1
|
Steigmann JC, Zhou X, Suttenberg LN, Salman I, Rehmathullah ZF, Weinberg JB. Effects of immunoproteasome inhibition on acute respiratory infection with murine hepatitis virus strain 1. J Virol 2024; 98:e0123824. [PMID: 39508578 PMCID: PMC11650983 DOI: 10.1128/jvi.01238-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 10/18/2024] [Indexed: 11/15/2024] Open
Abstract
The immunoproteasome (IP) is a predominantly inducible component of the ubiquitin proteasome system that plays key roles in multiple aspects of immune function, inflammation, and protein homeostasis. We used murine hepatitis virus strain 1 (MHV-1), a mouse coronavirus, to define the role of IP activity during acute coronavirus respiratory infection. Expression of the β5i subunit of the IP and cytokines that induce IP activity, including IFN-γ, TNF-α, and IFN-β, increased in lungs and livers of CH3/HeJ mice following intranasal infection with MHV-1. IP inhibition using ONX-0914 did not affect MHV-1 replication in bone marrow-derived dendritic cells in vitro. IP inhibition in vivo exacerbated virus-induced weight loss and mortality but had no effect on virus replication in lungs or livers. IP inhibition had minimal effect on virus-induced pulmonary inflammation but led to substantially increased liver pathology, including greater upregulation of pro-inflammatory cytokines and histological evidence of inflammation and necrosis. Those findings were associated with evidence of increased endoplasmic reticulum stress although not with accumulation of ubiquitinated protein. Our results indicate that the IP is a protective host factor during acute MHV-1 infection. IMPORTANCE Inflammatory responses triggered by acute infection by respiratory viruses such as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) drive morbidity and mortality. Infection of mice with murine hepatitis virus strain 1 (MHV-1), a mouse coronavirus, is a useful model to study the pathogenesis of coronavirus respiratory infections. The immunoproteasome is an inducible component of the ubiquitin proteasome system that is poised to contribute to multiple aspects of immune function, inflammation, and protein homeostasis during an infection. We used the MHV-1 model to define the role of the immunoproteasome in coronavirus pathogenesis. We found that immunoproteasome subunit expression increases in the lungs and the liver during acute MHV-1 respiratory infection. Inhibition of immunoproteasome activity did not affect MHV-1 replication but increased MHV-1-induced weight loss, mortality, and inflammation in lungs and livers. Thus, our findings indicate that the immunoproteasome is a critical protective host factor during coronavirus respiratory infection.
Collapse
Affiliation(s)
- Jacob C. Steigmann
- Department of Pediatrics, Division of Infectious Diseases, University of Michigan, Ann Arbor, Michigan, USA
| | - Xiaofeng Zhou
- Department of Microbiology & Immunology, University of Michigan, Ann Arbor, Michigan, USA
| | - Lauren N. Suttenberg
- Department of Pediatrics, Division of Infectious Diseases, University of Michigan, Ann Arbor, Michigan, USA
| | - Irha Salman
- Department of Pediatrics, Division of Infectious Diseases, University of Michigan, Ann Arbor, Michigan, USA
| | - Zainab F. Rehmathullah
- Department of Pediatrics, Division of Infectious Diseases, University of Michigan, Ann Arbor, Michigan, USA
| | - Jason B. Weinberg
- Department of Pediatrics, Division of Infectious Diseases, University of Michigan, Ann Arbor, Michigan, USA
- Department of Microbiology & Immunology, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
2
|
Queiroz-Junior CM, Santos ACPM, Gonçalves MR, Brito CB, Barrioni B, Almeida PJ, Gonçalves-Pereira MH, Silva T, Oliveira SR, Pereira MM, Santiago HC, Teixeira MM, Costa VV. Acute coronavirus infection triggers a TNF-dependent osteoporotic phenotype in mice. Life Sci 2023; 324:121750. [PMID: 37142087 PMCID: PMC10152759 DOI: 10.1016/j.lfs.2023.121750] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 04/24/2023] [Accepted: 04/27/2023] [Indexed: 05/06/2023]
Abstract
AIMS Millions of people died during the COVID-19 pandemic, but the vast majority of infected individuals survived. Now, some consequences of the disease, known as long COVID, are been revealed. Although the respiratory system is the target of Sars-CoV-2, COVID-19 can influence other parts of the body, including bone. The aim of this work was to investigate the impact of acute coronavirus infection in bone metabolism. MAIN METHODS We evaluated RANKL/OPG levels in serum samples of patients with and without acute COVID-19. In vitro, the effects of coronavirus in osteoclasts and osteoblasts were investigated. In vivo, we evaluated the bone phenotype in a BSL2 mouse model of SARS-like disease induced by murine coronavirus (MHV-3). KEY FINDINGS Patients with acute COVID-19 presented decreased OPG and increased RANKL/OPG ratio in the serum versus healthy individuals. In vitro, MHV-3 infected macrophages and osteoclasts, increasing their differentiation and TNF release. Oppositely, osteoblasts were not infected. In vivo, MHV-3 lung infection triggered bone resorption in the femur of mice, increasing the number of osteoclasts at 3dpi and decreasing at 5dpi. Indeed, apoptotic-caspase-3+ cells have been detected in the femur after infection as well as viral RNA. RANKL/OPG ratio and TNF levels also increased in the femur after infection. Accordingly, the bone phenotype of TNFRp55-/- mice infected with MHV-3 showed no signs of bone resorption or increase in the number of osteoclasts. SIGNIFICANCE Coronavirus induces an osteoporotic phenotype in mice dependent on TNF and on macrophage/osteoclast infection.
Collapse
Affiliation(s)
- Celso M Queiroz-Junior
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil.
| | - Anna C P M Santos
- Department of Microbiology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Matheus R Gonçalves
- Department of Microbiology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Camila B Brito
- Department of Microbiology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Breno Barrioni
- Institute of Engineering, Science and Technology, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Janaúba, MG, Brazil
| | - Pedro J Almeida
- Medical School, Ciências da Saúde: Infectologia e Medicina Tropical, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Marcela H Gonçalves-Pereira
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Tarcília Silva
- Department of Oral Surgery and Pathology, School of Dentistry, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Sicília R Oliveira
- Department of Oral Surgery and Pathology, School of Dentistry, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Marivalda M Pereira
- Department of Metallurgical Engineering and Materials, School of Engineering, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Helton C Santiago
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Mauro M Teixeira
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Vivian V Costa
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil; Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil.
