-Won Oh
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Myeong Soo Park
- Research Center, BIFIDO Co. Ltd., Hanam 12930, Republic of Korea
| | - Sanguine Byun
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| |
Collapse
|
37
|
Strine MS, Alfajaro MM, Graziano VR, Song J, Hsieh LL, Hill R, Guo J, VanDussen KL, Orchard RC, Baldridge MT, Lee S, Wilen CB. Tuft-cell-intrinsic and -extrinsic mediators of norovirus tropism regulate viral immunity. Cell Rep 2022; 41:111593. [PMID: 36351394 PMCID: PMC9662704 DOI: 10.1016/j.celrep.2022.111593] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 08/19/2022] [Accepted: 10/12/2022] [Indexed: 11/09/2022] Open
Abstract
Murine norovirus (MNoV) is a model for human norovirus and for interrogating mechanisms of viral tropism and persistence. We previously demonstrated that the persistent strain MNoVCR6 infects tuft cells, which are dispensable for the non-persistent strain MNoVCW3. We now show that diverse MNoV strains require tuft cells for chronic enteric infection. We also demonstrate that interferon-λ (IFN-λ) acts directly on tuft cells to cure chronic MNoVCR6 infection and that type I and III IFNs signal together via STAT1 in tuft cells to restrict MNoVCW3 tropism. We then develop an enteroid model and find that MNoVCR6 and MNoVCW3 similarly infect tuft cells with equal IFN susceptibility, suggesting that IFN derived from non-epithelial cells signals on tuft cells in trans to restrict MNoVCW3 tropism. Thus, tuft cell tropism enables MNoV persistence and is determined by tuft cell-intrinsic factors (viral receptor expression) and -extrinsic factors (immunomodulatory signaling by non-epithelial cells).
Collapse
Affiliation(s)
- Madison S Strine
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA; Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Mia Madel Alfajaro
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA; Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Vincent R Graziano
- Department of Immunology, University of Connecticut Health Center, Farmington, CT, USA
| | - Jaewon Song
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI, USA
| | - Leon L Hsieh
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Ryan Hill
- Division of Infectious Diseases, Department of Medicine, Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Jun Guo
- Department of Surgery, Washington University School of Medicine, Saint Louis, MO, USA
| | - Kelli L VanDussen
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center and University of Cincinnati, Cincinnati, OH, USA
| | - Robert C Orchard
- Department of Immunology, University of Texas Southwestern Medical School, Dallas, TX, USA
| | - Megan T Baldridge
- Department of Medicine, Division of Infectious Diseases, Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Sanghyun Lee
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI, USA.
| | - Craig B Wilen
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA; Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
38
|
Human Norovirus Efficiently Replicates in Differentiated 3D-Human Intestinal Enteroids. J Virol 2022; 96:e0085522. [PMID: 36342297 PMCID: PMC9683019 DOI: 10.1128/jvi.00855-22] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
The human norovirus (HNoV) clinical and socio-economic impact calls for immediate action in the development of anti-infectives. Physiologically relevant
in vitro
models are hence needed to study HNoV biology, tropism, and mechanisms of viral-associated disease, and also as a platform to identify antiviral agents.
