1
|
Srinivas MA, Pierce LR, Olson MC, Roberston SJ, Sturdevant GL, Best SM, Orchard RC. Trim7 does not have a role in the restriction of murine norovirus infection in vivo. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.17.618898. [PMID: 39464121 PMCID: PMC11507913 DOI: 10.1101/2024.10.17.618898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Trim7 is an E3 ubiquitin ligase that was recently identified as a central regulator of host-viral interactions with both pro-viral and anti-viral activity in cell culture. As an inhibitor, Trim7 overexpression ubiquitinates viral proteins by recognizing C-terminal glutamines that are hallmarks of 3C-like protease cleavage events. Here we sought to determine the physiological impact of Trim7 in resolving murine norovirus (MNV) infection of mice as MNV is potently inhibited by Trim7 in vitro. Utilizing two independently derived Trim7 deficient mouse lines we found no changes in the viral burden or tissue distribution of MNV in both an acute and persistent model of infection. Additionally, no changes in cytokine responses were observed after acute MNV infection of Trim7-deficient mice. Furthermore, removal of potentially confounding innate immune responses such as STING and STAT1 did not reveal any role for Trim7 in regulating MNV replication. Taken together, our data fails to find a physiological role for Trim7 in regulating MNV infection outcomes in mice and serves as a caution for defining Trim7 as a broad acting antiviral.
Collapse
Affiliation(s)
| | - Linley R. Pierce
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Mikayla C. Olson
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Shelly J. Roberston
- Laboratory of Neurological Infections and Immunity, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton MT, USA
| | - Gail L. Sturdevant
- Laboratory of Neurological Infections and Immunity, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton MT, USA
| | - Sonja M. Best
- Laboratory of Neurological Infections and Immunity, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton MT, USA
| | - Robert C. Orchard
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
2
|
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
|
3
|
Janicka P, Stygar D, Chełmecka E, Kuropka P, Miążek A, Studzińska A, Pogorzelska A, Pala K, Bażanów B. Oxidative Stress Markers and Histopathological Changes in Selected Organs of Mice Infected with Murine Norovirus 1 (MNV-1). Int J Mol Sci 2024; 25:3614. [PMID: 38612426 PMCID: PMC11011583 DOI: 10.3390/ijms25073614] [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: 02/25/2024] [Revised: 03/19/2024] [Accepted: 03/21/2024] [Indexed: 04/14/2024] Open
Abstract
This paper describes the effects of murine norovirus (MNV) infection on oxidative stress and histopathological changes in mice. This study uses histopathological assays, enzymatic and non-enzymatic antioxidant markers, and total oxidative status and capacity (TOS, TAC). The results suggest that MNV infection can lead to significant changes with respect to the above-mentioned parameters in various organs. Specifically, reduced superoxide dismutase (SOD), Mn superoxide dismutase (MnSOD), catalase (CAT), and glutathione reductase (GR) activities were observed in liver tissues, while higher MnSOD activity was observed in kidney tissues of MNV-infected mice when compared to the control. GR activity was lower in all tissues of MNV-infected mice tested, with the exception of lung tissue. This study also showed that norovirus infection led to increased TOS levels in the brain and liver and TAC levels in the brain, while TOS levels were significantly reduced in the kidneys. These changes may be due to the production of reactive oxygen species (ROS) caused by the viral infection. ROS can damage cells and contribute to oxidative stress. These studies help us to understand the pathogenesis of MNV infection and its potential effects on oxidative stress and histopathological changes in mice, and pave the way for further studies of the long-term effects of MNV infection.
Collapse
Affiliation(s)
- Paulina Janicka
- Department of Pathology, Wrocław University of Environmental and Life Sciences, Norwida 31, 50-375 Wrocław, Poland; (P.J.); (A.P.)
- Food4Future Technologies Sp. z o.o., Tarasa Szewczenki 24, 51-351 Wroclaw, Poland;
| | - Dominika Stygar
- Department of Physiology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Jordana 19, 41-808 Zabrze, Poland;
| | - Elżbieta Chełmecka
- Department of Statistics, Department of Instrumental Analysis, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, Ostrogórska 30, 41-200 Sosnowiec, Poland;
| | - Piotr Kuropka
- Division of Histology and Embryology, Department of Biostructure and Animal Physiology, Faculty of Veterinary Medicine, Wrocław University of Environmental and Life Sciences, Norwida 25, 50-635 Wroclaw, Poland;
| | - Arkadiusz Miążek
- Department of Biochemistry and Molecular Biology, Wroclaw University of Environmental and Life Sciences, Norwida 31, 50-375 Wroclaw, Poland; (A.M.); (A.S.)
| | - Aleksandra Studzińska
- Department of Biochemistry and Molecular Biology, Wroclaw University of Environmental and Life Sciences, Norwida 31, 50-375 Wroclaw, Poland; (A.M.); (A.S.)
| | - Aleksandra Pogorzelska
- Department of Pathology, Wrocław University of Environmental and Life Sciences, Norwida 31, 50-375 Wrocław, Poland; (P.J.); (A.P.)
| | - Katarzyna Pala
- Food4Future Technologies Sp. z o.o., Tarasa Szewczenki 24, 51-351 Wroclaw, Poland;
| | - Barbara Bażanów
- Department of Pathology, Wrocław University of Environmental and Life Sciences, Norwida 31, 50-375 Wrocław, Poland; (P.J.); (A.P.)
| |
Collapse
|
4
|
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
|
5
|
McAllaster MR, Bhushan J, Balce DR, Orvedahl A, Park A, Hwang S, Sullender ME, Sibley LD, Virgin HW. Autophagy gene-dependent intracellular immunity triggered by interferon-γ. mBio 2023; 14:e0233223. [PMID: 37905813 PMCID: PMC10746157 DOI: 10.1128/mbio.02332-23] [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: 09/08/2023] [Accepted: 09/19/2023] [Indexed: 11/02/2023] Open
Abstract
IMPORTANCE Interferon-γ (IFNγ) is a critical mediator of cell-intrinsic immunity to intracellular pathogens. Understanding the complex cellular mechanisms supporting robust interferon-γ-induced host defenses could aid in developing new therapeutics to treat infections. Here, we examined the impact of autophagy genes in the interferon-γ-induced host response. We demonstrate that genes within the autophagy pathway including Wipi2, Atg9, and Gate-16, as well as ubiquitin ligase complex genes Cul3 and Klhl9 are required for IFNγ-induced inhibition of murine norovirus (norovirus hereinafter) replication in mouse cells. WIPI2 and GATE-16 were also required for IFNγ-mediated restriction of parasite growth within the Toxoplasma gondii parasitophorous vacuole in human cells. Furthermore, we found that perturbation of UFMylation pathway components led to more robust IFNγ-induced inhibition of norovirus via regulation of endoplasmic reticulum (ER) stress. Enhancing or inhibiting these dynamic cellular components could serve as a strategy to control intracellular pathogens and maintain an effective immune response.
Collapse
Affiliation(s)
- Michael R. McAllaster
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, USA
- Vir Biotechnology, San Francisco, California, USA
| | - Jaya Bhushan
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Dale R. Balce
- Vir Biotechnology, San Francisco, California, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Anthony Orvedahl
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Arnold Park
- Vir Biotechnology, San Francisco, California, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | | | - Meagan E. Sullender
- Division of Infectious Diseases, Department of Medicine, Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - L. David Sibley
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Herbert W. Virgin
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
6
|
Muharram G, Thépaut M, Lobert PE, Grandjean T, Boulard O, Delacre M, Wakeford E, Wheeler R, Poulin LF, Boneca IG, Lafont F, Michallet MC, Hober D, Cadwell K, Chamaillard M. Activation of Nod2 signaling upon norovirus infection enhances antiviral immunity and susceptibility to colitis. Gut Microbes 2023; 15:2249960. [PMID: 37655966 PMCID: PMC10478738 DOI: 10.1080/19490976.2023.2249960] [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: 05/23/2023] [Revised: 08/13/2023] [Accepted: 08/16/2023] [Indexed: 09/02/2023] Open
Abstract
Over 90% of epidemic non-bacterial gastroenteritis are caused by human noroviruses (NoVs), which persist in a substantial subset of people allowing their spread worldwide. This has led to a significant number of endemic cases and up to 70,000 children deaths in developing countries. NoVs are primarily transmitted through the fecal-oral route. To date, studies have focused on the influence of the gut microbiota on enteric viral clearance by mucosal immunity. In this study, the use of mouse norovirus S99 (MNoV_S99) and CR6 (MNoV_CR6), two persistent strains, allowed us to provide evidence that the norovirus-induced exacerbation of colitis severity relied on bacterial sensing by nucleotide-binding oligomerization domain 2 (Nod2). Consequently, Nod2-deficient mice showed reduced levels of gravity of Dextran sodium sulfate (DSS)-induced colitis with both viral strains. And MNoV_CR6 viremia was heightened in Nod2-/- mice in comparison with animals hypomorphic for Atg16l1, which are prone to aggravated inflammation under DSS. Accordingly, the infection of macrophages derived from WT mice promoted the phosphorylation of Signal Transducer and Activator of Transcription 1 (STAT1) and NOD2's expression levels. Higher secretion of Tumor Necrosis Factor alpha (TNFα ) following NOD2 activation and better viral clearance were measured in these cells. By contrast, reduced levels of pSTAT1 and blunted downstream secretion of TNFα were found in Nod2-deficient macrophages infected by MNoV_S99. Hence, our results uncover a previously unidentified virus-host-bacterial interplay that may represent a novel therapeutic target for treating noroviral origin gastroenteritis that may be linked with susceptibility to several common illnesses such as Crohn's disease.
Collapse
Affiliation(s)
- Ghaffar Muharram
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, Lille, U1019 - UMR 9017 - CIIL -Centre d'Infection et d'Immunité de Lille, Lille, France
| | - Marion Thépaut
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, Lille, U1019 - UMR 9017 - CIIL -Centre d'Infection et d'Immunité de Lille, Lille, France
| | | | - Teddy Grandjean
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, Lille, U1019 - UMR 9017 - CIIL -Centre d'Infection et d'Immunité de Lille, Lille, France
| | - Olivier Boulard
- Faculté de Médecine, CHU Lille, Laboratoire de Virologie, Univ. Lille, Lille, France
| | - Myriam Delacre
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, Lille, U1019 - UMR 9017 - CIIL -Centre d'Infection et d'Immunité de Lille, Lille, France
| | - Emmrich Wakeford
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, Lille, U1019 - UMR 9017 - CIIL -Centre d'Infection et d'Immunité de Lille, Lille, France
| | - Richard Wheeler
- TERI (Tumor Escape, Resistance and Immunity), Centre de recherche en cancérologie de Lyon, Centre Léon Bérard, Université de Lyon, Université Claude Bernard Lyon 1, Inserm 1052, CNRS 5286, Lyon, France
| | - Lionel Franz Poulin
- Faculté de Médecine, CHU Lille, Laboratoire de Virologie, Univ. Lille, Lille, France
| | - Ivo Gomperts Boneca
- TERI (Tumor Escape, Resistance and Immunity), Centre de recherche en cancérologie de Lyon, Centre Léon Bérard, Université de Lyon, Université Claude Bernard Lyon 1, Inserm 1052, CNRS 5286, Lyon, France
| | - Frank Lafont
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, Lille, U1019 - UMR 9017 - CIIL -Centre d'Infection et d'Immunité de Lille, Lille, France
| | - Marie-Cécile Michallet
- Institut Pasteur, Université Paris Cité CNRS UMR6047, INSERM U1306, Unité de Biologie et génétique de la paroi bactérienne, Paris, France
| | - Didier Hober
- Laboratory of Cell Physiology, INSERM U1003, University of Lille, Lille, France
| | - Ken Cadwell
- Kimmel Center for Biology and Medicine at the Skirball Institute, New York University Grossman School of Medicine, New York, NY, USA
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, USA
- Division of Gastroenterology and Hepatology, Department of Medicine, New York University Langone Health, New York, NY, USA
| | - Mathias Chamaillard
- Faculté de Médecine, CHU Lille, Laboratoire de Virologie, Univ. Lille, Lille, France
| |
Collapse
|
7
|
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
|
8
|
Bai GH, Tsai MC, Lin SC, Hsu YH, Chen SY. Unraveling the interplay between norovirus infection, gut microbiota, and novel antiviral approaches: a comprehensive review. Front Microbiol 2023; 14:1212582. [PMID: 37485533 PMCID: PMC10359435 DOI: 10.3389/fmicb.2023.1212582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 06/15/2023] [Indexed: 07/25/2023] Open
Abstract
Norovirus infection is a leading cause of acute gastroenteritis worldwide and can also cause harmful chronic infections in individuals with weakened immune systems. The role of the gut microbiota in the interactions between the host and noroviruses has been extensively studied. While most past studies were conducted in vitro or focused on murine noroviruses, recent research has expanded to human noroviruses using in vivo or ex vivo human intestinal enteroids culture studies. The gut microbiota has been observed to have both promoting and inhibiting effects on human noroviruses. Understanding the interaction between noroviruses and the gut microbiota or probiotics is crucial for studying the pathogenesis of norovirus infection and its potential implications, including probiotics and vaccines for infection control. Recently, several clinical trials of probiotics and norovirus vaccines have also been published. Therefore, in this review, we discuss the current understanding and recent updates on the interactions between noroviruses and gut microbiota, including the impact of norovirus on the microbiota profile, pro-viral and antiviral effects of microbiota on norovirus infection, the use of probiotics for treating norovirus infections, and human norovirus vaccine development.