| |
Collapse
|
3
|
Abstract
The existence of coronaviruses has been known for many years. These viruses cause significant disease that primarily seems to affect agricultural species. Human coronavirus disease due to the 2002 outbreak of Severe Acute Respiratory Syndrome and the 2012 outbreak of Middle East Respiratory Syndrome made headlines; however, these outbreaks were controlled, and public concern quickly faded. This complacency ended in late 2019 when alarms were raised about a mysterious virus responsible for numerous illnesses and deaths in China. As we now know, this novel disease called Coronavirus Disease 2019 (COVID-19) was caused by Severe acute respiratory syndrome-related-coronavirus-2 (SARS-CoV-2) and rapidly became a worldwide pandemic. Luckily, decades of research into animal coronaviruses hastened our understanding of the genetics, structure, transmission, and pathogenesis of these viruses. Coronaviruses infect a wide range of wild and domestic animals, with significant economic impact in several agricultural species. Their large genome, low dependency on host cellular proteins, and frequent recombination allow coronaviruses to successfully cross species barriers and adapt to different hosts including humans. The study of the animal diseases provides an understanding of the virus biology and pathogenesis and has assisted in the rapid development of the SARS-CoV-2 vaccines. Here, we briefly review the classification, origin, etiology, transmission mechanisms, pathogenesis, clinical signs, diagnosis, treatment, and prevention strategies, including available vaccines, for coronaviruses that affect domestic, farm, laboratory, and wild animal species. We also briefly describe the coronaviruses that affect humans. Expanding our knowledge of this complex group of viruses will better prepare us to design strategies to prevent and/or minimize the impact of future coronavirus outbreaks.
Collapse
Key Words
- bcov, bovine coronavirus
- ccov, canine coronavirus
- cov(s), coronavirus(es)
- covid-19, coronavirus disease 2019
- crcov, canine respiratory coronavirus
- e, coronaviral envelope protein
- ecov, equine coronavirus
- fcov, feline coronavirus
- fipv, feline infectious peritonitis virus
- gfcov, guinea fowl coronavirus
- hcov, human coronavirus
- ibv, infectious bronchitis virus
- m, coronaviral membrane protein
- mers, middle east respiratory syndrome-coronavirus
- mhv, mouse hepatitis virus
- pedv, porcine epidemic diarrhea virus
- pdcov, porcine deltacoronavirus
- phcov, pheasant coronavirus
- phev, porcine hemagglutinating encephalomyelitis virus
- prcov, porcine respiratory coronavirus
- rt-pcr, reverse transcriptase polymerase chain reaction
- s, coronaviral spike protein
- sads-cov, swine acute diarrhea syndrome-coronavirus
- sars-cov, severe acute respiratory syndrome-coronavirus
- sars-cov-2, severe acute respiratory syndrome–coronavirus–2
- tcov, turkey coronavirus
- tgev, transmissible gastroenteritis virus
Collapse
Affiliation(s)
- Alfonso S Gozalo
- Comparative Medicine Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland;,
| | - Tannia S Clark
- Office of Laboratory Animal Medicine, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland
| | - David M Kurtz
- Comparative Medicine Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, Durham, North Carolina
| |
Collapse
|
4
|
Known Cellular and Receptor Interactions of Animal and Human Coronaviruses: A Review. Viruses 2022; 14:v14020351. [PMID: 35215937 PMCID: PMC8878323 DOI: 10.3390/v14020351] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 02/03/2022] [Accepted: 02/05/2022] [Indexed: 12/12/2022] Open
Abstract
This article aims to review all currently known interactions between animal and human coronaviruses and their cellular receptors. Over the past 20 years, three novel coronaviruses have emerged that have caused severe disease in humans, including SARS-CoV-2 (severe acute respiratory syndrome virus 2); therefore, a deeper understanding of coronavirus host-cell interactions is essential. Receptor-binding is the first stage in coronavirus entry prior to replication and can be altered by minor changes within the spike protein-the coronavirus surface glycoprotein responsible for the recognition of cell-surface receptors. The recognition of receptors by coronaviruses is also a major determinant in infection, tropism, and pathogenesis and acts as a key target for host-immune surveillance and other potential intervention strategies. We aim to highlight the need for a continued in-depth understanding of this subject area following on from the SARS-CoV-2 pandemic, with the possibility for more zoonotic transmission events. We also acknowledge the need for more targeted research towards glycan-coronavirus interactions as zoonotic spillover events from animals to humans, following an alteration in glycan-binding capability, have been well-documented for other viruses such as Influenza A.
Collapse
|
5
|
Nakayama M, Kyuwa S. Basic reproduction numbers of three strains of mouse hepatitis viruses in mice. Microbiol Immunol 2022; 66:166-172. [PMID: 34984727 PMCID: PMC9306726 DOI: 10.1111/1348-0421.12961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/29/2021] [Accepted: 12/26/2021] [Indexed: 11/26/2022]
Abstract
Mouse hepatitis virus (MHV) is a murine coronavirus and one of the most important pathogens in laboratory mice. Although various strains of MHV have been isolated, they are generally excreted in the feces and transmitted oronasally via aerosols and contaminated bedding. In this study, we attempted to determine the basic reproduction numbers (R0) of three strains of MHV to improve our understanding of MHV infections in mice. Five‐week‐old female C57BL/6J mice were inoculated intranasally with either the Y, NuU, or JHM variant strain of MHV and housed with two naïve mice. After 4 weeks, the presence or absence of anti‐MHV antibody in the mice was determined by ELISA. We also examined the distribution of MHV in the organs of Y, NuU, or JHM variant‐infected mice. Our data suggest that the transmissibility of MHV is correlated with viral growth in the gastrointestinal tract of infected mice. To the best of our knowledge, this is the first report to address the basic reproduction numbers among pathogens in laboratory animals.
Collapse
Affiliation(s)
- Masataka Nakayama
- Laboratory of Biomedical Science, Department of Veterinary Medical Science, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Shigeru Kyuwa
- Laboratory of Biomedical Science, Department of Veterinary Medical Science, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
6
|
Compton SR. Overview of Coronaviruses in Veterinary Medicine. Comp Med 2021; 71:333-341. [PMID: 34412731 PMCID: PMC8594256 DOI: 10.30802/aalas-cm-21-000007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 04/08/2021] [Accepted: 05/10/2021] [Indexed: 11/05/2022]
Abstract
Coronaviruses infect humans and a wide range of animals, causing predominantly respiratory and intestinal infections. This review provides background on the taxonomy of coronaviruses, the functions of viral proteins, and the life cycle of coronaviruses. In addition, the review focuses on coronaviral diseases in several agriculturally important, companion, and laboratory animal species (cats, cattle, chickens, dogs, mice, rats and swine) and briefly reviews human coronaviruses and their origins.