Collapse
|
39
|
Helm EW, Peiper AM, Phillips M, Williams CG, Sherman MB, Kelley T, Smith HQ, Jacobs SO, Shah D, Tatum SM, Iyer N, Grodzki M, Morales Aparicio JC, Kennedy EA, Manzi MS, Baldridge MT, Smith TJ, Karst SM. Environmentally-triggered contraction of the norovirus virion determines diarrheagenic potential. Front Immunol 2022; 13:1043746. [PMID: 36389732 PMCID: PMC9664153 DOI: 10.3389/fimmu.2022.1043746] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 10/18/2022] [Indexed: 11/07/2022] Open
Abstract
Noroviruses are the leading cause of severe childhood diarrhea and foodborne disease worldwide. While they are a major cause of disease in all age groups, infections in the very young can be quite severe with annual estimates of 50,000-200,000 fatalities in children under 5 years old. In spite of the remarkable disease burden associated with norovirus infections in people, very little is known about the pathogenic mechanisms underlying norovirus diarrhea, principally because of the lack of tractable small animal models. We recently demonstrated that wild-type neonatal mice are susceptible to murine norovirus (MNV)-induced acute self-resolving diarrhea in a time course mirroring human norovirus disease. Using this robust pathogenesis model system, we demonstrate that virulence is regulated by the responsiveness of the viral capsid to environmental cues that trigger contraction of the VP1 protruding (P) domain onto the particle shell, thus enhancing receptor binding and infectivity. The capacity of a given MNV strain to undergo this contraction positively correlates with infection of cells expressing low abundance of the virus receptor CD300lf, supporting a model whereby virion contraction triggers infection of CD300lflo cell types that are responsible for diarrhea induction. These findings directly link environmentally-influenced biophysical features with norovirus disease severity.
Collapse
Affiliation(s)
- Emily W. Helm
- Department of Molecular Genetics & Microbiology, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Amy M. Peiper
- Department of Molecular Genetics & Microbiology, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Matthew Phillips
- Department of Molecular Genetics & Microbiology, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Caroline G. Williams
- Department of Molecular Genetics & Microbiology, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Michael B. Sherman
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | - Theresa Kelley
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | - Hong Q. Smith
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | - Sorin O. Jacobs
- Department of Molecular Genetics & Microbiology, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Dhairya Shah
- Department of Molecular Genetics & Microbiology, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Sarah M. Tatum
- Department of Molecular Genetics & Microbiology, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Neha Iyer
- Department of Molecular Genetics & Microbiology, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Marco Grodzki
- Department of Molecular Genetics & Microbiology, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Joyce C. Morales Aparicio
- Department of Molecular Genetics & Microbiology, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Elizabeth A. Kennedy
- Division of Infectious Diseases, Department of Medicine, Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO, United States
| | - Mikayla S. Manzi
- Department of Molecular Genetics & Microbiology, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Megan T. Baldridge
- Division of Infectious Diseases, Department of Medicine, Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO, United States
| | - Thomas J. Smith
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | - Stephanie M. Karst
- Department of Molecular Genetics & Microbiology, College of Medicine, University of Florida, Gainesville, FL, United States
| |
Collapse
|
40
|
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
|
41
|
Abstract
The thick mucus layer covering of the intestinal epithelium has received increasing attention, owing to its protective role in intestinal infection. However, the exact mechanisms by which the mucus increases intestinal resistance against viral infection remain largely unclear. Here, we identify prominent antiviral activity of the small intestinal mucus and extracted total mucus proteins, as evidenced by their inhibitory effects against porcine epidemic diarrhea virus (PEDV) infection. Of all the extracted mucus proteins, mucin 2 and fraction III (~70 kDa) exhibited potent antiviral activity. We further evaluated the antiviral effects of three candidate factors in fraction III and found that calpain-1 contributed substantially to its antiviral activity. In vivo studies demonstrated that oral administration of calpain-1 provided effective protection against intestinal PEDV infection. As a calcium-activated cysteine protease, calpain-1 inhibited viral invasion by binding to and hydrolyzing the S1 domain of the viral spike protein. The region between amino acids 297 and 337 in the b domain of PEDV S1 protein was critical for calpain-1-mediated hydrolysis. Further investigation indicated that calpain-1 could be produced by goblet cells between intestinal epithelia. Taken together, the results of our study revealed calpain-1 to be a novel antiviral protein in porcine small intestinal mucus, suggesting that calpain-1 has potential for defending against intestinal infections.