Collapse
Affiliation(s)
- Geng-Hao Bai
- Department of Internal Medicine, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Meng-Chen Tsai
- Department of General Medicine, Taipei Medical University Hospital, Taipei, Taiwan
| | - Sheng-Chieh Lin
- Department of Pediatrics, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Pediatrics, Division of Allergy, Asthma and Immunology, Shuang Ho Hospital, New Taipei, Taiwan
| | - Yi-Hsiang Hsu
- Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, United States
- Broad Institute of MIT and Harvard, Cambridge, MA, United States
| | - Shih-Yen Chen
- Department of Pediatrics, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Pediatrics, Division of Pediatric Gastroenterology and Hepatology, Shuang Ho Hospital, New Taipei, Taiwan
- TMU Research Center for Digestive Medicine, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
9
|
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: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [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
|
10
|
Péloquin L, Goetz C, Jubinville E, Jean J. Protective Effect of Select Bacterial Species Representative of Fresh Produce on Human Norovirus Surrogates Exposed to Disinfecting Pulsed Light. Appl Environ Microbiol 2023; 89:e0004323. [PMID: 37154750 PMCID: PMC10231187 DOI: 10.1128/aem.00043-23] [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: 01/10/2023] [Accepted: 03/29/2023] [Indexed: 05/10/2023] Open
Abstract
Contamination of berries and leafy greens with human norovirus (HuNoV) is a major cause of outbreaks of epidemic gastroenteritis worldwide. Using murine norovirus type 1 (MNV-1) and Tulane virus, we studied the possible extension of HuNoV persistence by biofilm-producing epiphytic bacteria on fresh produce. Nine bacterial species frequently found on the surface of berries and leafy greens (Bacillus cereus, Enterobacter cloacae, Escherichia coli, Kocuria kristinae, Lactobacillus plantarum, Pantoea agglomerans, Pseudomonas fluorescens, Raoultella terrigena, and Xanthomonas campestris) were evaluated for the ability to form biofilms in the MBEC Assay Biofilm Inoculator and in 96-well microplates. The biofilm-forming bacteria were further tested for binding MNV-1 and Tulane virus and the ability to protect them against loss of capsid integrity upon exposure to disinfecting pulsed light at a fluence of 11.52 J/cm2. Based on viral reductions, MNV-1 did not benefit from attachment to biofilm whereas Tulane virus was significantly more resistant than the control when attached to biofilms of E. cloacae (P ≤ 0.01), E. coli (P ≤ 0.01), K. kristinae (P ≤ 0.01), P. agglomerans (P ≤ 0.05), or P. fluorescens (P ≤ 0.0001). Enzymatic dispersion of biofilm and microscopic observations suggest that the biofilm matrix composition may contribute to the virus resistance. Our results indicate that direct virus-biofilm interaction protects Tulane virus against disinfecting pulsed light, and that HuNoV on fresh produce therefore might resist such treatment more than suggested by laboratory tests so far. IMPORTANCE Recent studies have shown that bacteria may be involved in the attachment of HuNoV to the surface of fresh produce. Because these foods are difficult to disinfect by conventional methods without compromising product quality, nonthermal nonchemical disinfectants such as pulsed light are being investigated. We seek to understand how HuNoV interacts with epiphytic bacteria, particularly with biofilms formed by bacterial epiphytes, with cells and extracellular polymeric substances, and to determine if it thus escapes inactivation by pulsed light. The results of this study should advance understanding of the effects of epiphytic biofilms on the persistence of HuNoV particle integrity after pulsed light treatment and thus guide the design of novel pathogen control strategies in the food industry.
Collapse
Affiliation(s)
- Laurence Péloquin
- Department of Food Sciences, Institute of Nutrition and Functional Foods (INAF), Université Laval, Quebec City, Quebec, Canada
| | - Coralie Goetz
- Department of Food Sciences, Institute of Nutrition and Functional Foods (INAF), Université Laval, Quebec City, Quebec, Canada
| | - Eric Jubinville
- Department of Food Sciences, Institute of Nutrition and Functional Foods (INAF), Université Laval, Quebec City, Quebec, Canada
| | - Julie Jean
- Department of Food Sciences, Institute of Nutrition and Functional Foods (INAF), Université Laval, Quebec City, Quebec, Canada
| |
Collapse
|
11
|
Lee H, Lee G, Cho YH, Song Y, Ko G. Chemokine CCL6 Plays Key Role in the Inhibitory Effect of Vitamin A on Norovirus Infection. J Microbiol 2023:10.1007/s12275-023-00047-3. [PMID: 37233907 DOI: 10.1007/s12275-023-00047-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 03/17/2023] [Accepted: 03/20/2023] [Indexed: 05/27/2023]
Abstract
Norovirus (NoV) is the most common viral cause of acute gastroenteritis worldwide. Vitamin A has demonstrated the potential to protect against gastrointestinal infections. However, the effects of vitamin A on human norovirus (HuNoV) infections remain poorly understood. This study aimed to investigate how vitamin A administration affects NoV replication. We demonstrated that treatment with retinol or retinoic acid (RA) inhibited NoV replication in vitro based on their effects on HuNoV replicon-bearing cells and murine norovirus-1 (MNV-1) replication in murine cells. MNV replication in vitro showed significant transcriptomic changes, which were partially reversed by retinol treatment. RNAi knockdown of CCL6, a chemokine gene that was downregulated by MNV infection but upregulated by retinol administration, resulted in increased MNV replication in vitro. This suggested a role of CCL6 in the host response to MNV infections. Similar gene expression patterns were observed in the murine intestine after oral administration of RA and/or MNV-1.CW1. CCL6 directly decreased HuNoV replication in HG23 cells, and might indirectly regulate the immune response against NoV infection. Finally, relative replication levels of MNV-1.CW1 and MNV-1.CR6 were significantly increased in CCL6 knockout RAW 264.7 cells. This study is the first to comprehensively profile transcriptomes in response to NoV infection and vitamin A treatment in vitro, and thus may provide new insights into dietary prophylaxis and NoV infections.
Collapse
Affiliation(s)
- Heetae Lee
- College of Pharmacy, Sahmyook University, Seoul, 01795, Republic of Korea.
| | - Giljae Lee
- Center for Human and Environmental Microbiome, School of Public Health, Seoul National University, Seoul, 08826, Republic of Korea
| | - You-Hee Cho
- Department of Pharmacy, College of Pharmacy, CHA University, Seongnam, 13488, Republic of Korea
| | - Youngcheon Song
- College of Pharmacy, Sahmyook University, Seoul, 01795, Republic of Korea
| | - GwangPyo Ko
- Center for Human and Environmental Microbiome, School of Public Health, Seoul National University, Seoul, 08826, Republic of Korea.
| |
Collapse
|
12
|
Jahun AS, Sorgeloos F, Chaudhry Y, Arthur SE, Hosmillo M, Georgana I, Izuagbe R, Goodfellow IG. Leaked genomic and mitochondrial DNA contribute to the host response to noroviruses in a STING-dependent manner. Cell Rep 2023; 42:112179. [PMID: 36943868 DOI: 10.1016/j.celrep.2023.112179] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 10/11/2022] [Accepted: 02/12/2023] [Indexed: 03/23/2023] Open
Abstract
The cGAS-STING pathway is central to the interferon response against DNA viruses. However, recent studies are increasingly demonstrating its role in the restriction of some RNA viruses. Here, we show that the cGAS-STING pathway also contributes to the interferon response against noroviruses, currently the commonest causes of infectious gastroenteritis worldwide. We show a significant reduction in interferon-β induction and a corresponding increase in viral replication in norovirus-infected cells after deletion of STING, cGAS, or IFI16. Further, we find that immunostimulatory host genome-derived DNA and mitochondrial DNA accumulate in the cytosol of norovirus-infected cells. Lastly, overexpression of the viral NS4 protein is sufficient to drive the accumulation of cytosolic DNA. Together, our data find a role for cGAS, IFI16, and STING in the restriction of noroviruses and show the utility of host genomic DNA as a damage-associated molecular pattern in cells infected with an RNA virus.
Collapse
Affiliation(s)
- Aminu S Jahun
- Division of Virology, Department of Pathology, University of Cambridge, Addenbrooke's Hospital Level 5, Hills Road, Cambridge CB2 0QQ, UK.
| | - Frederic Sorgeloos
- Division of Virology, Department of Pathology, University of Cambridge, Addenbrooke's Hospital Level 5, Hills Road, Cambridge CB2 0QQ, UK; Université catholique de Louvain, de Duve Institute, MIPA-VIRO 74-49, 74 Avenue Hippocrate, B-1200 Brussels, Belgium
| | - Yasmin Chaudhry
- Division of Virology, Department of Pathology, University of Cambridge, Addenbrooke's Hospital Level 5, Hills Road, Cambridge CB2 0QQ, UK
| | - Sabastine E Arthur
- Division of Virology, Department of Pathology, University of Cambridge, Addenbrooke's Hospital Level 5, Hills Road, Cambridge CB2 0QQ, UK
| | - Myra Hosmillo
- Division of Virology, Department of Pathology, University of Cambridge, Addenbrooke's Hospital Level 5, Hills Road, Cambridge CB2 0QQ, UK
| | - Iliana Georgana
- Division of Virology, Department of Pathology, University of Cambridge, Addenbrooke's Hospital Level 5, Hills Road, Cambridge CB2 0QQ, UK
| | - Rhys Izuagbe
- Division of Virology, Department of Pathology, University of Cambridge, Addenbrooke's Hospital Level 5, Hills Road, Cambridge CB2 0QQ, UK
| | - Ian G Goodfellow
- Division of Virology, Department of Pathology, University of Cambridge, Addenbrooke's Hospital Level 5, Hills Road, Cambridge CB2 0QQ, UK.
| |
Collapse
|
13
|
Yamamoto S, Sudo-Yokoyama Y, Ogasawara N, Yokota SI. Rapid, simple, and cost-effective plaque assay for murine norovirus using microcrystalline cellulose. J Virol Methods 2023; 316:114715. [PMID: 36940863 DOI: 10.1016/j.jviromet.2023.114715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 02/22/2023] [Accepted: 03/17/2023] [Indexed: 03/23/2023]
Abstract
Murine norovirus (MNV) is used widely as a practical alternative to human norovirus (HuNoV). Plaque-forming assays for MNV are important for developing therapeutic agents against HuNoV infections. Although agarose-overlay MNV assays have been reported, recent improvements in cellulose derivatives suggest that they could be optimized further, particularly with respect to improving the overlay material. To determine which overlay material is optimal for the MNV plaque assay, we compared four typical cellulose derivatives [microcrystalline cellulose (MCC), hydroxyethyl cellulose (HEC), hydroxypropyl methylcellulose (HPMC), and carboxymethyl cellulose (CMC)] with conventional agarose. We found that 3.5% (w/v) MCC-containing medium provided clear round-shaped plaques on RAW 264.7 cells 1 day after inoculation; the visibility of plaques was comparable with that of the original agarose-overlay assay. Removing residual MCC powder from the MCC-overlay assay before fixing was important for obtaining distinct plaques that are clearly countable. Finally, after calculating the plaque diameter as a percentage of well diameter, we found that 12- and 24-well plates were better than other plates for accurate plaque counting. The MCC-based MNV plaque assay is cost-effective and rapid, and produces plaques that are easy to count. Accurate virus quantification using this optimized plaque assay will enable reliable estimation of norovirus titers.
Collapse
Affiliation(s)
- Soh Yamamoto
- Department of Microbiology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Yuka Sudo-Yokoyama
- Department of Microbiology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Noriko Ogasawara
- Department of Microbiology, Sapporo Medical University School of Medicine, Sapporo, Japan; Department of Otolaryngology-Head and Neck Surgery, Sapporo Medical University School of Medicine, Sapporo, Japan.
| | - Shin-Ichi Yokota
- Department of Microbiology, Sapporo Medical University School of Medicine, Sapporo, Japan
| |
Collapse
|
14
|
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
|
15
|
Zhang Q, Zhu S, Zhang X, Su L, Ni J, Zhang Y, Fang L. Recent insights into reverse genetics of norovirus. Virus Res 2023; 325:199046. [PMID: 36657615 DOI: 10.1016/j.virusres.2023.199046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Revised: 12/23/2022] [Accepted: 01/15/2023] [Indexed: 01/18/2023]
Abstract
Norovirus is the leading cause of viral gastroenteritis globally, and poses substantial threats to public health. Despite substantial progress made in preventing norovirus diseases, the lack of a robust virus culture system has hampered biological research and effective strategies to combat this pathogen. Reverse genetic system is the technique to generate infectious viruses from cloned genetic constructs, which is a powerful tool for the investigation of viral pathogenesis and for the development of novel drugs and vaccines. The strategies of reverse genetics include bacterial artificial chromosomes, vaccinia virus vectors, and entirely plasmid-based systems. Since each strategy has its pros and cons, choosing appropriate approaches will greatly improve the efficiency of virus rescue. Reverse genetic systems that have been employed for norovirus greatly extend its life cycle and facilitate the development of medical countermeasures. In this review, we summarize the current knowledge on the structure, transmission, genetic evolution and clinical manifestations of norovirus, and describe recent advances in the studies of norovirus reverse genetics as well as its future prospects for therapeutics and vaccine development.