Collapse
Key Words
- apn, aminopeptidase n
- bcov, bovine coronavirus
- fecv, feline enteric coronavirus
- fipv, feline infectious peritonitis virus
- ibv, infectious bronchitis virus
- mers-cov, middle east respiratory syndrome coronavirus
- mhv, mouse hepatitis virus
- pdcov, porcine deltacoronavirus
- pedv, porcine endemic diarrhea virus
- phev, porcine hemagglutinating encephalomyelitis virus
- prcv, porcine respiratory coronavirus
- rcv, rat coronavirus
- sars-cov, severe acute respiratory syndrome coronavirus
- seacov, swine enteric alphacoronavirus
- tgev, transmissible gastroenteritis virus
Collapse
|
7
|
Li B, Wang D, Lee MMS, Wang W, Tan Q, Zhao Z, Tang BZ, Huang X. Fabrics Attached with Highly Efficient Aggregation-Induced Emission Photosensitizer: Toward Self-Antiviral Personal Protective Equipment. ACS NANO 2021; 15:13857-13870. [PMID: 34313425 DOI: 10.1021/acsnano.1c06071] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Personal protective equipment (PPE) is vital for the prevention and control of SARS-CoV-2. However, conventional PPEs lack virucidal capabilities and arbitrarily discarding used PPEs may cause a high risk for cross-contamination and environmental pollution. Recently reported photothermal or photodynamic-mediated self-sterilizing masks show bactericidal-virucidal abilities but have some inherent disadvantages, such as generating unbearable heat during the photothermal process or requiring additional ultraviolet light irradiation to inactivate pathogens, which limit their practical applications. Here, we report the fabrication of a series of fabrics (derived from various PPEs) with real-time self-antiviral capabilities, on the basis of a highly efficient aggregation-induced emission photosensitizer (namely, ASCP-TPA). ASCP-TPA possesses facile synthesis, excellent biocompatibility, and extremely high reactive oxygen species generation capacity, which significantly outperforms the traditional photosensitizers. Meanwhile, the ASCP-TPA-attached fabrics (ATaFs) show tremendous photodynamic inactivation effects against MHV-A59, a surrogate coronavirus of SARS-CoV-2. Upon ultralow-power white light irradiation (3.0 mW cm-2), >99.999% virions (5 log) on the ATaFs are eliminated within 10 min. Such ultralow-power requirement and rapid virus-killing ability enable ATaFs-based PPEs to provide real-time protection for the wearers under indoor light irradiation. ATaFs' virucidal abilities are retained after 100 washings or continuous exposure to office light for 2 weeks, which offers the benefits of reusability and long-term usability. Furthermore, ATaFs show no toxicity to normal skin, even upon continuous high-power light illumination. This self-antiviral ATaFs-based strategy may also be applied to fight against other airborne pathogens and holds huge potential to alleviate global PPE supply shortages.
Collapse
Affiliation(s)
- Bin Li
- Center for Infection and Immunity, Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai 519000, Guangdong, China
- Southern Marine Science and Engineering Guangdong Laboratory, Zhuhai 519000, Guangdong, China
- Key Laboratory of Tropical Diseases Control, Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, Guangdong, China
| | - Dong Wang
- Center for AIE Research, College of Materials Science and Engineering, Shenzhen University, Shenzhen 518060, Guangdong, China
- Department of Chemistry, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon 999077, Hong Kong, China
| | - Michelle M S Lee
- Department of Chemistry, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon 999077, Hong Kong, China
| | - Wei Wang
- Center for Infection and Immunity, Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai 519000, Guangdong, China
| | - Qingqin Tan
- Center for Infection and Immunity, Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai 519000, Guangdong, China
| | - Zhaoyan Zhao
- Center for Infection and Immunity, Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai 519000, Guangdong, China
| | - Ben Zhong Tang
- Center for AIE Research, College of Materials Science and Engineering, Shenzhen University, Shenzhen 518060, Guangdong, China
- Department of Chemistry, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon 999077, Hong Kong, China
| | - Xi Huang
- Center for Infection and Immunity, Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai 519000, Guangdong, China
- Southern Marine Science and Engineering Guangdong Laboratory, Zhuhai 519000, Guangdong, China
- Key Laboratory of Tropical Diseases Control, Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, Guangdong, China
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan 511518, Guangdong, China
| |
Collapse
|
8
|
Abstract
SARS-CoV-2 infection activates TLR2 signaling, which results in robust expression of proinflammatory cytokines that may contribute to disease in severe COVID-19. Inhibition of this signaling pathway represents a potential target for COVID-19 therapeutics.
Collapse
Affiliation(s)
- Alan Sariol
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, USA
| | - Stanley Perlman
- Interdisciplinary Program in Immunology, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
9
|
Li B, Wang W, Song W, Zhao Z, Tan Q, Zhao Z, Tang L, Zhu T, Yin J, Bai J, Dong X, Tan S, Hu Q, Tang BZ, Huang X. Antiviral and Anti‐Inflammatory Treatment with Multifunctional Alveolar Macrophage‐Like Nanoparticles in a Surrogate Mouse Model of COVID‐19. ADVANCED SCIENCE 2021; 8:2003556. [PMCID: PMC8209923 DOI: 10.1002/advs.202003556] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
The pandemic of coronavirus disease 2019 (COVID‐19) is continually worsening. Clinical treatment for COVID‐19 remains primarily supportive with no specific medicines or regimens. Here, the development of multifunctional alveolar macrophage (AM)‐like nanoparticles (NPs) with photothermal inactivation capability for COVID‐19 treatment is reported. The NPs, made by wrapping polymeric cores with AM membranes, display the same surface receptors as AMs, including the coronavirus receptor and multiple cytokine receptors. By acting as AM decoys, the NPs block coronavirus from host cell entry and absorb various proinflammatory cytokines, thus achieving combined antiviral and anti‐inflammatory treatment. To enhance the antiviral efficiency, an efficient photothermal material based on aggregation‐induced emission luminogens is doped into the NPs for virus photothermal disruption under near‐infrared (NIR) irradiation. In a surrogate mouse model of COVID‐19 caused by murine coronavirus, treatment with multifunctional AM‐like NPs with NIR irradiation decreases virus burden and cytokine levels, reduces lung damage and inflammation, and confers a significant survival advantage to the infected mice. Crucially, this therapeutic strategy may be clinically applied for the treatment of COVID‐19 at early stage through atomization inhalation of the NPs followed by NIR irradiation of the respiratory tract, thus alleviating infection progression and reducing transmission risk.