Collapse
|
42
|
Lee JE, Kim SA, Park HJ, Mun H, Ha KS, Shim WB. Colorimetric detection of norovirus by helicase-dependent amplification method based on specific primers integrated with HRPzyme. Anal Bioanal Chem 2022; 414:6723-6733. [PMID: 35931785 DOI: 10.1007/s00216-022-04247-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 07/11/2022] [Accepted: 07/21/2022] [Indexed: 11/28/2022]
Abstract
Noroviruses (NoVs) are the most common causes of epidemic gastroenteritis, responsible for at least 50% of all gastroenteritis outbreaks worldwide and significant causes of foodborne illness. In the USA, approximately 21 million illnesses attributable to NoVs have annually occurred. Therefore, there is a great demand to develop a rapid, low-cost, and accurate detection method for NoVs. This study first reported colorimetric helicase-dependent amplification (HDA) methods based on specific primers integrated with HRPzyme for the rapid and sensitive detection of NoV GI and GII. The colorimetric HDA methods exhibited a detection limit of 10 copies mL-1 of each NoV GI and GII and were confirmed to be specific to each NoV GI and GII. The period required to complete the HDA method was 2 h, including a step of RNA extraction and cDNA synthesis without expensive instruments such as a thermal cycler and detector. The cutoff value of the method for the oyster artificially inoculated with a known amount of NoV was all 102 copies g-1 for NoV GI and GII. Therefore, the HDA method developed in this study can be useful tool for the on-site detection of NoVs in food samples.
Collapse
Affiliation(s)
- Jeong-Eun Lee
- Institute of Smart Farm, Gyeongsang National University, Jinju, 52828, Gyeongnam, Korea
| | - Sol-A Kim
- Division of Applied Life Science, Graduate School, Gyeongsang National University, Jinju, 52828, Gyeongnam, Korea
| | - Hyun-Jin Park
- Division of Applied Life Science, Graduate School, Gyeongsang National University, Jinju, 52828, Gyeongnam, Korea
| | - Hyoyoung Mun
- Apteasy MJ Inc., 333 Cheomdangwagi-ro, Technopark, Gwangju, 61008, Korea
| | - Kwang-Soo Ha
- Southeast Sea Fisheries Research Institute, National Institute of Fisheries Science, Tongyeong, 53085, Korea
| | - Won-Bo Shim
- Institute of Smart Farm, Gyeongsang National University, Jinju, 52828, Gyeongnam, Korea. .,Institute of Agriculture and Life Science, Gyeongsang National University, Jinju, 52828, Gyeongnam, Korea. .,Department of Food Science and Technology, Gyeongsang National University, Jinju, 52828, Gyeongnam, Korea.
| |
Collapse
|
43
|
Moreno-Valladares M, Moncho-Amor V, Bernal-Simon I, Agirre-Iturrioz E, Álvarez-Satta M, Matheu A. Norovirus Intestinal Infection and Lewy Body Disease in an Older Patient with Acute Cognitive Impairment. Int J Mol Sci 2022; 23:ijms23158376. [PMID: 35955510 PMCID: PMC9368907 DOI: 10.3390/ijms23158376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/15/2022] [Accepted: 07/26/2022] [Indexed: 11/16/2022] Open
Abstract
We present a case report on an older woman with unspecific symptoms and predominant long-term gastrointestinal disturbances, acute overall health deterioration with loss of autonomy for daily activities, and cognitive impairment. Autopsy revealed the presence of alpha-synuclein deposits spread into intestinal mucosa lesions, enteric plexuses, pelvic and retroperitoneal nerves and ganglia, and other organs as well as Lewy pathology in the central nervous system (CNS). Moreover, we isolated norovirus from the patient, indicating active infection in the colon and detected colocalization of norovirus and alpha-synuclein in different regions of the patient’s brain. In view of this, we report a concomitant norovirus infection with synthesis of alpha-synuclein in the gastrointestinal mucosa and Lewy pathology in the CNS, which might support Braak’s hypothesis about the pathogenic mechanisms underlying synucleinopathies.
Collapse
Affiliation(s)
- Manuel Moreno-Valladares
- Pathology Department, Donostia University Hospital, Osakidetza Basque Health Service, 20014 San Sebastian, Spain; (I.B.-S.); (E.A.-I.)
- Group of Cellular Oncology, Biodonostia Health Research Institute, 20014 San Sebastian, Spain; (V.M.-A.); (M.Á.-S.); (A.M.)