Collapse
Affiliation(s)
- Qinyi Zhang
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, China; Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, China
| | - Shuirong Zhu
- Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, China
| | | | - Lingxuan Su
- Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, China
| | - Jun Ni
- Department of Critical Care Medicine, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, China
| | - Yanjun Zhang
- Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, China.
| | - Lei Fang
- Department of Critical Care Medicine, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, China.
| |
Collapse
|
16
|
Sullender ME, Pierce LR, Annaswamy Srinivas M, Crockett SL, Dunlap BF, Rodgers R, Schriefer LA, Kennedy EA, Stewart BM, Doench JG, Baldridge MT, Orchard RC. Selective Polyprotein Processing Determines Norovirus Sensitivity to Trim7. J Virol 2022; 96:e0070722. [PMID: 35972292 PMCID: PMC9472627 DOI: 10.1128/jvi.00707-22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 07/28/2022] [Indexed: 11/20/2022] Open
Abstract
Noroviruses are a leading cause of gastroenteritis worldwide, yet the molecular mechanisms of how host antiviral factors restrict norovirus infection are poorly understood. Here, we present a CRISPR activation screen that identifies mouse genes which inhibit murine norovirus (MNV) replication. Detailed analysis of the major hit Trim7 demonstrates a potent inhibition of the early stages of MNV replication. Leveraging in vitro evolution, we identified MNV mutants that escape Trim7 restriction by altering the cleavage of the viral NS6-7 polyprotein precursor. NS6, but not the NS6-7 precursor, directly binds the substrate-binding domain of Trim7. Surprisingly, the selective polyprotein processing that enables Trim7 evasion inflicts a significant evolutionary burden, as viruses with decreased NS6-7 cleavage are strongly attenuated in viral replication and pathogenesis. Our data provide an unappreciated mechanism of viral evasion of cellular antiviral factors through selective polyprotein processing and highlight the evolutionary tradeoffs in acquiring resistance to host restriction factors. IMPORTANCE To maximize a limited genetic capacity, viruses encode polyproteins that can be subsequently separated into individual components by viral proteases. While classically viewed as a means of economy, recent findings have indicated that polyprotein processing can spatially and temporally coordinate the distinct phases of the viral life cycle. Here, we present a function for alternative polyprotein processing centered on immune defense. We discovered that selective polyprotein processing of the murine norovirus polyprotein shields MNV from restriction by the host antiviral protein Trim7. Trim7 can bind the viral protein NS6 but not the viral precursor protein NS6-7. Our findings provide insight into the evolutionary pressures that define patterns of viral polyprotein processing and uncover a trade-off between viral replication and immune evasion.
Collapse
Affiliation(s)
- Meagan E. Sullender
- Division of Infectious Diseases, Department of Medicine, Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Linley R. Pierce
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | | | - Stacey L. Crockett
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Bria F. Dunlap
- Division of Infectious Diseases, Department of Medicine, Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Rachel Rodgers
- Division of Infectious Diseases, Department of Medicine, Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Lawrence A. Schriefer
- Division of Infectious Diseases, Department of Medicine, Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine, St. Louis, Missouri, 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, Missouri, USA
| | - Brittany M. Stewart
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - John G. Doench
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, 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, Missouri, USA
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Robert C. Orchard
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
17
|
Bhar S, Zhao G, Bartel JD, Sterchele H, Del Mazo A, Emerson LE, Edelmann MJ, Jones MK. Bacterial extracellular vesicles control murine norovirus infection through modulation of antiviral immune responses. Front Immunol 2022; 13:909949. [PMID: 35990695 PMCID: PMC9386532 DOI: 10.3389/fimmu.2022.909949] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 07/11/2022] [Indexed: 11/13/2022] Open
Abstract
Human norovirus is the primary cause of non-bacterial gastroenteritis globally and is the second leading cause of diarrheal deaths in children in developing countries. However, effective therapeutics which prevent or clear norovirus infection are not yet available due to a lack of understanding regarding norovirus pathogenesis. Evidence shows that noroviruses can bind to the surface of commensal bacteria, and the presence of these bacteria alters both acute and persistent murine norovirus infection through the modulation of host immune responses. Interestingly, norovirus-bacterial interactions also affect the bacteria by inducing bacterial stress responses and increasing the production of bacterial extracellular vesicles. Given the established ability of these vesicles to easily cross the intestinal barriers, enter the lamina propria, and modulate host responses, we hypothesized that bacterial extracellular vesicles influence murine norovirus infection through modulation of the antiviral immune response. In this study, we show that murine norovirus can attach to purified bacterial vesicles, facilitating co-inoculation of target cells with both virus and vesicle. Furthermore, we have found that when murine noroviruses and vesicles are used to co-inoculate macrophages, viral infection is reduced compared to virus infection alone. Specifically, co-inoculation with bacterial vesicles results in higher production and release of pro-inflammatory cytokines in response to viral infection. Ultimately, given that murine norovirus infection increases bacterial vesicle production in vivo, these data indicate that bacterial vesicles may serve as a mechanism by which murine norovirus infection is ultimately controlled and limited to a short-term disease.
Collapse
|
18
|
Eldridge DE, Hsu CC. Antibody Production Remains Intact Despite Loss of Bone Marrow B cells in Murine Norovirus Infected Stat1-/- Mice. Comp Med 2021; 71:502-511. [PMID: 34794531 DOI: 10.30802/aalas-cm-21-000054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Murine norovirus (MNV), which can be used as a model system to study human noroviruses, can infect macrophages/monocytes, neutrophils, dendritic, intestinal epithelial, T and B cells, and is highly prevalent in laboratory mice. We previouslyshowed that MNV infection significantly reduces bone marrow B cell populations in a Stat1-dependent manner. We show here that while MNV-infected Stat1-/- mice have significant losses of bone marrow B cells, splenic B cells capable of mounting an antibody response to novel antigens retain the ability to expand. We also investigated whether increased granulopoiesis after MNV infection was causing B cell loss. We found that administration of anti-G-CSF antibody inhibits the pronounced bone marrow granulopoiesis induced by MNV infection of Stat1-/- mice, but this inhibition did not rescue bone marrow B cell losses. Therefore, MNV-infected Stat1-/- mice can still mount a robust humoral immune response despite decreased bone marrow B cells. This suggests that further investigation will be needed to identify other indirect factors or mechanisms that are responsible for the bone marrow B cell losses seen after MNV infection. In addition, this work contributes to our understanding of the potential physiologic effects of Stat1-related disruptions in research mouse colonies that may be endemically infected with MNV.
Collapse
|
19
|
Akt plays differential roles during the life cycles of acute and persistent murine norovirus strains in macrophages. J Virol 2021; 96:e0192321. [PMID: 34787460 DOI: 10.1128/jvi.01923-21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Akt (Protein kinase B) is a key signaling protein in eukaryotic cells that controls many cellular processes such as glucose metabolism and cell proliferation for survival. As obligate intracellular pathogens, viruses modulate host cellular processes, including Akt signaling, for optimal replication. The mechanisms by which viruses modulate Akt and the resulting effects on the infectious cycle differ widely depending on the virus. In this study, we explored the effect of Akt serine 473 phosphorylation (p-Akt) during murine norovirus (MNV) infection. p-Akt increased during infection of murine macrophages with acute MNV-1 and persistent CR3 and CR6 strains. Inhibition of Akt with MK2206, an inhibitor of all three isoforms of Akt (Akt1/2/3), reduced infectious virus progeny of all three virus strains. This reduction was due to decreased viral genome replication (CR3), defective virus assembly (MNV-1), or diminished cellular egress (CR3 and CR6) in a virus strain-dependent manner. Collectively, our data demonstrate that Akt activation increases in macrophages during the later stages of the MNV infectious cycle, which may enhance viral infection in unique ways for different virus strains. The data, for the first time, indicate a role for Akt signaling in viral assembly and highlight additional phenotypic differences between closely related MNV strains. Importance Human noroviruses (HNoV) are a leading cause of viral gastroenteritis, resulting in high annual economic burden and morbidity; yet there are no small animal models supporting productive HNoV infection, or robust culture systems producing cell culture-derived virus stocks. As a result, research on drug discovery and vaccine development against norovirus infection has been challenging, and no targeted antivirals or vaccines against HNoV are approved. On the other hand, murine norovirus (MNV) replicates to high titers in cell culture and is a convenient and widespread model in norovirus research. Our data demonstrate the importance of Akt signaling during the late stage of the MNV life cycle. Notably, the effect of Akt signaling on genome replication, virus assembly and cellular egress is virus strain specific, highlighting the diversity of biological phenotypes despite small genetic variability among norovirus strains. This study is the first to demonstrate a role for Akt in viral assembly.
Collapse
|
20
|
Abstract
Human noroviruses (HuNoVs) are increasingly becoming the main cause of transmissible gastroenteritis worldwide, with hundreds of thousands of deaths recorded annually. Yet, decades after their discovery, there is still no effective treatment or vaccine. Efforts aimed at developing vaccines or treatment will benefit from a greater understanding of norovirus-host interactions, including the host response to infection. In this review, we provide a concise overview of the evidence establishing the significance of type I and type III interferon (IFN) responses in the restriction of noroviruses. We also critically examine our current understanding of the molecular mechanisms of IFN induction in norovirus-infected cells, and outline the diverse strategies deployed by noroviruses to supress and/or avoid host IFN responses. It is our hope that this review will facilitate further discussion and increase interest in this area.
Collapse
Affiliation(s)
- Aminu S. Jahun
- Division of Virology, Department of Pathology, University of Cambridge, Addenbrooke’s Hospital, Cambridge CB2 0QQ, UK
- *Correspondence: Aminu S. Jahun,
| | - Ian G. Goodfellow
- Division of Virology, Department of Pathology, University of Cambridge, Addenbrooke’s Hospital, Cambridge CB2 0QQ, UK
| |
Collapse
|
21
|
Dallari S, Heaney T, Rosas-Villegas A, Neil JA, Wong SY, Brown JJ, Urbanek K, Herrmann C, Depledge DP, Dermody TS, Cadwell K. Enteric viruses evoke broad host immune responses resembling those elicited by the bacterial microbiome. Cell Host Microbe 2021; 29:1014-1029.e8. [PMID: 33894129 PMCID: PMC8192460 DOI: 10.1016/j.chom.2021.03.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 02/04/2021] [Accepted: 03/23/2021] [Indexed: 02/08/2023]
Abstract
The contributions of the viral component of the microbiome-the virome-to the development of innate and adaptive immunity are largely unknown. Here, we systematically defined the host response in mice to a panel of eukaryotic enteric viruses representing six different families. Infections with most of these viruses were asymptomatic in the mice, the magnitude and duration of which was dependent on the microbiota. Flow cytometric and transcriptional profiling of mice mono-associated with these viruses unveiled general adaptations by the host, such as lymphocyte differentiation and IL-22 signatures in the intestine, as well as numerous viral-strain-specific responses that persisted. Comparison with a dataset derived from analogous bacterial mono-association in mice identified bacterial species that evoke an immune response comparable with the viruses we examined. These results expand an understanding of the immune space occupied by the enteric virome and underscore the importance of viral exposure events.
Collapse
Affiliation(s)
- Simone Dallari
- Kimmel Center for Biology and Medicine at the Skirball Institute, New York University Grossman School of Medicine, New York, NY, USA
| | - Thomas Heaney
- Kimmel Center for Biology and Medicine at the Skirball Institute, New York University Grossman School of Medicine, New York, NY, USA
| | - Adriana Rosas-Villegas
- Kimmel Center for Biology and Medicine at the Skirball Institute, New York University Grossman School of Medicine, New York, NY, USA
| | - Jessica A Neil
- Kimmel Center for Biology and Medicine at the Skirball Institute, New York University Grossman School of Medicine, New York, NY, USA
| | - Serre-Yu Wong
- Kimmel Center for Biology and Medicine at the Skirball Institute, New York University Grossman School of Medicine, New York, NY, USA; Department of Medicine, Henry D. Janowitz Division of Gastroenterology, Susan and Leonard Feinstein Inflammatory Bowel Disease Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Judy J Brown
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, TN, USA; Department of Biology, Trevecca Nazarene University, Nashville, TN, USA
| | - Kelly Urbanek
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Christin Herrmann
- Kimmel Center for Biology and Medicine at the Skirball Institute, New York University Grossman School of Medicine, New York, NY, USA
| | - Daniel P Depledge
- Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - Terence S Dermody
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Microbiology, New York University Grossman School of Medicine, New York, NY, USA
| | - Ken Cadwell
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, USA; Division of Gastroenterology and Hepatology, Department of Medicine, New York University Langone Health, New York, NY, USA.
| |
Collapse
|
22
|
Walker FC, Hassan E, Peterson ST, Rodgers R, Schriefer LA, Thompson CE, Li Y, Kalugotla G, Blum-Johnston C, Lawrence D, McCune BT, Graziano VR, Lushniak L, Lee S, Roth AN, Karst SM, Nice TJ, Miner JJ, Wilen CB, Baldridge MT. Norovirus evolution in immunodeficient mice reveals potentiated pathogenicity via a single nucleotide change in the viral capsid. PLoS Pathog 2021; 17:e1009402. [PMID: 33705489 PMCID: PMC7987144 DOI: 10.1371/journal.ppat.1009402] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 03/23/2021] [Accepted: 02/17/2021] [Indexed: 02/06/2023] Open
Abstract
Interferons (IFNs) are key controllers of viral replication, with intact IFN responses suppressing virus growth and spread. Using the murine norovirus (MNoV) system, we show that IFNs exert selective pressure to limit the pathogenic evolutionary potential of this enteric virus. In animals lacking type I IFN signaling, the nonlethal MNoV strain CR6 rapidly acquired enhanced virulence via conversion of a single nucleotide. This nucleotide change resulted in amino acid substitution F514I in the viral capsid, which led to >10,000-fold higher replication in systemic organs including the brain. Pathogenicity was mediated by enhanced recruitment and infection of intestinal myeloid cells and increased extraintestinal dissemination of virus. Interestingly, the trade-off for this mutation was reduced fitness in an IFN-competent host, in which CR6 bearing F514I exhibited decreased intestinal replication and shedding. In an immunodeficient context, a spontaneous amino acid change can thus convert a relatively avirulent viral strain into a lethal pathogen.