Collapse
Affiliation(s)
- Bin Li
- Center for Infection and ImmunityGuangdong Provincial Key Laboratory of Biomedical ImagingThe Fifth Affiliated Hospital of Sun Yat‐sen UniversityZhuhaiGuangdong519000China
- Southern Marine Science and Engineering Guangdong LaboratoryZhuhaiGuangdong519000China
| | - Wei Wang
- Center for Infection and ImmunityGuangdong Provincial Key Laboratory of Biomedical ImagingThe Fifth Affiliated Hospital of Sun Yat‐sen UniversityZhuhaiGuangdong519000China
- Engineering Research Center of Tibetan Medicine Detection Technology, Ministry of EducationXizang Minzu UniversityXianyangShaanxi712082China
| | - Weifeng Song
- Center for Infection and ImmunityGuangdong Provincial Key Laboratory of Biomedical ImagingThe Fifth Affiliated Hospital of Sun Yat‐sen UniversityZhuhaiGuangdong519000China
| | - Zheng Zhao
- Department of ChemistryThe Hong Kong University of Science and TechnologyClear Water BayKowloonHong Kong999077China
| | - Qingqin Tan
- Center for Infection and ImmunityGuangdong Provincial Key Laboratory of Biomedical ImagingThe Fifth Affiliated Hospital of Sun Yat‐sen UniversityZhuhaiGuangdong519000China
- Southern Marine Science and Engineering Guangdong LaboratoryZhuhaiGuangdong519000China
| | - Zhaoyan Zhao
- Center for Infection and ImmunityGuangdong Provincial Key Laboratory of Biomedical ImagingThe Fifth Affiliated Hospital of Sun Yat‐sen UniversityZhuhaiGuangdong519000China
- Southern Marine Science and Engineering Guangdong LaboratoryZhuhaiGuangdong519000China
| | - Lantian Tang
- Center for Infection and ImmunityGuangdong Provincial Key Laboratory of Biomedical ImagingThe Fifth Affiliated Hospital of Sun Yat‐sen UniversityZhuhaiGuangdong519000China
- Southern Marine Science and Engineering Guangdong LaboratoryZhuhaiGuangdong519000China
| | - Tianchuan Zhu
- Center for Infection and ImmunityGuangdong Provincial Key Laboratory of Biomedical ImagingThe Fifth Affiliated Hospital of Sun Yat‐sen UniversityZhuhaiGuangdong519000China
- Southern Marine Science and Engineering Guangdong LaboratoryZhuhaiGuangdong519000China
| | - Jialing Yin
- Center for Infection and ImmunityGuangdong Provincial Key Laboratory of Biomedical ImagingThe Fifth Affiliated Hospital of Sun Yat‐sen UniversityZhuhaiGuangdong519000China
- Southern Marine Science and Engineering Guangdong LaboratoryZhuhaiGuangdong519000China
| | - Jun Bai
- Center for Infection and ImmunityGuangdong Provincial Key Laboratory of Biomedical ImagingThe Fifth Affiliated Hospital of Sun Yat‐sen UniversityZhuhaiGuangdong519000China
- Southern Marine Science and Engineering Guangdong LaboratoryZhuhaiGuangdong519000China
| | - Xin Dong
- Center for Infection and ImmunityGuangdong Provincial Key Laboratory of Biomedical ImagingThe Fifth Affiliated Hospital of Sun Yat‐sen UniversityZhuhaiGuangdong519000China
- Southern Marine Science and Engineering Guangdong LaboratoryZhuhaiGuangdong519000China
| | - Siyi Tan
- Center for Infection and ImmunityGuangdong Provincial Key Laboratory of Biomedical ImagingThe Fifth Affiliated Hospital of Sun Yat‐sen UniversityZhuhaiGuangdong519000China
- Southern Marine Science and Engineering Guangdong LaboratoryZhuhaiGuangdong519000China
| | - Qunying Hu
- Engineering Research Center of Tibetan Medicine Detection Technology, Ministry of EducationXizang Minzu UniversityXianyangShaanxi712082China
| | - Ben Zhong Tang
- Department of ChemistryThe Hong Kong University of Science and TechnologyClear Water BayKowloonHong Kong999077China
| | - Xi Huang
- Center for Infection and ImmunityGuangdong Provincial Key Laboratory of Biomedical ImagingThe Fifth Affiliated Hospital of Sun Yat‐sen UniversityZhuhaiGuangdong519000China
- Southern Marine Science and Engineering Guangdong LaboratoryZhuhaiGuangdong519000China
| |
Collapse
|
10
|
Sariol A, Perlman S. Lessons for COVID-19 Immunity from Other Coronavirus Infections. Immunity 2020; 53:248-263. [PMID: 32717182 PMCID: PMC7359787 DOI: 10.1016/j.immuni.2020.07.005] [Citation(s) in RCA: 225] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 07/07/2020] [Accepted: 07/08/2020] [Indexed: 12/13/2022]
Abstract
A key goal to controlling coronavirus disease 2019 (COVID-19) is developing an effective vaccine. Development of a vaccine requires knowledge of what constitutes a protective immune response and also features that might be pathogenic. Protective and pathogenic aspects of the response to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) are not well understood, partly because the virus has infected humans for only 6 months. However, insight into coronavirus immunity can be informed by previous studies of immune responses to non-human coronaviruses, common cold coronaviruses, and SARS-CoV and Middle East respiratory syndrome coronavirus (MERS-CoV). Here, we review the literature describing these responses and discuss their relevance to the SARS-CoV-2 immune response.
Collapse
Affiliation(s)
- Alan Sariol
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA 52242, USA
| | - Stanley Perlman
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA 52242, USA; Department of Microbiology and Immunology, University of Iowa, Iowa City, IA 52242, USA.
| |
Collapse
|
11
|
Abstract
A key goal to controlling coronavirus disease 2019 (COVID-19) is developing an effective vaccine. Development of a vaccine requires knowledge of what constitutes a protective immune response and also features that might be pathogenic. Protective and pathogenic aspects of the response to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) are not well understood, partly because the virus has infected humans for only 6 months. However, insight into coronavirus immunity can be informed by previous studies of immune responses to non-human coronaviruses, common cold coronaviruses, and SARS-CoV and Middle East respiratory syndrome coronavirus (MERS-CoV). Here, we review the literature describing these responses and discuss their relevance to the SARS-CoV-2 immune response.
Collapse
Affiliation(s)
- Alan Sariol
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA 52242, USA
| | - Stanley Perlman
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA 52242, USA; Department of Microbiology and Immunology, University of Iowa, Iowa City, IA 52242, USA.
| |
Collapse
|
12
|
Körner RW, Majjouti M, Alcazar MAA, Mahabir E. Of Mice and Men: The Coronavirus MHV and Mouse Models as a Translational Approach to Understand SARS-CoV-2. Viruses 2020; 12:E880. [PMID: 32806708 PMCID: PMC7471983 DOI: 10.3390/v12080880] [Citation(s) in RCA: 107] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 08/07/2020] [Accepted: 08/10/2020] [Indexed: 02/06/2023] Open
Abstract
The fatal acute respiratory coronavirus disease 2019 (COVID-19) is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Since COVID-19 was declared a pandemic by the World Health Organization in March 2020, infection and mortality rates have been rising steadily worldwide. The lack of a vaccine, as well as preventive and therapeutic strategies, emphasize the need to develop new strategies to mitigate SARS-CoV-2 transmission and pathogenesis. Since mouse hepatitis virus (MHV), severe acute respiratory syndrome coronavirus (SARS-CoV), and SARS-CoV-2 share a common genus, lessons learnt from MHV and SARS-CoV could offer mechanistic insights into SARS-CoV-2. This review provides a comprehensive review of MHV in mice and SARS-CoV-2 in humans, thereby highlighting further translational avenues in the development of innovative strategies in controlling the detrimental course of SARS-CoV-2. Specifically, we have focused on various aspects, including host species, organotropism, transmission, clinical disease, pathogenesis, control and therapy, MHV as a model for SARS-CoV and SARS-CoV-2 as well as mouse models for infection with SARS-CoV and SARS-CoV-2. While MHV in mice and SARS-CoV-2 in humans share various similarities, there are also differences that need to be addressed when studying murine models. Translational approaches, such as humanized mouse models are pivotal in studying the clinical course and pathology observed in COVID-19 patients. Lessons from prior murine studies on coronavirus, coupled with novel murine models could offer new promising avenues for treatment of COVID-19.