- CIBER on Frailty and Healthy Aging (CIBERfes), Institute of Health Carlos III, 28029 Madrid, Spain
- Correspondence: ; Tel.: +34-943007151
| | - Veronica Moncho-Amor
- Group of Cellular Oncology, Biodonostia Health Research Institute, 20014 San Sebastian, Spain; (V.M.-A.); (M.Á.-S.); (A.M.)
| | - Iraide Bernal-Simon
- Pathology Department, Donostia University Hospital, Osakidetza Basque Health Service, 20014 San Sebastian, Spain; (I.B.-S.); (E.A.-I.)
| | - Eñaut Agirre-Iturrioz
- Pathology Department, Donostia University Hospital, Osakidetza Basque Health Service, 20014 San Sebastian, Spain; (I.B.-S.); (E.A.-I.)
| | - María Álvarez-Satta
- Group of Cellular Oncology, Biodonostia Health Research Institute, 20014 San Sebastian, Spain; (V.M.-A.); (M.Á.-S.); (A.M.)
- CIBER on Frailty and Healthy Aging (CIBERfes), Institute of Health Carlos III, 28029 Madrid, Spain
| | - Ander Matheu
- Group of Cellular Oncology, Biodonostia Health Research Institute, 20014 San Sebastian, Spain; (V.M.-A.); (M.Á.-S.); (A.M.)
- CIBER on Frailty and Healthy Aging (CIBERfes), Institute of Health Carlos III, 28029 Madrid, Spain
- IKERBASQUE, Basque Foundation for Science, 48011 Bilbao, Spain
| |
Collapse
|
44
|
Nolan LS, Baldridge MT. Advances in understanding interferon-mediated immune responses to enteric viruses in intestinal organoids. Front Immunol 2022; 13:943334. [PMID: 35935957 PMCID: PMC9354881 DOI: 10.3389/fimmu.2022.943334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 06/30/2022] [Indexed: 11/16/2022] Open
Abstract
Interferons (IFN) are antiviral cytokines with critical roles in regulating pathogens at epithelial barriers, but their capacity to restrict human enteric viruses has been incompletely characterized in part due to challenges in cultivating some viruses in vitro, particularly human norovirus. Accordingly, advancements in the development of antiviral therapies and vaccine strategies for enteric viral infections have been similarly constrained. Currently emerging is the use of human intestinal enteroids (HIEs) to investigate mechanisms of human enteric viral pathogenesis. HIEs provide a unique opportunity to investigate host-virus interactions using an in vitro system that recapitulates the cellular complexity of the in vivo gastrointestinal epithelium. This approach permits the exploration of intestinal epithelial cell interactions with enteric viruses as well as the innate immune responses mediated by IFNs and IFN-stimulated genes. Here, we describe recent findings related to the production, signaling, and function of IFNs in the response to enteric viral infections, which will ultimately help to reveal important aspects of pathogenesis and facilitate the future development of therapeutics and vaccines.