Collapse
Affiliation(s)
- Forrest C. Walker
- Division of Infectious Diseases, Department of Medicine, Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Ebrahim Hassan
- Division of Infectious Diseases, Department of Medicine, Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - 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, Missouri, United States of America
| | - Rachel Rodgers
- Division of Infectious Diseases, Department of Medicine, Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Lawrence A. Schriefer
- Division of Infectious Diseases, Department of Medicine, Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Cassandra E. Thompson
- Division of Infectious Diseases, Department of Medicine, Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Yuhao Li
- Division of Infectious Diseases, Department of Medicine, Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Gowri Kalugotla
- Division of Infectious Diseases, Department of Medicine, Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Carla Blum-Johnston
- Division of Infectious Diseases, Department of Medicine, Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Dylan Lawrence
- Division of Infectious Diseases, Department of Medicine, Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Broc T. McCune
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Vincent R. Graziano
- Departments of Laboratory Medicine & Immunobiology, Yale School of Medicine, New Haven, Connecticut, United States of America
| | - Larissa Lushniak
- Division of Infectious Diseases, Department of Medicine, Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Sanghyun Lee
- Division of Infectious Diseases, Department of Medicine, Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Alexa N. Roth
- Department of Molecular Genetics & Microbiology, Emerging Pathogens Institute, University of Florida, Gainesville, Florida, United States of America
| | - Stephanie M. Karst
- Department of Molecular Genetics & Microbiology, Emerging Pathogens Institute, University of Florida, Gainesville, Florida, United States of America
| | - Timothy J. Nice
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Jonathan J. Miner
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Craig B. Wilen
- Departments of Laboratory Medicine & Immunobiology, Yale School of Medicine, New Haven, Connecticut, United States of America
| | - 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, Missouri, United States of America
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| |
Collapse
|
23
|
CD300lf Conditional Knockout Mouse Reveals Strain-Specific Cellular Tropism of Murine Norovirus. J Virol 2021; 95:JVI.01652-20. [PMID: 33177207 DOI: 10.1128/jvi.01652-20] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 11/02/2020] [Indexed: 02/07/2023] Open
Abstract
Noroviruses are a leading cause of gastrointestinal infection in humans and mice. Understanding human norovirus (HuNoV) cell tropism has important implications for our understanding of viral pathogenesis. Murine norovirus (MNoV) is extensively used as a surrogate model for HuNoV. We previously identified CD300lf as the receptor for MNoV. Here, we generated a Cd300lf conditional knockout (CD300lfF/F ) mouse to elucidate the cell tropism of persistent and nonpersistent strains of murine norovirus. Using this mouse model, we demonstrated that CD300lf expression on intestinal epithelial cells (IECs), and on tuft cells in particular, is essential for transmission of the persistent MNoV strain CR6 (MNoVCR6) in vivo In contrast, the nonpersistent MNoV strain CW3 (MNoVCW3) does not require CD300lf expression on IECs for infection. However, deletion of CD300lf in myelomonocytic cells (LysM Cre+) partially reduces CW3 viral load in lymphoid and intestinal tissues. Disruption of CD300lf expression on B cells (CD19 Cre), neutrophils (Mrp8 Cre), and dendritic cells (CD11c Cre) did not affect MNoVCW3 viral RNA levels. Finally, we show that the transcription factor STAT1, which is critical for the innate immune response, partially restricts the cell tropism of MNoVCW3 to LysM+ cells. Taken together, these data demonstrate that CD300lf expression on tuft cells is essential for MNoVCR6; that myelomonocytic cells are a major, but not exclusive, target cell of MNoVCW3; and that STAT1 signaling restricts the cellular tropism of MNoVCW3 This study provides the first genetic system for studying the cell type-specific role of CD300lf in norovirus pathogenesis.IMPORTANCE Human noroviruses (HuNoVs) are a leading cause of gastroenteritis resulting in up to 200,000 deaths each year. The receptor and cell tropism of HuNoV in immunocompetent humans are unclear. We use murine norovirus (MNoV) as a model for HuNoV. We recently identified CD300lf as the sole physiologic receptor for MNoV. Here, we leverage this finding to generate a Cd300lf conditional knockout mouse to decipher the contributions of specific cell types to MNoV infection. We demonstrate that persistent MNoVCR6 requires CD300lf expression on tuft cells. In contrast, multiple CD300lf+ cell types, dominated by myelomonocytic cells, are sufficient for nonpersistent MNoVCW3 infection. CD300lf expression on epithelial cells, B cells, neutrophils, and dendritic cells is not critical for MNoVCW3 infection. Mortality associated with the MNoVCW3 strain in Stat1-/- mice does not require CD300lf expression on LysM+ cells, highlighting that both CD300lf receptor expression and innate immunity regulate MNoV cell tropism in vivo.
Collapse
|
24
|
Park BJ, Ahn HS, Han SH, Go HJ, Kim DH, Choi C, Jung S, Myoung J, Lee JB, Park SY, Song CS, Lee SW, Lee HT, Choi IS. Analysis of the Immune Responses in the Ileum of Gnotobiotic Pigs Infected with the Recombinant GII.p12_GII.3 Human Norovirus by mRNA Sequencing. Viruses 2021; 13:v13010092. [PMID: 33440894 PMCID: PMC7826840 DOI: 10.3390/v13010092] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 12/29/2020] [Accepted: 01/08/2021] [Indexed: 11/20/2022] Open
Abstract
Norovirus genogroup II (NoV GII) induces acute gastrointestinal food-borne illness in humans. Because gnotobiotic pigs can be infected with human norovirus (HuNoV) GII, they are frequently used to analyze the associated pathogenic mechanisms and immune responses, which remain poorly understood. Recently, mRNA sequencing analysis (RNA-Seq) has been used to identify cellular responses to viruses. In this study, we investigated the host immune response and possible mechanisms involved in virus evasion in the ileum of gnotobiotic pigs infected with HuNoV by RNA-Seq. HuNoV was detected in the feces, blood, and tissues of the jejunum, ileum, colon, mesenteric lymph node, and spleen of pigs infected with HuNoV. In analysis of mRNA sequencing, expression of anti-viral protein genes such as OAS1, MX1, and MX2 were largely decreased, whereas type I IFN was increased in pigs infected with HuNoV. In addition, expression of TNF and associated anti-inflammatory cytokine genes such as IL10 was increased in HuNoV-infected pigs. Expression of genes related to natural killer (NK) cell cytotoxicity and CD8+ T cell exhaustion was increased, whereas that of MHC class I genes was decreased. Expression profiles of selected genes were further confirmed by qRT-PCR and Western blot. These results suggest that infection with HuNoV induces NK cell-mediated cytotoxicity but suppresses type I IFN- and CD8+ T cell-mediated antiviral responses.
Collapse
Affiliation(s)
- Byung-Joo Park
- Department of Infectious Diseases, College of Veterinary Medicine, Konkuk University, Gwangjin-gu, Seoul 05029, Korea; (B.-J.P.); (H.-S.A.); (S.-H.H.); (H.-J.G.); (D.-H.K.); (J.-B.L.); (S.-Y.P.); (C.-S.S.); (S.-W.L.)
| | - Hee-Seop Ahn
- Department of Infectious Diseases, College of Veterinary Medicine, Konkuk University, Gwangjin-gu, Seoul 05029, Korea; (B.-J.P.); (H.-S.A.); (S.-H.H.); (H.-J.G.); (D.-H.K.); (J.-B.L.); (S.-Y.P.); (C.-S.S.); (S.-W.L.)
| | - Sang-Hoon Han
- Department of Infectious Diseases, College of Veterinary Medicine, Konkuk University, Gwangjin-gu, Seoul 05029, Korea; (B.-J.P.); (H.-S.A.); (S.-H.H.); (H.-J.G.); (D.-H.K.); (J.-B.L.); (S.-Y.P.); (C.-S.S.); (S.-W.L.)
| | - Hyeon-Jeong Go
- Department of Infectious Diseases, College of Veterinary Medicine, Konkuk University, Gwangjin-gu, Seoul 05029, Korea; (B.-J.P.); (H.-S.A.); (S.-H.H.); (H.-J.G.); (D.-H.K.); (J.-B.L.); (S.-Y.P.); (C.-S.S.); (S.-W.L.)
| | - Dong-Hwi Kim
- Department of Infectious Diseases, College of Veterinary Medicine, Konkuk University, Gwangjin-gu, Seoul 05029, Korea; (B.-J.P.); (H.-S.A.); (S.-H.H.); (H.-J.G.); (D.-H.K.); (J.-B.L.); (S.-Y.P.); (C.-S.S.); (S.-W.L.)
| | - Changsun Choi
- Department of Food and Nutrition, College of Biotechnology and Natural Resources, Chung-Ang University, Anseong, Gyeonggi 17546, Korea; (C.C.); (S.J.)
| | - Soontag Jung
- Department of Food and Nutrition, College of Biotechnology and Natural Resources, Chung-Ang University, Anseong, Gyeonggi 17546, Korea; (C.C.); (S.J.)
| | - Jinjong Myoung
- Korea Zoonosis Research Institute, Chonbuk National University, Jeonju, Jeollabuk-do 54896, Korea;
| | - Joong-Bok Lee
- Department of Infectious Diseases, College of Veterinary Medicine, Konkuk University, Gwangjin-gu, Seoul 05029, Korea; (B.-J.P.); (H.-S.A.); (S.-H.H.); (H.-J.G.); (D.-H.K.); (J.-B.L.); (S.-Y.P.); (C.-S.S.); (S.-W.L.)
| | - Seung-Yong Park
- Department of Infectious Diseases, College of Veterinary Medicine, Konkuk University, Gwangjin-gu, Seoul 05029, Korea; (B.-J.P.); (H.-S.A.); (S.-H.H.); (H.-J.G.); (D.-H.K.); (J.-B.L.); (S.-Y.P.); (C.-S.S.); (S.-W.L.)
| | - Chang-Seon Song
- Department of Infectious Diseases, College of Veterinary Medicine, Konkuk University, Gwangjin-gu, Seoul 05029, Korea; (B.-J.P.); (H.-S.A.); (S.-H.H.); (H.-J.G.); (D.-H.K.); (J.-B.L.); (S.-Y.P.); (C.-S.S.); (S.-W.L.)
| | - Sang-Won Lee
- Department of Infectious Diseases, College of Veterinary Medicine, Konkuk University, Gwangjin-gu, Seoul 05029, Korea; (B.-J.P.); (H.-S.A.); (S.-H.H.); (H.-J.G.); (D.-H.K.); (J.-B.L.); (S.-Y.P.); (C.-S.S.); (S.-W.L.)
| | - Hoon-Taek Lee
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Korea;
| | - In-Soo Choi
- Department of Infectious Diseases, College of Veterinary Medicine, Konkuk University, Gwangjin-gu, Seoul 05029, Korea; (B.-J.P.); (H.-S.A.); (S.-H.H.); (H.-J.G.); (D.-H.K.); (J.-B.L.); (S.-Y.P.); (C.-S.S.); (S.-W.L.)
- Correspondence: ; Tel.: +82-2049-6228
| |
Collapse
|
25
|
Na Z, Bo J, Yifei Y, Fuyuan C, Bin H, Yanshu Z, Huan J, Jingliang S, Shuang L. Isolation and Identification of a Murine Norovirus Persistent Infection Strain in China. Front Vet Sci 2020; 7:571730. [PMID: 33335918 PMCID: PMC7736604 DOI: 10.3389/fvets.2020.571730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 11/02/2020] [Indexed: 11/13/2022] Open
Abstract
Murine Norovirus (MNV) is one of the most known viruses among viruses in mice. Because of the high prevalence of MNV in frequently used laboratory animals in biomedical researches, there is a significant impact of MNV. There may be different prevalence degrees and molecular characteristics of MNV in different regions around the world. Here, we reported an MNV strain "designated HBTS-1806" isolation from commercial mice's feces that caused a detectable cytopathic effect (CPE) in RAW264.7 cells. According to electron microscopy, the virus was 50-70 nm in diameter. The complete genome of HBTS-1806 is 7383 nucleotides with a structure similar to that of MNV reference strains. According to phylogenetic analysis on the basis of the whole genome, HBTS-1806 shared nucleotide sequence identities of 90.2-95.4% with other Chinese isolates reported. Analysis of amino acid sequence on the basis of ORF1 and ORF2 suggested that the isolated strain may be derived from recombination. Although no gross lesions or histopathological changes were found from mice infected with 5 × 105 TCLD50 of MNV by oral gavage inoculation, the intestinal virus loads lasted 12 weeks, suggesting a persistent infection strain of MNV isolate in China.
Collapse
Affiliation(s)
- Zhao Na
- The Experiment Animal Center, North China University of Science and Technology, Tangshan, China
| | - Jiang Bo
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agricultural and Forestry Sciences, Beijing, China
| | - Yang Yifei
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Cao Fuyuan
- The Experiment Animal Center, North China University of Science and Technology, Tangshan, China
| | - He Bin
- The Experiment Animal Center, North China University of Science and Technology, Tangshan, China
| | - Zhang Yanshu
- The Experiment Animal Center, North China University of Science and Technology, Tangshan, China
| | - Jin Huan
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agricultural and Forestry Sciences, Beijing, China
| | - Su Jingliang
- Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Li Shuang
- The Experiment Animal Center, North China University of Science and Technology, Tangshan, China
| |
Collapse
|
26
|
Kim H, Lim CY, Lee DB, Seok JH, Kim KH, Chung MS. Inhibitory Effects of Laminaria japonica Fucoidans Against Noroviruses. Viruses 2020; 12:E997. [PMID: 32906822 PMCID: PMC7552056 DOI: 10.3390/v12090997] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Revised: 08/28/2020] [Accepted: 09/03/2020] [Indexed: 12/18/2022] Open
Abstract
Norovirus is the leading cause of nonbacterial foodborne disease outbreaks. Human noroviruses (HuNoVs) bind to histo-blood group antigens as the host receptor for infection. In this study, the inhibitory effects of fucoidans from brown algae, Laminaria japonica (LJ), Undaria pinnatifida and Undaria pinnatifida sporophyll, were evaluated against murine norovirus (MNoV), feline calicivirus (FCV) and HuNoV. Pretreatment of MNoV or FCV with the fucoidans at 1 mg/mL showed high antiviral activities, with 1.1 average log reductions of viral titers in plaque assays. They also showed significant inhibition on the binding of the P domains of HuNoV GII.4 and GII.17 to A- or O-type saliva and the LJ fucoidan was the most effective, reaching 54-72% inhibition at 1 mg/mL. In STAT1-/- mice infected with MNoV, oral administration of the LJ fucoidan, composed of mainly sulfated fucose and minor amounts of glucose and galactose, improved the survival rates of mice and significantly reduced the viral titers in their feces. Overall, these results provide the LJ fucoidan can be used to reduce NoV outbreaks.