Collapse
Affiliation(s)
- Robert W. Körner
- Department of Pediatrics, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany;
| | - Mohamed Majjouti
- Comparative Medicine, Center for Molecular Medicine Cologne (CMMC), University of Cologne, Faculty of Medicine and University Hospital Cologne, 50931 Cologne, Germany;
| | - Miguel A. Alejandre Alcazar
- Department of Pediatric and Adolescent Medicine, Translational Experimental Pediatrics—Experimental Pulmonology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany;
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany
- Member of the German Center for Lung Research (DZL), Institute for Lung Health, University of Giessen and Marburg Lung Center (UGMLC), 50937 Cologne, Germany
| | - Esther Mahabir
- Comparative Medicine, Center for Molecular Medicine Cologne (CMMC), University of Cologne, Faculty of Medicine and University Hospital Cologne, 50931 Cologne, Germany;
| |
Collapse
|
13
|
Islam MM, Toohey B, Purcell DFJ, Kannourakis G. Suppression subtractive hybridization method for the identification of a new strain of murine hepatitis virus from xenografted SCID mice. Arch Virol 2015; 160:2945-55. [PMID: 26347284 PMCID: PMC4635179 DOI: 10.1007/s00705-015-2592-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 08/27/2015] [Indexed: 11/21/2022]
Abstract
During attempts to clone retroviral determinants associated with a mouse model of Langerhans cell histiocytosis (LCH), suppression subtractive hybridization (SSH) was used to identify unique viruses in the liver of severe combined immunodeficiency (SCID) mice transplanted with LCH tissues. A partial genomic sequence of a murine coronavirus was identified, and the whole genome (31428 bp) of the coronavirus was subsequently sequenced using PCR cloning techniques. Nucleotide sequence comparisons revealed that the genome sequence of the new virus was 91-93% identical to those of known murine hepatitis viruses (MHVs). The predicted open reading frame from the nucleotide sequence encoded all known proteins of MHVs. Analysis at the protein level showed that the virus was closely related to the highly virulent MHV-JHM strain. The virus strain was named MHV-MI. No type D retroviruses were found. Degenerate PCR targeting of type D retrovirus and 5'-RACE targeting of other types of retroviruses confirmed the absence of any retroviral association with the LCH xenografted SCID mice.
Collapse
Affiliation(s)
- Mohammed M Islam
- Fiona Elsey Cancer Research Institute, 106-110 Lydiard Street South, Ballarat Technology Park Central, Ballarat, VIC, 3353, Australia.
- School of Applied and Biomedical Sciences, Federation University Australia, Ballarat, VIC, 3350, Australia.
| | - Brendan Toohey
- Fiona Elsey Cancer Research Institute, 106-110 Lydiard Street South, Ballarat Technology Park Central, Ballarat, VIC, 3353, Australia
| | - Damian F J Purcell
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - George Kannourakis
- Fiona Elsey Cancer Research Institute, 106-110 Lydiard Street South, Ballarat Technology Park Central, Ballarat, VIC, 3353, Australia
- School of Applied and Biomedical Sciences, Federation University Australia, Ballarat, VIC, 3350, Australia
| |
Collapse
|
14
|
Yang Z, Du J, Chen G, Zhao J, Yang X, Su L, Cheng G, Tang H. Coronavirus MHV-A59 infects the lung and causes severe pneumonia in C57BL/6 mice. Virol Sin 2014; 29:393-402. [PMID: 25547683 PMCID: PMC7090691 DOI: 10.1007/s12250-014-3530-y] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2014] [Accepted: 12/10/2014] [Indexed: 12/21/2022] Open
Abstract
It remains challenging to develop animal models of lung infection and severe pneumonia by severe acute respiratory syndrome coronavirus (SARS-CoV) and Middle East respiratory syndrome cornavirus (MERS-CoV) without high level of containment. This inevitably hinders understanding of virushost interaction and development of appropriate countermeasures. Here we report that intranasal inoculation of sublethal doses of murine coronavirus mouse hepatitis virus A-59 (MHV-A59), a hepatic and neuronal tropic coronavirus, can induce acute pneumonia and severe lung injuries in C57BL/6 mice. Inflammatory leukocyte infiltrations, hemorrhages and fibrosis of alveolar walls can be observed 2–11 days after MHV-A59 infection. This pathological manifestation is associated with dramatical elevation of tissue IP-10 and IFN-γ and moderate increase of TNF-α and IL-1β, but inability of anti-viral type I interferon response. These results suggest that intranasal infection of MHV-A59 would serve as a surrogate mouse model of acute respiratory distress syndrome by SARS-CoV and MERS-CoV infections.
Collapse
Affiliation(s)
- Zhangsheng Yang
- Key Laboratory of Infection and Immunity (CASKLII), Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Chaoyang District, Beijing, 100101, China
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Slobodskaya O, Snijder EJ, Spaan WJM. Organ tropism of murine coronavirus does not correlate with the expression levels of the membrane-anchored or secreted isoforms of the carcinoembryonic antigen-related cell adhesion molecule 1 receptor. J Gen Virol 2012; 93:1918-1923. [PMID: 22673933 DOI: 10.1099/vir.0.043190-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1) is the sole known receptor of murine hepatitis virus (MHV) A59, but the available, often qualitative, data about CEACAM1 expression does not explain MHV organ tropism. Ceacam1 transcripts undergo alternative splicing resulting in multiple isoforms, including secreted CEACAM1 isoforms that can neutralize the virus. We determined the quantities of Ceacam1 transcripts encoding membrane-bound and secreted isoforms in mouse organs and a set of cell lines. In vivo, the lowest receptor mRNA levels were found in brain and muscle and these were similar to those in easily infectable cultured cells. While the quantities of the receptor transcripts varied between mouse organs, their abundance did not correlate with susceptibility to MHV infection. The proportion of transcripts encoding secreted isoforms also could not explain the selection of sites for virus replication, as it was constant in all organs. Our data suggest that neither of the two CEACAM1 isoforms defines MHV organ tropism.