Collapse
Affiliation(s)
- Lila S. Nolan
- Department of Pediatrics, Division of Newborn Medicine, Washington University School of Medicine, St. Louis Children’s Hospital, St. Louis, MO, United States
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO, United States
| | - Megan T. Baldridge
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO, United States
- Department of Medicine, Division of Infectious Diseases, Washington University School of Medicine, St. Louis, MO, United States
| |
Collapse
|
45
|
Tsuruta S, Kawasaki T, Machida M, Iwatsuki K, Inaba A, Shibata S, Shindo T, Nakabayashi K, Hakamada K, Umezawa A, Akutsu H. Development of Human Gut Organoids With Resident Tissue Macrophages as a Model of Intestinal Immune Responses. Cell Mol Gastroenterol Hepatol 2022; 14:726-729.e5. [PMID: 35760286 PMCID: PMC9421619 DOI: 10.1016/j.jcmgh.2022.06.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 06/15/2022] [Accepted: 06/16/2022] [Indexed: 12/10/2022]
Affiliation(s)
- Satoru Tsuruta
- Center for Regenerative Medicine, National Center for Child Health and Development, Tokyo, Japan; Department of Gastroenterological Surgery, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Tomoyuki Kawasaki
- Center for Regenerative Medicine, National Center for Child Health and Development, Tokyo, Japan
| | - Masakazu Machida
- Center for Regenerative Medicine, National Center for Child Health and Development, Tokyo, Japan
| | - Ken Iwatsuki
- Department of Nutritional Science and Food Safety, Faculty of Applied Bioscience, Tokyo University of Agriculture, Tokyo, Japan
| | - Akihiko Inaba
- Department of Nutritional Science and Food Safety, Faculty of Applied Bioscience, Tokyo University of Agriculture, Tokyo, Japan
| | - Shinsuke Shibata
- Electron Microscope Laboratory, Keio University School of Medicine, Tokyo, Japan; Division of Microscopic Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Tomoko Shindo
- Electron Microscope Laboratory, Keio University School of Medicine, Tokyo, Japan
| | - Kazuhiko Nakabayashi
- Department of Maternal-Fetal Biology, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Kenichi Hakamada
- Department of Gastroenterological Surgery, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Akihiro Umezawa
- Center for Regenerative Medicine, National Center for Child Health and Development, Tokyo, Japan
| | - Hidenori Akutsu
- Center for Regenerative Medicine, National Center for Child Health and Development, Tokyo, Japan
| |
Collapse
|
46
|
A highly quantitative detection system for cell entry of human norovirus-like particles based on the complementation of NanoLuc luciferase. Virology 2022; 573:23-28. [DOI: 10.1016/j.virol.2022.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 05/10/2022] [Accepted: 06/01/2022] [Indexed: 11/21/2022]
|
47
|
Estienney M, Tarris G, Abou-Hamad N, Rouleau A, Boireau W, Chassagnon R, Ayouni S, Daval-Frerot P, Martin L, Bouyer F, Le Pendu J, de Rougemont A, Belliot G. Epidemiological Impact of GII.17 Human Noroviruses Associated With Attachment to Enterocytes. Front Microbiol 2022; 13:858245. [PMID: 35572680 PMCID: PMC9094630 DOI: 10.3389/fmicb.2022.858245] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 03/23/2022] [Indexed: 01/19/2023] Open
Abstract
For the last 30 years, molecular surveys have shown that human norovirus (HuNoV), predominantly the GII.4 genotype, is one of the main causative agents of gastroenteritis. However, epidemiological surveys have revealed the worldwide emergence of GII.17 HuNoVs. Genetic analysis confirmed that GII.17 strains are distributed into three variants (i.e., Kawasaki 308, Kawasaki 323, and CS-E1). Here, virus-like particles (VLPs) were baculovirus-expressed from these variants to study putative interactions with HBGA. Qualitative analysis of the HBGA binding profile of each variant showed that the most recent and predominant GII.17 variant, Kawasaki 308, possesses a larger binding spectrum. The retrospective study of GII.17 strains documented before the emergence of the dominant Kawasaki 308 variant showed that the emergence of a new GII.17 variant could be related to an increased binding capacity toward HBGA. The use of duodenal histological sections confirmed that recognition of enterocytes involved HBGA for the three GII.17 variants. Finally, we observed that the relative affinity of recent GII.17 VLPs for HBGA remains lower than that of the GII.4-2012 variant. These observations suggest a model whereby a combination of virological factors, such as polymerase fidelity and increased affinity for HBGA, and immunological factors was responsible for the incomplete and non-persistent replacement of GII.4 by new GII.17 variants.