Collapse
Affiliation(s)
- Hyojin Kim
- Department of Food and Nutrition, Duksung Women’s University, Seoul 01369, Korea; (H.K.); (C.Y.L.)
| | - Chae Yeon Lim
- Department of Food and Nutrition, Duksung Women’s University, Seoul 01369, Korea; (H.K.); (C.Y.L.)
| | - Dan Bi Lee
- Department of Biotechnology and Bioinformatics, Korea University, Sejong 30019, Korea; (D.B.L.); (J.H.S.)
| | - Jong Hyeon Seok
- Department of Biotechnology and Bioinformatics, Korea University, Sejong 30019, Korea; (D.B.L.); (J.H.S.)
| | - Kyung Hyun Kim
- Department of Biotechnology and Bioinformatics, Korea University, Sejong 30019, Korea; (D.B.L.); (J.H.S.)
| | - Mi Sook Chung
- Department of Food and Nutrition, Duksung Women’s University, Seoul 01369, Korea; (H.K.); (C.Y.L.)
| |
Collapse
|
27
|
Roth AN, Helm EW, Mirabelli C, Kirsche E, Smith JC, Eurell LB, Ghosh S, Altan-Bonnet N, Wobus CE, Karst SM. Norovirus infection causes acute self-resolving diarrhea in wild-type neonatal mice. Nat Commun 2020; 11:2968. [PMID: 32528015 PMCID: PMC7289885 DOI: 10.1038/s41467-020-16798-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 05/25/2020] [Indexed: 01/08/2023] Open
Abstract
Human noroviruses are the leading cause of severe childhood diarrhea worldwide, yet we know little about their pathogenic mechanisms. Murine noroviruses cause diarrhea in interferon-deficient adult mice but these hosts also develop systemic pathology and lethality, reducing confidence in the translatability of findings to human norovirus disease. Herein we report that a murine norovirus causes self-resolving diarrhea in the absence of systemic disease in wild-type neonatal mice, thus mirroring the key features of human norovirus disease and representing a norovirus small animal disease model in wild-type mice. Intriguingly, lymphocytes are critical for controlling acute norovirus replication while simultaneously contributing to disease severity, likely reflecting their dual role as targets of viral infection and key components of the host response.
Collapse
Affiliation(s)
- Alexa N Roth
- 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
| | - Carmen Mirabelli
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI, USA
| | - Erin Kirsche
- Department of Molecular Genetics & Microbiology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Jonathan C Smith
- 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
| | - Sourish Ghosh
- Laboratory of Host-Pathogen Dynamics, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Nihal Altan-Bonnet
- Laboratory of Host-Pathogen Dynamics, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Christiane E Wobus
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI, USA
| | - Stephanie M Karst
- Department of Molecular Genetics & Microbiology, College of Medicine, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
28
|
Graziano VR, Walker FC, Kennedy EA, Wei J, Ettayebi K, Strine MS, Filler RB, Hassan E, Hsieh LL, Kim AS, Kolawole AO, Wobus CE, Lindesmith LC, Baric RS, Estes MK, Orchard RC, Baldridge MT, Wilen CB. CD300lf is the primary physiologic receptor of murine norovirus but not human norovirus. PLoS Pathog 2020; 16:e1008242. [PMID: 32251490 PMCID: PMC7162533 DOI: 10.1371/journal.ppat.1008242] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 04/16/2020] [Accepted: 03/03/2020] [Indexed: 12/18/2022] Open
Abstract
Murine norovirus (MNoV) is an important model of human norovirus (HNoV) and mucosal virus infection more broadly. Viral receptor utilization is a major determinant of cell tropism, host range, and pathogenesis. The bona fide receptor for HNoV is unknown. Recently, we identified CD300lf as a proteinaceous receptor for MNoV. Interestingly, its paralogue CD300ld was also sufficient for MNoV infection in vitro. Here we explored whether CD300lf is the sole physiologic receptor in vivo and whether HNoV can use a CD300 ortholog as an entry receptor. We report that both CD300ld and CD300lf are sufficient for infection by diverse MNoV strains in vitro. We further demonstrate that CD300lf is essential for both oral and parenteral MNoV infection and to elicit anti-MNoV humoral responses in vivo. In mice deficient in STAT1 signaling, CD300lf is required for MNoV-induced lethality. Finally, we demonstrate that human CD300lf (huCD300lf) is not essential for HNoV infection, nor does huCD300lf inhibit binding of HNoV virus-like particles to glycans. Thus, we report huCD300lf is not a receptor for HNoV.
Collapse
Affiliation(s)
- Vincent R. Graziano
- Departments of Laboratory Medicine and Immunobiology, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Forrest C. Walker
- Department of Medicine, Division of Infectious Diseases, Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine, Saint Louis, Missouri, United States of America
| | - Elizabeth A. Kennedy
- Department of Medicine, Division of Infectious Diseases, Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine, Saint Louis, Missouri, United States of America
| | - Jin Wei
- Departments of Laboratory Medicine and Immunobiology, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Khalil Ettayebi
- Departments of Medicine and Molecular Virology & Microbiology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Madison S. Strine
- Departments of Laboratory Medicine and Immunobiology, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Renata B. Filler
- Departments of Laboratory Medicine and Immunobiology, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Ebrahim Hassan
- Department of Medicine, Division of Infectious Diseases, Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine, Saint Louis, Missouri, United States of America
| | - Leon L. Hsieh
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Arthur S. Kim
- Department of Medicine, Division of Infectious Diseases, Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine, Saint Louis, Missouri, United States of America
| | - Abimbola O. Kolawole
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Christiane E. Wobus
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Lisa C. Lindesmith
- Department of Epidemiology, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Ralph S. Baric
- Department of Epidemiology, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Mary K. Estes
- Departments of Medicine and Molecular Virology & Microbiology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Robert C. Orchard
- Department of Immunology, University of Texas Southwestern Medical School, Dallas, Texas, United States of America
| | - 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, Missouri, United States of America
| | - Craig B. Wilen
- Departments of Laboratory Medicine and Immunobiology, Yale University School of Medicine, New Haven, Connecticut, United States of America
| |
Collapse
|
29
|
Hosmillo M, Chaudhry Y, Nayak K, Sorgeloos F, Koo BK, Merenda A, Lillestol R, Drumright L, Zilbauer M, Goodfellow I. Norovirus Replication in Human Intestinal Epithelial Cells Is Restricted by the Interferon-Induced JAK/STAT Signaling Pathway and RNA Polymerase II-Mediated Transcriptional Responses. mBio 2020; 11:e00215-20. [PMID: 32184238 PMCID: PMC7078467 DOI: 10.1128/mbio.00215-20] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 02/04/2020] [Indexed: 02/06/2023] Open
Abstract
Human noroviruses (HuNoV) are a leading cause of viral gastroenteritis worldwide and a significant cause of morbidity and mortality in all age groups. The recent finding that HuNoV can be propagated in B cells and mucosa-derived intestinal epithelial organoids (IEOs) has transformed our ability to dissect the life cycle of noroviruses. Using transcriptome sequencing (RNA-Seq) of HuNoV-infected intestinal epithelial cells (IECs), we have found that replication of HuNoV in IECs results in interferon (IFN)-induced transcriptional responses and that HuNoV replication in IECs is sensitive to IFN. This contrasts with previous studies that suggested that the innate immune response may play no role in the restriction of HuNoV replication in immortalized cells. We demonstrated that inhibition of Janus kinase 1 (JAK1)/JAK2 enhanced HuNoV replication in IECs. Surprisingly, targeted inhibition of cellular RNA polymerase II-mediated transcription was not detrimental to HuNoV replication but instead enhanced replication to a greater degree than blocking of JAK signaling directly. Furthermore, we demonstrated for the first time that IECs generated from genetically modified intestinal organoids, engineered to be deficient in the interferon response, were more permissive to HuNoV infection. Taking the results together, our work revealed that IFN-induced transcriptional responses restrict HuNoV replication in IECs and demonstrated that inhibition of these responses mediated by modifications of the culture conditions can greatly enhance the robustness of the norovirus culture system.IMPORTANCE Noroviruses are a major cause of gastroenteritis worldwide, and yet the challenges associated with their growth in culture have greatly hampered the development of therapeutic approaches and have limited our understanding of the cellular pathways that control infection. Here, we show that human intestinal epithelial cells, which represent the first point of entry of human noroviruses into the host, limit virus replication by induction of innate responses. Furthermore, we show that modulating the ability of intestinal epithelial cells to induce transcriptional responses to HuNoV infection can significantly enhance human norovirus replication in culture. Collectively, our findings provide new insights into the biological pathways that control norovirus infection but also identify mechanisms that enhance the robustness of norovirus culture.
Collapse
Affiliation(s)
- Myra Hosmillo
- Division of Virology, Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | - Yasmin Chaudhry
- Division of Virology, Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | - Komal Nayak
- Department of Paediatrics, University of Cambridge, Cambridge, United Kingdom
| | - Frederic Sorgeloos
- Division of Virology, Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | - Bon-Kyoung Koo
- Wellcome Trust-Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna Biocenter (VBC), Vienna, Austria
| | - Alessandra Merenda
- Wellcome Trust-Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
| | - Reidun Lillestol
- Department of Medicine, Addenbrooke's Hospital, University of Cambridge, Cambridge, United Kingdom
| | - Lydia Drumright
- Department of Medicine, Addenbrooke's Hospital, University of Cambridge, Cambridge, United Kingdom
| | - Matthias Zilbauer
- Department of Paediatrics, University of Cambridge, Cambridge, United Kingdom
| | - Ian Goodfellow
- Division of Virology, Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
30
|
Hsu CC, Patil K, Seamons A, Brabb TL, Treuting PM, Paik J, Meeker SM, Maggio-Price L. Lack of Effect of Murine Norovirus Infection on the CD4 + CD45RB high T-cell Adoptive Transfer Mouse Model of Inflammatory Bowel Disease. Comp Med 2020; 70:16-24. [PMID: 31937392 PMCID: PMC7024779 DOI: 10.30802/aalas-cm-19-000009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 02/24/2019] [Accepted: 04/22/2019] [Indexed: 11/05/2022]
Abstract
Murine norovirus (MNV) infection is highly prevalent in laboratory mice. Although MNV infection does not typically induce clinical disease in most laboratory mice, infection may nonetheless affect mouse models of disease by altering immune responses. We previously reported that MNV altered the bacterial-induced mouse model of inflammatory bowel disease (IBD) using Helicobacter-infected Mdr1a-/- mice. Therefore, we hypothesized that MNV infection would exacerbate another mouse model of IBD, the T-cell adoptive transfer (AT) model. In this model, Helicobacter infection is used to accelerate the progression of IBD induced by AT of naïve CD4+CD45RBhigh T cells into B6.129S7- Rag1tm1Mom/J (Rag1-/-) mice. We evaluated the effects of MNV infection in both Helicobacter-accelerated as well as Helicobacter-free AT models. In our studies, Helicobacter-infected Rag1-/- mice that received CD4+CD45RBhigh T cells through AT rapidly developed weight loss and typhlocolitis; MNV infection had no effect on disease severity or rate of progression. In the absence of Helicobacter infection, progression of IBD caused by AT of CD4+CD45RBhigh T cells was slower and typhlocolitis was less severe; this inflammation likewise was unaltered by MNV infection. These results indicate that MNV infection does not alter IBD progression and severity in the CD4+CD45RBhigh T-cell AT model in Rag1-/- mice.
Collapse
Affiliation(s)
- Charlie C Hsu
- Department of Comparative Medicine, School of Medicine, University of Washington, Seattle, Washington;,
| | | | - Audrey Seamons
- Department of Comparative Medicine, School of Medicine, University of Washington, Seattle, Washington
| | - Thea L Brabb
- Department of Comparative Medicine, School of Medicine, University of Washington, Seattle, Washington
| | - Piper M Treuting
- Department of Comparative Medicine, School of Medicine, University of Washington, Seattle, Washington
| | - Jisun Paik
- Department of Comparative Medicine, School of Medicine, University of Washington, Seattle, Washington
| | - Stacey M Meeker
- University Laboratory Animal Resources, Department of Veterinary Preventive Medicine, Ohio State University, Columbus, Ohio
| | - Lillian Maggio-Price
- Department of Comparative Medicine, School of Medicine, University of Washington, Seattle, Washington
| |
Collapse
|
31
|
Radaelli E, Santagostino SF, Sellers RS, Brayton CF. Immune Relevant and Immune Deficient Mice: Options and Opportunities in Translational Research. ILAR J 2019; 59:211-246. [PMID: 31197363 PMCID: PMC7114723 DOI: 10.1093/ilar/ily026] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 12/03/2018] [Indexed: 12/29/2022] Open
Abstract
In 1989 ILAR published a list and description of immunodeficient rodents used in research. Since then, advances in understanding of molecular mechanisms; recognition of genetic, epigenetic microbial, and other influences on immunity; and capabilities in manipulating genomes and microbiomes have increased options and opportunities for selecting mice and designing studies to answer important mechanistic and therapeutic questions. Despite numerous scientific breakthroughs that have benefitted from research in mice, there is debate about the relevance and predictive or translational value of research in mice. Reproducibility of results obtained from mice and other research models also is a well-publicized concern. This review summarizes resources to inform the selection and use of immune relevant mouse strains and stocks, aiming to improve the utility, validity, and reproducibility of research in mice. Immune sufficient genetic variations, immune relevant spontaneous mutations, immunodeficient and autoimmune phenotypes, and selected induced conditions are emphasized.