Collapse
Affiliation(s)
- Olga Slobodskaya
- Molecular Virology Laboratory, Department of Medical Microbiology, Center of Infectious Diseases, Leiden University Medical Center, The Netherlands
| | - Eric J Snijder
- Molecular Virology Laboratory, Department of Medical Microbiology, Center of Infectious Diseases, Leiden University Medical Center, The Netherlands
| | - Willy J M Spaan
- Molecular Virology Laboratory, Department of Medical Microbiology, Center of Infectious Diseases, Leiden University Medical Center, The Netherlands
| |
Collapse
|
16
|
Ribes JM, Ortego J, Ceriani J, Montava R, Enjuanes L, Buesa J. Transmissible gastroenteritis virus (TGEV)-based vectors with engineered murine tropism express the rotavirus VP7 protein and immunize mice against rotavirus. Virology 2011; 410:107-18. [PMID: 21094967 PMCID: PMC7111951 DOI: 10.1016/j.virol.2010.10.036] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2010] [Revised: 08/18/2010] [Accepted: 10/27/2010] [Indexed: 01/26/2023]
Abstract
A coronavirus vector based on the genome of the porcine transmissible gastroenteritis virus (TGEV) expressing the rotavirus VP7 protein was constructed to immunize and protect against rotavirus infections in a murine model. The tropism of this TGEV-derived vector was modified by replacing the spike S protein with the homologous protein from mouse hepatitis virus (MHV). The rotavirus gene encoding the VP7 protein was cloned into the coronavirus cDNA. BALB/c and STAT1-deficient mice were inoculated with the recombinant viral vector rTGEV(S-MHV)-VP7, which replicates in the intestine and spreads to other organs such as liver, spleen and lungs. TGEV-specific antibodies were detected in all the inoculated BALB/c mice, while rotavirus-specific antibodies were found only after immunization by the intraperitoneal route. Partial protection against rotavirus-induced diarrhea was achieved in suckling BALB/c mice born to dams immunized with the recombinant virus expressing VP7 when they were orally challenged with the homotypic rotavirus strain.
Collapse
Affiliation(s)
- Juan Manuel Ribes
- Department of Microbiology and Ecology, School of Medicine, University of Valencia, Avda. Blasco Ibáñez, 17, 46010 Valencia, Spain
| | | | | | | | | | | |
Collapse
|
17
|
Compton SR. Prevention of murine norovirus infection in neonatal mice by fostering. JOURNAL OF THE AMERICAN ASSOCIATION FOR LABORATORY ANIMAL SCIENCE : JAALAS 2008; 47:25-30. [PMID: 18459709 PMCID: PMC2654003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 11/16/2007] [Revised: 12/18/2007] [Accepted: 12/25/2007] [Indexed: 05/26/2023]
Abstract
Murine norovirus (MNV) causes subclinical chronic infections in adult immunocompetent mice and is endemic in many mouse colonies. The susceptibility of neonatal mice to MNV infection was investigated. Intestinal homogenates from Swiss Webster (SW) mice inoculated orally with MNV-L on postpartum days (ppd) 1 to 3 were negative for MNV by RT-PCR at postinoculation days (pid) 3 and 7. In contrast, 69% of intestinal homogenates prepared on pid 3 and 7 from mice inoculated orally at ppd 5 to 8 were MNV-positive by RT-PCR. Because only mice 10 d of age or older were infected by contact with infected dams, a study was performed to determine whether fostering of neonatal mice from MNV-infected to MNV-naïve dams could be effective at preventing infection of neonatal mice. Four litters each of 1-, 2-, 4-, or 6 d-old mice from MNV-L- infected dams were transferred to naïve dams with similar-aged litters and vice versa. On ppd 21, feces from all MNV-infected dams and litters transferred to them were MNV-positive. In contrast on ppd 21, feces from all MNV-naïve dams and litters transferred to them were MNV-negative. Fostering of 2-d-old mice from 5 of 5 MNV-C-, 5 of 6 MNV-D-, and 7 of 8 MNV-G- infected dams onto MNV-naïve dams prevented MNV infection of the foster mice. In the 2 litters where MNV was detected, dams were infected within 7 d of transfer, suggesting that the neonatal mice had served as fomites. In summary, fostering was effective at preventing MNV infection in 33 of 35 litters of neonatal mice. Precautions to prevent transmission of virus on the surface of neonatal mice to foster dams could increase the efficiency of the fostering process.
Collapse
Affiliation(s)
- Susan R Compton
- Section of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
18
|
Pratelli A. Canine coronavirus inactivation with physical and chemical agents. Vet J 2007; 177:71-9. [PMID: 17513145 PMCID: PMC7110435 DOI: 10.1016/j.tvjl.2007.03.019] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2006] [Revised: 03/02/2007] [Accepted: 03/25/2007] [Indexed: 12/31/2022]
Abstract
Canine coronavirus (CCoV) is responsible for mild or moderate enteritis in puppies. The virus is highly contagious and avoiding contact with infected dogs and their excretions is the only way to ensure disease prevention. Since no studies have yet focused on the sensitivity of CCoV to chemical biocides the present investigation examined the efficiency of physical and chemical methods of viral inactivation. CCoV infectivity was stable at +56 degrees C for up to 30 min, but tended to decrease rapidly at +65 degrees C and +75 degrees C. Germicidal ultra-violet (UV-C) light exposure demonstrated no significant effects on virus inactivation for up to 3 days. CCoV was observed to be more stable at pH 6.0-6.5 while extreme acidic conditions inactivated the virus. Two tested aldehydes inactivated the virus but their action was temperature- and time-dependent. The methods for CCoV inactivation could be applied as animal models to study human coronavirus infection, reducing the risk of accidental exposure of researchers to pathogens during routine laboratory procedures.
Collapse
Affiliation(s)
- Annamaria Pratelli
- Department of Animal Health and Well-being, Strada per Casamassima km 3, 70010 Valenzano, Bari, Italy.
| |
Collapse
|
19
|
Kyuwa S, Ohsawa K, Sato H, Urano T. Replication of enterotropic and polytropic murine coronaviruses in cultured cell lines of mouse origin. Exp Anim 2000; 49:251-7. [PMID: 11109550 DOI: 10.1538/expanim.49.251] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
To understand the virus-cell interactions that occur during murine coronavirus infection, six murine cell lines (A3-1M, B16, CMT-93, DBT, IC-21 and J774A.1) were inoculated with eight murine coronaviruses, including prototype strains of both polytropic and enterotropic biotypes, and new isolates. All virus strains produced a cytopathic effect (CPE) with cell-to-cell fusion in B16, DBT, IC-21 and J774A.1 cells. The CPE was induced most rapidly in IC-21 cells and was visible microscopically in all cell lines tested. In contrast, the coronaviruses produced little CPE in A3-1M and CMT-93 cells. Although most virus-infected cells, except KQ3E-infected A3-1M, CMT-93 and J774A.1 cells, produced progeny viruses in the supernatants when assayed by plaque formation on DBT cells, the kinetics of viral replication were dependent on both the cell line and virus strain; replication of prototype strains was higher than that of new isolates. There was no significant difference in replication of enterotropic and polytropic strains. B16 cells supported the highest level of viral replication. To determine the sensitivity of the cell lines to murine coronaviruses, the 50% tissue culture infectious dose of the coronaviruses was determined with B16, DBT, IC-21 and J774A.1 cells, and compared to that with DBT cells. The results indicate that IC-21 cells were the most sensitive to murine coronaviruses. These data suggest that B16 and IC-21 cells are suitable for large-scale preparation and isolation of murine coronaviruses, respectively.