Collapse
Affiliation(s)
- Marie Estienney
- National Reference Centre for Gastroenteritis Viruses, Laboratory of Virology, University Hospital of Dijon, Dijon, France.,UMR PAM A 02.102 Procédés Alimentaires et Microbiologiques, Université de Bourgogne, Franche-Comté/AgroSup Dijon, Dijon, France
| | - Georges Tarris
- Department of Pathology, University Hospital of Dijon, Dijon, France.,Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Dijon, France
| | - Nicole Abou-Hamad
- National Reference Centre for Gastroenteritis Viruses, Laboratory of Virology, University Hospital of Dijon, Dijon, France.,UMR PAM A 02.102 Procédés Alimentaires et Microbiologiques, Université de Bourgogne, Franche-Comté/AgroSup Dijon, Dijon, France.,Laboratoire Interdisciplinaire Carnot de Bourgogne, UMR CNRS 6303, Université Bourgogne Franche-Comté, Dijon, France
| | - Alain Rouleau
- FEMTO-ST Institute, CNRS UMR-6174, Université de Bourgogne Franche-Comté, Besançon, France
| | - Wilfrid Boireau
- FEMTO-ST Institute, CNRS UMR-6174, Université de Bourgogne Franche-Comté, Besançon, France
| | - Rémi Chassagnon
- Laboratoire Interdisciplinaire Carnot de Bourgogne, UMR CNRS 6303, Université Bourgogne Franche-Comté, Dijon, France
| | - Siwar Ayouni
- National Reference Centre for Gastroenteritis Viruses, Laboratory of Virology, University Hospital of Dijon, Dijon, France
| | - Philippe Daval-Frerot
- National Reference Centre for Gastroenteritis Viruses, Laboratory of Virology, University Hospital of Dijon, Dijon, France
| | - Laurent Martin
- Department of Pathology, University Hospital of Dijon, Dijon, France.,Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Dijon, France
| | - Frédéric Bouyer
- Laboratoire Interdisciplinaire Carnot de Bourgogne, UMR CNRS 6303, Université Bourgogne Franche-Comté, Dijon, France
| | | | - Alexis de Rougemont
- National Reference Centre for Gastroenteritis Viruses, Laboratory of Virology, University Hospital of Dijon, Dijon, France.,UMR PAM A 02.102 Procédés Alimentaires et Microbiologiques, Université de Bourgogne, Franche-Comté/AgroSup Dijon, Dijon, France
| | - Gael Belliot
- National Reference Centre for Gastroenteritis Viruses, Laboratory of Virology, University Hospital of Dijon, Dijon, France.,UMR PAM A 02.102 Procédés Alimentaires et Microbiologiques, Université de Bourgogne, Franche-Comté/AgroSup Dijon, Dijon, France
| |
Collapse
|
48
|
Lee S. Norovirus cell tropism: The road to uncovering its secret hideout. Cell Host Microbe 2022; 30:454-457. [PMID: 35421342 DOI: 10.1016/j.chom.2022.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Here, I revisit our early Cell Host & Microbe publications, which show how norovirus builds its comfortable home in an extremely rare intestinal cell population for persistent infection. This commentary covers insights from previous works and advances in the current research.
Collapse
Affiliation(s)
- Sanghyun Lee
- Department of Molecular Microbiology and Immunology, Division of Biology and Medicine, Brown University, Providence, RI, USA.
| |
Collapse
|
49
|
Surwase SS, Shahriar SMS, An JM, Ha J, Mirzaaghasi A, Bagheri B, Park JH, Lee YK, Kim YC. Engineered Nanoparticles inside a Microparticle Oral System for Enhanced Mucosal and Systemic Immunity. ACS APPLIED MATERIALS & INTERFACES 2022; 14:11124-11143. [PMID: 35227057 DOI: 10.1021/acsami.1c24982] [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] [Indexed: 06/14/2023]
Abstract
Antigen delivery through an oral route requires overcoming multiple challenges, including gastrointestinal enzymes, mucus, and epithelial tight junctions. Although each barrier has a crucial role in determining the final efficiency of the oral vaccination, transcytosis of antigens through follicle-associated epithelium (FAE) represents a major challenge. Most of the research is focused on delivering an antigen to the M-cell for FAE transcytosis because M-cells can easily transport the antigen from the luminal site. However, the fact is that the M-cell population is less than 1% of the total gastrointestinal cells, and most of the oral vaccines have failed to show any effect in clinical trials. To challenge the current dogma of M-cell targeting, in this study, we designed a novel tandem peptide with a FAE-targeting peptide at the front position and a cell-penetrating peptide at the back position. The tandem peptide was attached to a smart delivery system, which overcomes the enzymatic barrier and the mucosal barrier. The result showed that the engineered system could target the FAE (enterocytes and M-cells) and successfully penetrate the enterocytes to reach the dendritic cells located at the subepithelium dome. There was successful maturation and activation of dendritic cells in vitro confirmed by a significant increase in maturation markers such as CD40, CD86, presentation marker MHC I, and proinflammatory cytokines (TNF-α, IL-6, and IL-10). The in vivo results showed a high production of CD4+ T-lymphocytes (helper T-cell) and a significantly higher production of CD8+ T-lymphocytes (killer T-cell). Finally, the production of mucosal immunity (IgA) in the trachea, intestine, and fecal extracts and systemic immunity (IgG, IgG1, and IgG2a) was successfully confirmed. To the best of our knowledge, this is the first study that designed a novel tandem peptide to target the FAE, which includes M-cells and enterocytes rather than M-cell targeting and showed that a significant induction of both the mucosal and systemic immune response was achieved compared to M-cell targeting.