Collapse
Affiliation(s)
- Enrico Radaelli
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Sara F Santagostino
- Department of Safety Assessment, Genentech, Inc., South San Francisco, California
| | | | - Cory F Brayton
- Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
32
|
Peñaflor-Téllez Y, Trujillo-Uscanga A, Escobar-Almazán JA, Gutiérrez-Escolano AL. Immune Response Modulation by Caliciviruses. Front Immunol 2019; 10:2334. [PMID: 31632406 PMCID: PMC6779827 DOI: 10.3389/fimmu.2019.02334] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 09/16/2019] [Indexed: 12/29/2022] Open
Abstract
Noroviruses and Sapoviruses, classified in the Caliciviridae family, are small positive-stranded RNA viruses, considered nowadays the leading cause of acute gastroenteritis globally in both children and adults. Although most noroviruses have been associated with gastrointestinal disease in humans, almost 50 years after its discovery, there is still a lack of comprehensive evidence regarding its biology and pathogenesis mainly because they can be neither conveniently grown in cultured cells nor propagated in animal models. However, other members of this family such as Feline calicivirus (FCV), Murine norovirus (MNV), Rabbit hemorrhagic disease virus (RHDV), and Porcine sapovirus (PS), from which there are accessible propagation systems, have been useful to study the calicivirus replication strategies. Using cell cultures and animal models, many of the functions of the viral proteins in the viral replication cycles have been well-characterized. Moreover, evidence of the role of viral proteins from different members of the family in the establishment of infection has been generated and the mechanism of their immunopathogenesis begins to be understood. In this review, we discuss different aspects of how caliciviruses are implicated in membrane rearrangements, apoptosis, and evasion of the immune responses, highlighting some of the pathogenic mechanisms triggered by different members of the Caliciviridae family.
Collapse
Affiliation(s)
- Yoatzin Peñaflor-Téllez
- Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados, IPN, Mexico City, Mexico
| | - Adrian Trujillo-Uscanga
- Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados, IPN, Mexico City, Mexico
| | - Jesús Alejandro Escobar-Almazán
- Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados, IPN, Mexico City, Mexico
| | - Ana Lorena Gutiérrez-Escolano
- Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados, IPN, Mexico City, Mexico
| |
Collapse
|
33
|
Walker FC, Baldridge MT. Interactions between noroviruses, the host, and the microbiota. Curr Opin Virol 2019; 37:1-9. [PMID: 31096124 PMCID: PMC6768699 DOI: 10.1016/j.coviro.2019.04.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Revised: 04/11/2019] [Accepted: 04/12/2019] [Indexed: 12/25/2022]
Abstract
In recent years, appreciation has been growing for the role that the microbiota plays in interactions between the host and various pathogens, including norovirus. Proviral and antiviral effects of the microbiota have been observed for both human and murine noroviruses, and it has become clear that direct effects of microbes and their metabolites as well as indirect effects of commensals on the host are key in modulating pathogenesis. In particular, a common thread has emerged in the ability of members of the microbiota to regulate the host interferon response, thereby modulating norovirus infection. Here, we highlight key differences between human and murine noroviruses and their interactions with the microbiota, while also underscoring shared characteristics between noroviruses and other gastrointestinal viruses.
Collapse
Affiliation(s)
- Forrest C Walker
- Division of Infectious Diseases, Department of Medicine, Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine, St. Louis, MO, 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.
| |
Collapse
|
34
|
Bolsega S, Basic M, Smoczek A, Buettner M, Eberl C, Ahrens D, Odum KA, Stecher B, Bleich A. Composition of the Intestinal Microbiota Determines the Outcome of Virus-Triggered Colitis in Mice. Front Immunol 2019; 10:1708. [PMID: 31396223 PMCID: PMC6664081 DOI: 10.3389/fimmu.2019.01708] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 07/08/2019] [Indexed: 12/14/2022] Open
Abstract
The intestinal microbiota is a complex ecosystem implicated in host health and disease. Inflammatory bowel disease (IBD) is a multifactorial chronic disorder of the gastrointestinal mucosa. Even though the exact mechanisms are still unknown, the intestinal microbiota is crucial in IBD development. We previously showed that murine norovirus (MNV) induces colitis in the Il10-deficient (Il10−/−) mouse model of IBD in a microbiota-dependent manner. Thus, in this study we analyzed whether distinct minimal bacterial consortia influence the outcome of MNV-triggered colitis in Il10−/− mice. Gnotobiotic Il10−/− mice associated with Oligo-Mouse-Microbiota 12 (OMM12) or Altered Schaedler Flora (ASF) developed little to no inflammatory lesions in the colon and cecum. MNV infection exacerbated colitis severity only in ASF-colonized mice, but not in those associated with OMM12. Four weeks after MNV infection, inflammatory lesions in ASF-colonized Il10−/− mice were characterized by epithelial hyperplasia, infiltration of inflammatory cells, and increased barrier permeability. Co-colonization of ASF-colonized Il10−/− mice with segmented filamentous bacteria (SFB) abolished MNV-induced colitis, whereas histopathological scores in SFB-OMM12-co-colonized mice stayed unchanged. Moreover, SFB only colonized mice associated with ASF. The SFB-mediated protective effects in ASF-colonized mice involved enhanced activation of intestinal barrier defense mechanisms and mucosal immune responses in the chronic and acute phase of MNV infection. SFB colonization strengthened intestinal barrier function by increasing expression of tight junction proteins, antimicrobial peptides and mucus. Furthermore, SFB colonization enhanced the expression of pro-inflammatory cytokines such as Tnfα, Il1β, and Il12a, as well as the expression of the regulatory cytokine Tgfβ. Altogether, our results showed that MNV-triggered colitis depends on the microbial context.
Collapse
Affiliation(s)
- Silvia Bolsega
- Hannover Medical School, Institute for Laboratory Animal Science, Hanover, Germany
| | - Marijana Basic
- Hannover Medical School, Institute for Laboratory Animal Science, Hanover, Germany
| | - Anna Smoczek
- Hannover Medical School, Institute for Laboratory Animal Science, Hanover, Germany
| | - Manuela Buettner
- Hannover Medical School, Institute for Laboratory Animal Science, Hanover, Germany
| | - Claudia Eberl
- Faculty of Medicine, Max von Pettenkofer Institute of Hygiene and Medical Microbiology, LMU Munich, Munich, Germany
| | - Daniel Ahrens
- Hannover Medical School, Institute for Laboratory Animal Science, Hanover, Germany
| | - Kodwo Appoh Odum
- Hannover Medical School, Institute for Laboratory Animal Science, Hanover, Germany
| | - Bärbel Stecher
- Faculty of Medicine, Max von Pettenkofer Institute of Hygiene and Medical Microbiology, LMU Munich, Munich, Germany.,German Center of Infection Research (DZIF), Partner Site Munich, Munich, Germany
| | - Andre Bleich
- Hannover Medical School, Institute for Laboratory Animal Science, Hanover, Germany
| |
Collapse
|
35
|
Robinson BA, Van Winkle JA, McCune BT, Peters AM, Nice TJ. Caspase-mediated cleavage of murine norovirus NS1/2 potentiates apoptosis and is required for persistent infection of intestinal epithelial cells. PLoS Pathog 2019; 15:e1007940. [PMID: 31329638 PMCID: PMC6675124 DOI: 10.1371/journal.ppat.1007940] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 08/01/2019] [Accepted: 06/24/2019] [Indexed: 12/05/2022] Open
Abstract
Human norovirus (HNoV) is the leading cause of acute gastroenteritis and is spread by fecal shedding that can often persist for weeks to months after the resolution of symptoms. Elimination of persistent viral reservoirs has the potential to prevent outbreaks. Similar to HNoV, murine norovirus (MNV) is spread by persistent shedding in the feces and provides a tractable model to study molecular mechanisms of enteric persistence. Previous studies have identified non-structural protein 1 (NS1) from the persistent MNV strain CR6 as critical for persistent infection in intestinal epithelial cells (IECs), but its mechanism of action remains unclear. We now find that the function of CR6 NS1 is regulated by apoptotic caspase cleavage. Following induction of apoptosis in infected cells, caspases cleave the precursor NS1/2 protein, and this cleavage is prevented by mutation of caspase target motifs. These mutations profoundly compromise CR6 infection of IECs and persistence in the intestine. Conversely, NS1/2 cleavage is not strictly required for acute replication in extra-intestinal tissues or in cultured myeloid cells, suggesting an IEC-centric role. Intriguingly, we find that caspase cleavage of CR6 NS1/2 reciprocally promotes caspase activity, potentiates cell death, and amplifies spread among cultured IEC monolayers. Together, these data indicate that the function of CR6 NS1 is regulated by apoptotic caspases, and suggest that apoptotic cell death enables epithelial spread and persistent shedding. Human Norovirus infection is highly contagious and the most common cause of acute gastroenteritis. Norovirus can be persistently shed after resolution of symptoms, perpetuating or initiating new outbreaks. Murine norovirus (MNV) is also persistently shed, enabling study of host and viral determinants of norovirus pathogenesis. We previously identified a critical role for MNV non-structural protein 1 (NS1), in persistence. Herein we find that regulation of NS1 by host apoptotic caspases is required for infection of intestinal epithelial cells, but not for extra-intestinal spread. Additionally, we demonstrate that NS1 reciprocally promotes cell death and spread among epithelial cells. These data identify regulation of NS1 by host proteases and suggest that apoptotic death is a determinant of epithelial spread and persistence.
Collapse
Affiliation(s)
- Bridget A. Robinson
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, Oregon, United States of America
| | - Jacob A. Van Winkle
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, Oregon, United States of America
| | - Broc T. McCune
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX, United States of America
| | - A. Mack Peters
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, Oregon, United States of America
| | - Timothy J. Nice
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, Oregon, United States of America
- * E-mail:
| |
Collapse
|
36
|
Cortez V, Sharp B, Yao J, Livingston B, Vogel P, Schultz-Cherry S. Characterizing a Murine Model for Astrovirus Using Viral Isolates from Persistently Infected Immunocompromised Mice. J Virol 2019; 93:e00223-19. [PMID: 30971471 PMCID: PMC6580942 DOI: 10.1128/jvi.00223-19] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 03/26/2019] [Indexed: 12/11/2022] Open
Abstract
Human astroviruses are single-stranded RNA enteric viruses that cause a spectrum of disease ranging from asymptomatic infection to systemic extragastrointestinal spread; however, they are among the least-characterized enteric viruses, and there is a lack of a well-characterized small animal model. Finding that immunocompromised mice were resistant to human astrovirus infection via multiple routes of inoculation, our studies aimed to determine whether murine astrovirus (MuAstV) could be used to model human astrovirus disease. We experimentally infected wild-type mice with MuAstV isolated from immunocompromised mice and found that the virus was detected throughout the gastrointestinal tract, including the stomach, but was not associated with diarrhea. The virus was also detected in the lung. Although virus levels were higher in recently weaned mice, the levels were similar in male and female adult mice. Using two distinct viruses isolated from different immunocompromised mouse strains, we observed virus strain-specific differences in the duration of infection (3 versus 10 weeks) in wild-type mice, indicating that the within-host immune pressure from donor mice shaped the virus kinetics in immunocompetent recipient hosts. Both virus strains elicited minimal pathology and a lack of sustained immunity. In summary, MuAstV represents a useful model for studying asymptomatic human infection and gaining insight into the astrovirus pathogenesis and immunity.IMPORTANCE Astroviruses are widespread in both birds and mammals; however, little is known about the pathogenesis and the immune response to the virus due to the lack of a well-characterized small-animal model. Here we describe two distinct strains of murine astrovirus that cause infections in immunocompetent mice that mirror aspects of asymptomatic human infections, including minimal pathology and short-lived immunity. However, we noted that the duration of infection differed greatly between the strains, highlighting an important facet of these viruses that was not previously appreciated. The ubiquitous nature and diversity of murine astroviruses coupled with the continuous likelihood of reinfection raise the possibility of viral interference with other mouse models of disease.
Collapse
Affiliation(s)
- Valerie Cortez
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Bridgett Sharp
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Jiangwei Yao
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Brandi Livingston
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Peter Vogel
- Veterinary Pathology Core, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Stacey Schultz-Cherry
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| |
Collapse
|
37
|
Bhar S, Jones MK. In Vitro Replication of Human Norovirus. Viruses 2019; 11:v11060547. [PMID: 31212759 PMCID: PMC6630950 DOI: 10.3390/v11060547] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 06/06/2019] [Accepted: 06/10/2019] [Indexed: 12/18/2022] Open
Abstract
Human norovirus (HuNoV) infection is a major cause of gastroenteritis all over the world. Despite this, these non-enveloped RNA viruses are poorly characterized due to the lack of robust and widely available HuNoV culture systems. The two published systems (B cell line and stem cell-derived enteroids) support replication of HuNoVs but the levels of replication are not sufficient for the generation of highly purified virus stocks or the development of culture-based quantification assays. Therefore, improvement of HuNoV in vitro replication is still needed. Murine norovirus and other caliciviruses have provided insights into norovirus replication that paved the way for the development of the current HuNoV culture systems and may also aid in the improvement of these systems. This review will highlight ways in which previous research guided and impacted the development of HuNoV culture systems and discuss ways in which more recent discoveries might be utilized to improve the quality of the HuNoV in vitro replication.