Collapse
Affiliation(s)
- S Kyuwa
- Division of Microbiology and Genetics, Kumamoto University, Japan
| | | | | | | |
Collapse
|
20
|
Wessner DR, Shick PC, Lu JH, Cardellichio CB, Gagneten SE, Beauchemin N, Holmes KV, Dveksler GS. Mutational analysis of the virus and monoclonal antibody binding sites in MHVR, the cellular receptor of the murine coronavirus mouse hepatitis virus strain A59. J Virol 1998; 72:1941-8. [PMID: 9499047 PMCID: PMC109486 DOI: 10.1128/jvi.72.3.1941-1948.1998] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/1997] [Accepted: 11/26/1997] [Indexed: 02/06/2023] Open
Abstract
The primary cellular receptor for mouse hepatitis virus (MHV), a murine coronavirus, is MHVR (also referred to as Bgp1a or C-CAM), a transmembrane glycoprotein with four immunoglobulin-like domains in the murine biliary glycoprotein (Bgp) subfamily of the carcinoembryonic antigen (CEA) family. Other murine glycoproteins in the Bgp subfamily, including Bgp1b and Bgp2, also can serve as MHV receptors when transfected into MHV-resistant cells. Previous studies have shown that the 108-amino-acid N-terminal domain of MHVR is essential for virus receptor activity and is the binding site for monoclonal antibody (MAb) CC1, an antireceptor MAb that blocks MHV infection in vivo and in vitro. To further elucidate the regions of MHVR required for virus receptor activity and MAb CC1 binding, we constructed chimeras between MHVR and other members of the CEA family and tested them for MHV strain A59 (MHV-A59) receptor activity and MAb CC1 binding activity. In addition, we used site-directed mutagenesis to introduce selected amino acid changes into the N-terminal domains of MHVR and these chimeras and tested the abilities of these mutant glycoproteins to bind MAb CC1 and to function as MHV receptors. Several recombinant glycoproteins exhibited virus receptor activity but did not bind MAb CC1, indicating that the virus and MAb binding sites on the N-terminal domain of MHVR are not identical. Analysis of the recombinant glycoproteins showed that a short region of MHVR, between amino acids 34 and 52, is critical for MHV-A59 receptor activity. Additional regions of the N-terminal variable domain and the constant domains, however, greatly affected receptor activity. Thus, the molecular context in which the amino acids critical for MHV-A59 receptor activity are found profoundly influences the virus receptor activity of the glycoprotein.
Collapse
Affiliation(s)
- D R Wessner
- Department of Pathology, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20814, USA
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Godfraind C, Holmes KV, Coutelier JP. Thymus involution induced by mouse hepatitis virus A59 in BALB/c mice. J Virol 1995; 69:6541-7. [PMID: 7666556 PMCID: PMC189556 DOI: 10.1128/jvi.69.10.6541-6547.1995] [Citation(s) in RCA: 50] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Mouse hepatitis virus A59 (MHV-A59) infection of adult BALB/c mice induced a severe, transient atrophy of the thymus. The effect was maximal at 1 week after infection, and thymuses returned to normal size by 2 weeks after infection. There was no effect of glucocorticoids, since thymus atrophy was also found in adrenalectomized, infected mice. In infected thymus, immature CD4+ CD8+ lymphocytes were selectively depleted, and apoptosis of lymphocytes was increased. The MHV receptor glycoprotein MHVR was detected on thymus epithelial cells but not on T lymphocytes. In a small number of stromal epithelial cells, but in very few lymphocytes, the viral genome was detectable by in situ hybridization. These observations suggested that MHV-A59-induced thymic atrophy results not from a generalized lytic infection of T lymphocytes but rather from apoptosis of immature double-positive T cells that might be caused by infection of a small proportion of thymus epithelial cells or from inappropriate secretion of some factor, such as a cytokine.
Collapse
Affiliation(s)
- C Godfraind
- Laboratory of Pathology, St.-Luc Hospital, Catholic University of Louvain, Brussels, Belgium
| | | | | |
Collapse
|
22
|
Homberger FR. Sequence analysis of the nucleoprotein genes of three enterotropic strains of murine coronavirus. Arch Virol 1995; 140:571-9. [PMID: 7733827 PMCID: PMC7087234 DOI: 10.1007/bf01718432] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/1994] [Accepted: 11/15/1994] [Indexed: 01/26/2023]
Abstract
The nucleotide sequences of the nucleoprotein genes of three enterotropic strains of the murine coronavirus mouse hepatitis virus (MHV-Y, MHV-RI and DVIM) were determined and compared with previously reported sequences of three polytropic (respiratory) strains (MHV-A59, MHV-JHM and MHV-S). Greater than 92% homology was found among the six strains by pair-wise comparison at the nucleotide level. The genes encoded proteins of 451 to 455 residues and the deduced amino acid sequences were more than 91% homologous. A unique deletion of twelve nucleotides was found at the carboxy terminus of MHV-Y and a three nucleotide deletion was found in MHV-RI, which corresponded to the one previously reported in MHV-A59 and MHV-S. Two internal open reading frames were found within the coding region of the nucleoprotein, the smaller one was specific for the enterotropic strains. It could potentially encode a truncated version of the hypothetical protein described for MHV-A59 and MHV-S. Sequence relationship of the N gene showed no correlation with tissue tropism and no sequence or even single amino acid change unique to either tropism group was found. This indicates that the nucleoprotein of MHV probably has no part in the determination of the primary tissue tropism of an MHV strain. The role of the potential internal protein warrants further investigation.