Collapse
Affiliation(s)
- Sachin S Surwase
- Department of Chemical & Biomolecular Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - S M Shatil Shahriar
- Department of Surgery-Transplant and Mary & Dick Holland Regenerative Medicine Program, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska 68198-5940, United States
- KB Biomed Inc., Chungju 27469, Republic of Korea
- Department of Chemical & Biological Engineering, Korea National University of Transportation, Chungju 27469, Republic of Korea
| | - Jeong Man An
- Department of Chemical & Biological Engineering, Korea National University of Transportation, Chungju 27469, Republic of Korea
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul 04763, Republic of Korea
| | - JongHoon Ha
- Department of Chemical & Biomolecular Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Amin Mirzaaghasi
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Babak Bagheri
- Department of Chemical & Biomolecular Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Ji-Ho Park
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Yong-Kyu Lee
- KB Biomed Inc., Chungju 27469, Republic of Korea
- Department of Chemical & Biological Engineering, Korea National University of Transportation, Chungju 27469, Republic of Korea
| | - Yeu-Chun Kim
- Department of Chemical & Biomolecular Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| |
Collapse
|
50
|
Abstract
Although tuft cells were discovered over 60 years ago, their functions have long been enigmatic, especially in human health. Nonetheless, tuft cells have recently emerged as key orchestrators of the host response to diverse microbial infections in the gut and airway. While tuft cells are epithelial in origin, they exhibit functions akin to immune cells and mediate important interkingdom interactions between the host and helminths, protists, viruses, and bacteria. With broad intra- and intertissue heterogeneity, tuft cells sense and respond to microbes with exquisite specificity. Tuft cells can recognize helminth and protist infection, driving a type 2 immune response to promote parasite expulsion. Tuft cells also serve as the primary physiologic target of persistent murine norovirus (MNV) and promote immune evasion. Recently, tuft cells were also shown to be infected by rotavirus. Other viral infections, such as influenza A virus, can induce tuft cell–dependent tissue repair. In the context of coinfection, tuft cells promote neurotropic flavivirus replication by dampening antiviral adaptive immune responses. Commensal and pathogenic bacteria can regulate tuft cell abundance and function and, in turn, tuft cells are implicated in modulating bacterial infiltration and mucosal barrier integrity. However, the contribution of tuft cells to microbial sensing in humans and their resulting effector responses are poorly characterized. Herein, we aim to provide a comprehensive overview of microbial activation of tuft cells with an emphasis on tuft cell heterogeneity and differences between mouse and human tuft cell biology as it pertains to human health and disease.
Collapse
Affiliation(s)
- Madison S. Strine
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, Connecticut, United States of America
- Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut, United States of America
| | - Craig B. Wilen
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, Connecticut, United States of America
- Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut, United States of America
- Yale Cancer Center, Yale School of Medicine, New Haven, Connecticut, United States of America
- * E-mail: ,
| |
Collapse
|