Collapse
Affiliation(s)
- Sutonuka Bhar
- Microbiology and Cell Science Department, University of Florida, Gainesville, FL 32611, USA.
| | - Melissa K Jones
- Microbiology and Cell Science Department, University of Florida, Gainesville, FL 32611, USA.
| |
Collapse
|
38
|
The Antigenic Topology of Norovirus as Defined by B and T Cell Epitope Mapping: Implications for Universal Vaccines and Therapeutics. Viruses 2019; 11:v11050432. [PMID: 31083353 PMCID: PMC6563215 DOI: 10.3390/v11050432] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 05/07/2019] [Accepted: 05/08/2019] [Indexed: 12/11/2022] Open
Abstract
Human norovirus (HuNoV) is the leading cause of acute nonbacterial gastroenteritis. Vaccine design has been confounded by the antigenic diversity of these viruses and a limited understanding of protective immunity. We reviewed 77 articles published since 1988 describing the isolation, function, and mapping of 307 unique monoclonal antibodies directed against B cell epitopes of human and murine noroviruses representing diverse Genogroups (G). Of these antibodies, 91, 153, 21, and 42 were reported as GI-specific, GII-specific, MNV GV-specific, and G cross-reactive, respectively. Our goal was to reconstruct the antigenic topology of noroviruses in relationship to mapped epitopes with potential for therapeutic use or inclusion in universal vaccines. Furthermore, we reviewed seven published studies of norovirus T cell epitopes that identified 18 unique peptide sequences with CD4- or CD8-stimulating activity. Both the protruding (P) and shell (S) domains of the major capsid protein VP1 contained B and T cell epitopes, with the majority of neutralizing and HBGA-blocking B cell epitopes mapping in or proximal to the surface-exposed P2 region of the P domain. The majority of broadly reactive B and T cell epitopes mapped to the S and P1 arm of the P domain. Taken together, this atlas of mapped B and T cell epitopes offers insight into the promises and challenges of designing universal vaccines and immunotherapy for the noroviruses.
Collapse
|
39
|
Nlrp3 inflammasome activation and Gasdermin D-driven pyroptosis are immunopathogenic upon gastrointestinal norovirus infection. PLoS Pathog 2019; 15:e1007709. [PMID: 31017981 PMCID: PMC6502405 DOI: 10.1371/journal.ppat.1007709] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 05/06/2019] [Accepted: 03/15/2019] [Indexed: 12/22/2022] Open
Abstract
Norovirus infection is the leading cause of food-borne gastroenteritis worldwide, being responsible for over 200,000 deaths annually. Studies with murine norovirus (MNV) showed that protective STAT1 signaling controls viral replication and pathogenesis, but the immune mechanisms that noroviruses exploit to induce pathology are elusive. Here, we show that gastrointestinal MNV infection leads to widespread IL-1β maturation in MNV-susceptible STAT1-deficient mice. MNV activates the canonical Nlrp3 inflammasome in macrophages, leading to maturation of IL-1β and to Gasdermin D (GSDMD)-dependent pyroptosis. STAT1-deficient macrophages displayed increased MAVS-mediated expression of pro-IL-1β, facilitating elevated Nlrp3-dependent release of mature IL-1β upon MNV infection. Accordingly, MNV-infected Stat1-/- mice showed Nlrp3-dependent maturation of IL-1β as well as Nlrp3-dependent pyroptosis as assessed by in vivo cleavage of GSDMD to its active N-terminal fragment. While MNV-induced diarrheic responses were not affected, Stat1-/- mice additionally lacking either Nlrp3 or GSDMD displayed lower levels of the fecal inflammatory marker Lipocalin-2 as well as delayed lethality after gastrointestinal MNV infection. Together, these results uncover new insights into the mechanisms of norovirus-induced inflammation and cell death, thereby revealing Nlrp3 inflammasome activation and ensuing GSDMD-driven pyroptosis as contributors to MNV-induced immunopathology in susceptible STAT1-deficient mice. Gastrointestinal norovirus infections form a serious socio-economic worldwide problem, with about 684 million cases annually worldwide and mortality occurring both in underdeveloped countries and in immunocompromised patients. Despite the urgent global need, no specific treatments are available for norovirus induced gastroenteritis, partly because innate immune responses upon gastrointestinal norovirus infection are largely unresolved. Using the murine norovirus (MNV) model we showed that MNV infection in macrophages leads to a lytic form of cell death termed pyroptosis as well as to the maturation and release of the pro-inflammatory cytokine IL-1β. Maturation of IL-1β was observed also in vivo, after gastrointestinal infection of MNV-susceptible Stat1 knockout mice. We found that these innate immune responses upon MNV infection crucially depended on activation of the Nlrp3 inflammasome leading to Gasdermin D-driven pyroptosis, and inactivating this signaling pathway delayed lethality of MNV-susceptible STAT1 knockout mice after gastrointestinal MNV infection. We thus identified Nlrp3 inflammasome activation and ensuing pyroptosis as a signaling pathway contributing to norovirus-induced immunopathology. Taken together, this study resulted in a more detailed understanding of MNV-induced inflammatory and cell death pathways and provided insights into how gastrointestinal viruses induce immunopathology.
Collapse
|
40
|
de Souza WM, Fumagalli MJ, de Araujo J, Ometto T, Modha S, Thomazelli LM, Durigon EL, Murcia PR, Figueiredo LTM. Discovery of novel astrovirus and calicivirus identified in ruddy turnstones in Brazil. Sci Rep 2019; 9:5556. [PMID: 30944402 PMCID: PMC6447618 DOI: 10.1038/s41598-019-42110-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 03/25/2019] [Indexed: 01/06/2023] Open
Abstract
Birds are the natural reservoir of viruses with zoonotic potential, as well as contributing to the evolution, emergence, and dissemination of novel viruses. In this study, we applied a high-throughput screening approach to identify the diversity of viruses in 118 samples of birds captured between October 2006 to October 2010 in the North and Northeast regions of Brazil. We found nearly complete genomes of novel species of astrovirus and calicivirus in cloacal swabs of ruddy turnstones (Arenaria interpres) collected in Coroa do Avião islet, Pernambuco State. These viruses are positive-sense single-stranded RNA with a genome of ~7 to 8 kb, and were designated as Ruddy turnstone astrovirus (RtAstV) and Ruddy turnstone calicivirus (RTCV), respectively. Phylogenetic analysis showed that RtAstV and RTCV grouped in a monophyletic clade with viruses identified from poultry samples (i.e., chicken, goose, and turkey), including viruses associated with acute nephritis in chickens. Attempts of viral propagation in monkey and chicken cell lines for both viruses were unsuccessful. Also, we found genomes related with viral families that infect invertebrates and plants, suggesting that they might be ingested in the birds' diet. In sum, these findings shed new light on the diversity of viruses in migratory birds with the notable characterization of a novel astrovirus and calicivirus.
Collapse
Affiliation(s)
- William Marciel de Souza
- Virology Research Center, School of Medicine of Ribeirão Preto of University of São Paulo, Ribeirão Preto, 14049-900, SP, Brazil.
- MRC-University of Glasgow Centre for Virus Research, Glasgow, G61 1QH, Scotland, United Kingdom.
- Department of Genetics, Evolution and Bioagents, Institute of Biology, University of Campinas, 13083-862, Campinas, São Paulo, Brazil.
| | - Marcílio Jorge Fumagalli
- Virology Research Center, School of Medicine of Ribeirão Preto of University of São Paulo, Ribeirão Preto, 14049-900, SP, Brazil
| | - Jansen de Araujo
- Institute of Biomedical Sciences, University of São Paulo, São Paulo, 05508-900, SP, Brazil
| | - Tatiana Ometto
- Institute of Biomedical Sciences, University of São Paulo, São Paulo, 05508-900, SP, Brazil
| | - Sejal Modha
- MRC-University of Glasgow Centre for Virus Research, Glasgow, G61 1QH, Scotland, United Kingdom
| | | | - Edison Luís Durigon
- Institute of Biomedical Sciences, University of São Paulo, São Paulo, 05508-900, SP, Brazil
| | - Pablo Ramiro Murcia
- MRC-University of Glasgow Centre for Virus Research, Glasgow, G61 1QH, Scotland, United Kingdom
| | - Luiz Tadeu Moraes Figueiredo
- Virology Research Center, School of Medicine of Ribeirão Preto of University of São Paulo, Ribeirão Preto, 14049-900, SP, Brazil
| |
Collapse
|
41
|
Natural Secretory Immunoglobulins Promote Enteric Viral Infections. J Virol 2018; 92:JVI.00826-18. [PMID: 30232191 DOI: 10.1128/jvi.00826-18] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 09/05/2018] [Indexed: 12/27/2022] Open
Abstract
Noroviruses are enteric pathogens causing significant morbidity, mortality, and economic losses worldwide. Secretory immunoglobulins (sIg) are a first line of mucosal defense against enteric pathogens. They are secreted into the intestinal lumen via the polymeric immunoglobulin receptor (pIgR), where they bind to antigens. However, whether natural sIg protect against norovirus infection remains unknown. To determine if natural sIg alter murine norovirus (MNV) pathogenesis, we infected pIgR knockout (KO) mice, which lack sIg in mucosal secretions. Acute MNV infection was significantly reduced in pIgR KO mice compared to controls, despite increased MNV target cells in the Peyer's patch. Natural sIg did not alter MNV binding to the follicle-associated epithelium (FAE) or crossing of the FAE into the lymphoid follicle. Instead, naive pIgR KO mice had enhanced levels of the antiviral inflammatory molecules interferon gamma (IFN-γ) and inducible nitric oxide synthase (iNOS) in the ileum compared to controls. Strikingly, depletion of the intestinal microbiota in pIgR KO and control mice resulted in comparable IFN-γ and iNOS levels, as well as MNV infectious titers. IFN-γ treatment of wild-type (WT) mice and neutralization of IFN-γ in pIgR KO mice modulated MNV titers, implicating the antiviral cytokine in the phenotype. Reduced gastrointestinal infection in pIgR KO mice was also observed with another enteric virus, reovirus. Collectively, our findings suggest that natural sIg are not protective during enteric virus infection, but rather, that sIg promote enteric viral infection through alterations in microbial immune responses.IMPORTANCE Enteric virus, such as norovirus, infections cause significant morbidity and mortality worldwide. However, direct antiviral infection prevention strategies are limited. Blocking host entry and initiation of infection provides an established avenue for intervention. Here, we investigated the role of the polymeric immunoglobulin receptor (pIgR)-secretory immunoglobulin (sIg) cycle during enteric virus infections. The innate immune functions of sIg (agglutination, immune exclusion, neutralization, and expulsion) were not required during control of acute murine norovirus (MNV) infection. Instead, lack of pIgR resulted in increased IFN-γ levels, which contributed to reduced MNV titers. Another enteric virus, reovirus, also showed decreased infection in pIgR KO mice. Collectively, our data point to a model in which sIg-mediated microbial sensing promotes norovirus and reovirus infection. These data provide the first evidence of the proviral role of natural sIg during enteric virus infections and provide another example of how intestinal bacterial communities indirectly influence MNV pathogenesis.
Collapse
|
42
|
Antiviral activity of Schizonepeta tenuifolia Briquet against noroviruses via induction of antiviral interferons. J Microbiol 2018; 56:683-689. [DOI: 10.1007/s12275-018-8228-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 05/11/2018] [Accepted: 05/21/2018] [Indexed: 01/02/2023]
|
43
|
Lee W, Kim M, Lee SH, Jung HG, Oh JW. Prophylactic efficacy of orally administered Bacillus poly-γ-glutamic acid, a non-LPS TLR4 ligand, against norovirus infection in mice. Sci Rep 2018; 8:8667. [PMID: 29875467 PMCID: PMC5989232 DOI: 10.1038/s41598-018-26935-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 01/17/2018] [Indexed: 12/22/2022] Open
Abstract
Poly-gamma-glutamic acid (γ-PGA), an extracellular biopolymer produced by Bacillus sp., is a non-canonical toll-like receptor 4 (TLR4) agonist. Here we show its antiviral efficacy against noroviruses. γ-PGA with a molecular mass of 2,000-kDa limited murine norovirus (MNV) replication in the macrophage cell line RAW264.7 by inducing interferon (IFN)-β and conferred resistance to viral infection-induced cell death. Additionally, γ-PGA interfered with viral entry into cells. The potent antiviral state mounted by γ-PGA was not attributed to the upregulation of TLR4 or TLR3, a sensor known to recognize norovirus RNA. γ-PGA sensing by TLR4 required the two TLR4-associated accessory factors MD2 and CD14. In ex vivo cultures of mouse ileum, γ-PGA selectively increased the expression of IFN-β in villi. In contrast, IFN-β induction was negligible in the ileal Peyer’s patches (PPs) where its expression was primarily induced by the replication of MNV. Oral administration of γ-PGA, which increased serum IFN-β levels without inducing proinflammatory cytokines, reduced MNV loads in the ileum with PPs and mesenteric lymph nodes in mice. Our results disclose a γ-PGA-mediated non-conventional TLR4 signaling in the ileum, highlighting the potential use of γ-PGA as a prophylactic antiviral agent against noroviruses.
Collapse
Affiliation(s)
- Wooseong Lee
- Department of Biotechnology, Yonsei University, Seoul, 03722, Korea
| | - Minwoo Kim
- Department of Biotechnology, Yonsei University, Seoul, 03722, Korea
| | - Seung-Hoon Lee
- Department of Biotechnology, Yonsei University, Seoul, 03722, Korea
| | - Hae-Gwang Jung
- Department of Biotechnology, Yonsei University, Seoul, 03722, Korea
| | - Jong-Won Oh
- Department of Biotechnology, Yonsei University, Seoul, 03722, Korea.
| |
Collapse
|
44
|
Thorne L, Lu J, Chaudhry Y, Bailey D, Goodfellow I. Targeting macrophage- and intestinal epithelial cell-specific microRNAs against norovirus restricts replication in vivo. J Gen Virol 2018; 99:1621-1632. [PMID: 29683421 DOI: 10.1099/jgv.0.001065] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Until recently, our understanding of the cellular tropism of human norovirus (HuNoV), a major cause of viral gastroenteritis, has been limited. Immune cells and intestinal epithelial cells (IECs) have been proposed as targets of HuNoV replication in vivo, although the contribution of each to pathogenesis and transmission is unknown. Murine norovirus (MNV) is widely used as a surrogate model for HuNoV, as it replicates in cultured immune cells. The importance of the complete MNV immune cell tropism in vivo has not been determined. Recent work has linked replication in IECs to viral persistence in vivo. MNV provides a model to assess the relative contribution of each cell tropism to viral replication in immunocompetent native hosts. Here we exploited cell-specific microRNAs to control MNV replication, through insertion of microRNA target sequences into the MNV genome. We demonstrated the utility of this approach for MNV in vitro by selectively reducing replication in microglial cells, using microglial-specific miR-467c. We then showed that inserting a target sequence for the haematopoietic-specific miR-142-3p abrogated replication in a macrophage cell line. The presence of a target sequence for either miR-142-3p or IEC miR-215 significantly reduced viral secretion during the early stages of a persistent infection in immunocompetent mice, confirming that both cell types support viral replication in vivo. This study provides additional evidence that MNV shares the IEC tropism of HuNoVs in vivo, and now provides a model to dissect the contribution of replication in each cell type to viral pathogenesis and transmission in a native host.
Collapse
Affiliation(s)
- Lucy Thorne
- 2Division of Infection and Immunity, University College London, Medical Research Council, London, UK.,1Division of Virology, Department of Pathology, University of Cambridge, Cambridge, UK
| | - Jia Lu
- 1Division of Virology, Department of Pathology, University of Cambridge, Cambridge, UK
| | - Yasmin Chaudhry
- 1Division of Virology, Department of Pathology, University of Cambridge, Cambridge, UK
| | - Dalan Bailey
- 3The Pirbright Institute, Pirbright, Woking, UK
| | - Ian Goodfellow
- 1Division of Virology, Department of Pathology, University of Cambridge, Cambridge, UK
| |
Collapse
|
45
|
Hsu CC, Meeker SM, Escobar S, Brabb TL, Paik J, Park H, Iritani BM, Maggio-Price L. Murine norovirus inhibits B cell development in the bone marrow of STAT1-deficient mice. Virology 2017; 515:123-133. [PMID: 29287229 PMCID: PMC5801037 DOI: 10.1016/j.virol.2017.12.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 12/12/2017] [Accepted: 12/13/2017] [Indexed: 12/13/2022]
Abstract
Noroviruses are a leading cause of gastroenteritis in humans and it was recently revealed that noroviruses can infect B cells. We demonstrate that murine norovirus (MNV) infection can significantly impair B cell development in the bone marrow in a signal transducer and activator of transcription 1 (STAT1) dependent, but interferon signaling independent manner. We also show that MNV replication is more pronounced in the absence of STAT1 in ex vivo cultured B cells. Interestingly, using bone marrow transplantation studies, we found that impaired B cell development requires Stat1-/- hematopoietic cells and Stat1-/- stromal cells, and that the presence of wild-type hematopoietic or stromal cells was sufficient to restore normal development of Stat1-/- B cells. These results suggest that B cells normally restrain norovirus replication in a cell autonomous manner, and that wild-type STAT1 is required to protect B cell development during infection.
Collapse
Affiliation(s)
- Charlie C Hsu
- Department of Comparative Medicine, University of Washington, Seattle, WA, United States
| | - Stacey M Meeker
- Department of Comparative Medicine, University of Washington, Seattle, WA, United States
| | - Sabine Escobar
- Department of Comparative Medicine, University of Washington, Seattle, WA, United States
| | - Thea L Brabb
- Department of Comparative Medicine, University of Washington, Seattle, WA, United States
| | - Jisun Paik
- Department of Comparative Medicine, University of Washington, Seattle, WA, United States
| | - Heon Park
- Department of Comparative Medicine, University of Washington, Seattle, WA, United States
| | - Brian M Iritani
- Department of Comparative Medicine, University of Washington, Seattle, WA, United States
| | - Lillian Maggio-Price
- Department of Comparative Medicine, University of Washington, Seattle, WA, United States
| |
Collapse
|
46
|
Grau KR, Roth AN, Zhu S, Hernandez A, Colliou N, DiVita BB, Philip DT, Riffe C, Giasson B, Wallet SM, Mohamadzadeh M, Karst SM. The major targets of acute norovirus infection are immune cells in the gut-associated lymphoid tissue. Nat Microbiol 2017; 2:1586-1591. [PMID: 29109476 PMCID: PMC5705318 DOI: 10.1038/s41564-017-0057-7] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 10/09/2017] [Indexed: 11/16/2022]
Abstract
Noroviruses are the leading cause of food-borne gastroenteritis outbreaks and childhood diarrhoea globally, estimated to be responsible for 200,000 deaths in children each year 1-4 . Thus, reducing norovirus-associated disease is a critical priority. Development of vaccines and therapeutics has been hindered by the limited understanding of basic norovirus pathogenesis and cell tropism. While macrophages, dendritic cells, B cells and stem-cell-derived enteroids can all support infection of certain noroviruses in vitro 5-7 , efforts to define in vivo norovirus cell tropism have generated conflicting results. Some studies detected infected intestinal immune cells 8-12 , other studies detected epithelial cells 13 , and still others detected immune and epithelial cells 14-16 . Major limitations of these studies are that they were performed on tissue sections from immunocompromised or germ-free hosts, chronically infected hosts where the timing of infection was unknown, or following non-biologically relevant inoculation routes. Here, we report that the dominant cellular targets of a murine norovirus inoculated orally into immunocompetent mice are macrophages, dendritic cells, B cells and T cells in the gut-associated lymphoid tissue. Importantly, we also demonstrate that a norovirus can infect T cells, a previously unrecognized target, in vitro. These findings represent the most extensive analyses to date of in vivo norovirus cell tropism in orally inoculated, immunocompetent hosts at the peak of acute infection and thus they significantly advance our basic understanding of norovirus pathogenesis.
Collapse
Affiliation(s)
- Katrina R Grau
- Department of Molecular Genetics & Microbiology, Emerging Pathogens Institute, Center for Inflammation and Mucosal Immunology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Alexa N Roth
- Department of Molecular Genetics & Microbiology, Emerging Pathogens Institute, Center for Inflammation and Mucosal Immunology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Shu Zhu
- Department of Molecular Genetics & Microbiology, Emerging Pathogens Institute, Center for Inflammation and Mucosal Immunology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Abel Hernandez
- Department of Molecular Genetics & Microbiology, Emerging Pathogens Institute, Center for Inflammation and Mucosal Immunology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Natacha Colliou
- Department of Infectious Diseases & Immunology, Center for Inflammation & Mucosal Immunology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Bayli B DiVita
- Department of Oral Biology, College of Dentistry, University of Florida, Gainesville, FL, USA
| | - Drake T Philip
- Department of Molecular Genetics & Microbiology, Emerging Pathogens Institute, Center for Inflammation and Mucosal Immunology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Cara Riffe
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Benoit Giasson
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Shannon M Wallet
- Department of Oral Biology, College of Dentistry, University of Florida, Gainesville, FL, USA
| | - Mansour Mohamadzadeh
- Department of Infectious Diseases & Immunology, Center for Inflammation & Mucosal Immunology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Stephanie M Karst
- Department of Molecular Genetics & Microbiology, Emerging Pathogens Institute, Center for Inflammation and Mucosal Immunology, College of Medicine, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
47
|
Lee S, Wilen CB, Orvedahl A, McCune BT, Kim KW, Orchard RC, Peterson ST, Nice TJ, Baldridge MT, Virgin HW. Norovirus Cell Tropism Is Determined by Combinatorial Action of a Viral Non-structural Protein and Host Cytokine. Cell Host Microbe 2017; 22:449-459.e4. [PMID: 28966054 PMCID: PMC5679710 DOI: 10.1016/j.chom.2017.08.021] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 07/19/2017] [Accepted: 08/29/2017] [Indexed: 01/24/2023]
Abstract
Cellular tropism during persistent viral infection is commonly conferred by the interaction of a viral surface protein with a host receptor complex. Norovirus, the leading global cause of gastroenteritis, can be persistently shed during infection, but its in vivo cellular tropism and tropism determinants remain unidentified. Using murine norovirus (MNoV), we determine that a small number of intestinal epithelial cells (IECs) serve as the reservoir for fecal shedding and persistence. The viral non-structural protein NS1, rather than a viral surface protein, determines IEC tropism. Expression of NS1 from a persistent MNoV strain is sufficient for an acute MNoV strain to target IECs and persist. In addition, interferon-lambda (IFN-λ) is a key host determinant blocking MNoV infection in IECs. The inability of acute MNoV to shed and persist is rescued in Ifnlr1-/- mice, suggesting that NS1 evades IFN-λ-mediated antiviral immunity. Thus, NS1 and IFN-λ interactions govern IEC tropism and persistence of MNoV.
Collapse
Affiliation(s)
- Sanghyun Lee
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Craig B Wilen
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Anthony Orvedahl
- Department of Pediatrics, Division of Infectious Diseases, Washington University School of Medicine, St. Louis, MO, USA
| | - Broc T McCune
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Ki-Wook Kim
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Robert C Orchard
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Stefan T Peterson
- Department of Medicine, Division of Infectious Diseases, Washington University School of Medicine, St. Louis, MO, USA
| | - Timothy J Nice
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR, USA
| | - Megan T Baldridge
- Department of Medicine, Division of Infectious Diseases, Washington University School of Medicine, St. Louis, MO, USA.
| | - Herbert W Virgin
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
48
|
Enosi Tuipulotu D, Netzler NE, Lun JH, Mackenzie JM, White PA. RNA Sequencing of Murine Norovirus-Infected Cells Reveals Transcriptional Alteration of Genes Important to Viral Recognition and Antigen Presentation. Front Immunol 2017; 8:959. [PMID: 28848558 PMCID: PMC5554501 DOI: 10.3389/fimmu.2017.00959] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Accepted: 07/26/2017] [Indexed: 01/02/2023] Open
Abstract
Viruses inherently exploit normal cellular functions to promote replication and survival. One mechanism involves transcriptional control of the host, and knowledge of the genes modified and their molecular function can aid in understanding viral-host interactions. Norovirus pathogenesis, despite the recent advances in cell cultivation, remains largely uncharacterized. Several studies have utilized the related murine norovirus (MNV) to identify innate response, antigen presentation, and cellular recognition components that are activated during infection. In this study, we have used next-generation sequencing to probe the transcriptomic changes of MNV-infected mouse macrophages. Our in-depth analysis has revealed that MNV is a potent stimulator of the innate response including genes involved in interferon and cytokine production pathways. We observed that genes involved in viral recognition, namely IFIH1, DDX58, and DHX58 were significantly upregulated with infection, whereas we observed significant downregulation of cytokine receptors (Il17rc, Il1rl1, Cxcr3, and Cxcr5) and TLR7. Furthermore, we identified that pathways involved in protein degradation (including genes Psmb3, Psmb4, Psmb5, Psmb9, and Psme2), antigen presentation, and lymphocyte activation are downregulated by MNV infection. Thus, our findings illustrate that MNV induces perturbations in the innate immune transcriptome, particularly in MHC maturation and viral recognition that can contribute to disease pathogenesis.
Collapse
Affiliation(s)
- Daniel Enosi Tuipulotu
- Faculty of Science, School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Natalie E Netzler
- Faculty of Science, School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Jennifer H Lun
- Faculty of Science, School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Jason M Mackenzie
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Peter A White
- Faculty of Science, School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
49
|
Lee H, Ko G. New perspectives regarding the antiviral effect of vitamin A on norovirus using modulation of gut microbiota. Gut Microbes 2017; 8:616-620. [PMID: 28727498 PMCID: PMC5730389 DOI: 10.1080/19490976.2017.1353842] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Gut microbiota has been revealed to play an important role in various health conditions, and recent studies have suggested the modulation of gut microbiota as a therapeutic strategy. There is no effective treatment of norovirus infection, though vitamin A has been suggested to have an antiviral effect in an epidemiological study. We demonstrated that vitamin A significantly inhibited murine norovirus replication. Vitamin A supplementation significantly increased the abundance of Lactobacillus sp. during norovirus infection, which played a crucial role in antiviral efficacy, inhibiting murine norovirus. Therefore, we elaborated the antiviral effect of vitamin A via modulation of gut microbiota. Furthermore, we suggest a novel strategy, using potential probiotics, as having a protective and therapeutic effect on noroviral infection.
Collapse
Affiliation(s)
- Heetae Lee
- College of Pharmacy, Sahmyook University, Seoul, Korea
| | - GwangPyo Ko
- Center for Human and Environmental Microbiome, Institute of Health and Environment, School of Public Health, Seoul National University, Seoul, Korea,CONTACT GwangPyo Ko School of Public Health, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 151-742, Korea
| |
Collapse
|
50
|
Lee S, Baldridge MT. Interferon-Lambda: A Potent Regulator of Intestinal Viral Infections. Front Immunol 2017; 8:749. [PMID: 28713375 PMCID: PMC5491552 DOI: 10.3389/fimmu.2017.00749] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 06/13/2017] [Indexed: 12/12/2022] Open
Abstract
Interferon-lambda (IFN-λ) is a recently described cytokine found to be of critical importance in innate immune regulation of intestinal viruses. Endogenous IFN-λ has potent antiviral effects and has been shown to control multiple intestinal viruses and may represent a factor that contributes to human variability in response to infection. Importantly, recombinant IFN-λ has therapeutic potential against enteric viral infections, many of which lack other effective treatments. In this mini-review, we describe recent advances regarding IFN-λ-mediated regulation of enteric viruses with important clinical relevance including rotavirus, reovirus, and norovirus. We also briefly discuss IFN-λ interactions with other cytokines important in the intestine, and how IFN-λ may play a role in regulation of intestinal viruses by the commensal microbiome. Finally, we indicate currently outstanding questions regarding IFN-λ control of enteric infections that remain to be explored to enhance our understanding of this important immune molecule.
Collapse
Affiliation(s)
- Sanghyun Lee
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, United States
| | - Megan T Baldridge
- Department of Medicine, Division of Infectious Diseases, Washington University School of Medicine, St. Louis, MO, United States
| |
Collapse
|