Collapse
Affiliation(s)
- F R Homberger
- Institute of Laboratory Animal Science, University of Zurich, Switzerland
| |
Collapse
|
23
|
Rangel HDA, Verinaud L, Camargo IJ, Gilioli R, Sakurada JK. Murine virus contaminant of Trypanosoma cruzi experimental infection. Rev Inst Med Trop Sao Paulo 1994; 36:423-31. [PMID: 7569609 DOI: 10.1590/s0036-46651994000500006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
The possibility that some virus contaminants could be altering host response to Trypanosoma cruzi experimental infection was investigated. Data obtained showed that CBA/J mice infected with stocks of parasite maintained in mice (YIUEC) presented higher level of parasitemia and shorter survival times than those infected with a stock (YITC) which was also maintained in mice but had been previously passaged in cell culture. Mouse antibody production tests, performed with the filtered plasma of mice infected with YIUEC, indicated the presence of mouse hepatitis virus (MHV) while no virus was detected when testing the plasma of YITC infected mice. Filtered plasma of YIEUC infected mice was shown to contain a factor able to enhance the level of parasitemia and to reduce the mean survival time of mice challenged with 10(5) YITC. This factor, that could be serially passaged to naïve mice was shown to be a coronavirus by neutralization tests.
Collapse
Affiliation(s)
- H de A Rangel
- Department of Microbiology and Immunology, CEMIB-UNICAMP, Campinas, São Paulo, Brazil
| | | | | | | | | |
Collapse
|
24
|
Barthold SW, Smith AL. Role of host age and genotype in murine enterotropic coronavirus infection. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 1994; 342:371-6. [PMID: 8209756 DOI: 10.1007/978-1-4615-2996-5_57] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Affiliation(s)
- S W Barthold
- Section of Comparative Medicine, Yale University School of Medicine, New Haven, Connecticut 06510
| | | |
Collapse
|
25
|
Décimo D, Boespflug O, Meunier-Rotival M, Hadchouel M, Tardieu M. Genetic restriction of murine hepatitis virus type 3 expression in liver and brain: comparative study in BALB/c and C3H mice by immunochemistry and hybridization in situ. Arch Virol 1993; 130:269-77. [PMID: 8390822 PMCID: PMC7086966 DOI: 10.1007/bf01309659] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
To study the host-dependent genetic variations in murine hepatitis virus type 3 (MHV 3) induced diseases, we localized the sites of MHV 3 (Mill Hill strain) expression within liver and brain by immunohistochemistry or hybridization in situ. Two strains of mice were studied: BALB/c mice, which develop an acute and lethal hepatitis and C3H mice which develop a chronic brain infection. In BALB/c mice, viral RNA and antigens appeared during the first 24h post infection (p.i.) in liver, whereas viral RNA was barely detectable in brain, up until death at day 3 p.i. In C3H mice, viral RNA and antigens were detected simultaneously in liver and brain only at day 2 p.i. In brain, the virus was detected in meningeal and ependymal cells and in perivascular cortical areas (days 5 and 7 p.i.). After day 49, the virus was no longer detected in brain parenchyma, but persisted in meningeal cells. Two host-dependent genetic differences in viral processing were observed in the liver: (1) the virus was first detected in Kupffer cells in BALB/c mice and mostly in hepatocytes in C3H mice; (2) in BALB/c mice, the 180 kDa S viral glycoprotein appeared more frequently cleaved in 90 kDa form than in C3H mice.
Collapse
Affiliation(s)
- D Décimo
- Laboratoire de Neurovirologie et de Neuroimmunologie, Université Paris XI, France
| | | | | | | | | |
Collapse
|
26
|
Casebolt DB, Stephensen CB. Monoclonal antibody solution hybridization assay for detection of mouse hepatitis virus infection. J Clin Microbiol 1992; 30:608-12. [PMID: 1313046 PMCID: PMC265119 DOI: 10.1128/jcm.30.3.608-612.1992] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
A monoclonal antibody solution hybridization (MASH) assay was developed to detect fecal excretion of mouse hepatitis virus (MHV). The assay used a biotinylated cDNA probe to detect viral RNA target sequences by hybridization in solution, capture of hybrids on the solid phase with antibiotin antibody, and immunoassay with an enzyme-labelled monoclonal antibody specific for DNA-RNA hybrids. The MASH assay was used to monitor the time course of enterotropic MHV excretion after oronasal inoculation. Infectivity of the inoculated mice was simultaneously monitored with sentinel animals. The MASH assay detected MHV excretion in all inoculated mice, with the highest mean excretion levels occurring from day 3 through day 9 postinoculation. Mean excretion then decreased gradually to below detection limits by day 21 postinoculation. Sentinels became infected on exposure to inoculated mice up to but not after day 21 postinoculation. Infected sentinel mice showed a time course of virus excretion similar to that of inoculated mice. These results indicate that the MASH assay is useful for rapid, sensitive, and specific detection of MHV in clinical specimens from laboratory mice.
Collapse
Affiliation(s)
- D B Casebolt
- Department of Comparative Medicine, School of Medicine, University of Alabama, Birmingham 35294-0019
| | | |
Collapse
|
27
|
Barthold SW, Smith AL. Viremic dissemination of mouse hepatitis virus-JHM following intranasal inoculation of mice. Arch Virol 1992; 122:35-44. [PMID: 1309644 PMCID: PMC7086624 DOI: 10.1007/bf01321116] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Using a sensitive infant mouse bioassay to detect infectious virus, the pattern of mouse hepatitis virus (MHV) JHM dissemination in blood and other tissues was examined during the first 5 days following intranasal inoculation. MHV replicated in nasal turbinates of both susceptible BALB and resistant SJL mice from days 1 through 5, but BALB mice had higher titers on days 1 and 2. Viremia was detectable on days 1 through 5 in BALB mice, but only on days 3 and 5 in SJL mice. Transient virus replication occurred in the lungs of both mouse genotypes at 1 and 2 days, then ceased. This correlated with more consistently demonstrable virus in blood collected from the left atrium of the heart, compared to jugular vein, portal vein and right atrial blood. Virus was associated equally with the plasma and cellular fractions of blood on day 3, but was primarily in the buffy coat of the cellular fraction on day 5. Interferon-alpha/beta was detected in serum and spleen, but not liver or brain of BALB mice or in any tissue of SJL mice. BALB serum and spleen interferon was first detected at 36 h, peaked between 48 and 72 h, and was undetectable by 108 h. The distribution of virus in nose, cervical, axillary and mesenteric lymph nodes, spleen, Peyer's patch, thymus, bone marrow and liver was examined at 1, 2, and 3 days. The resulting pattern suggested lymphatic spread of virus to cervical lymph node and mesenteric lymph node as pathways of dissemination in addition to viremia.
Collapse
Affiliation(s)
- S W Barthold
- Section of Comparative Medicine, Yale University School of Medicine, New Haven, Connecticut
| | | |
Collapse
|
28
|
Homberger FR, Smith AL, Barthold SW. Detection of rodent coronaviruses in tissues and cell cultures by using polymerase chain reaction. J Clin Microbiol 1991; 29:2789-93. [PMID: 1661745 PMCID: PMC270434 DOI: 10.1128/jcm.29.12.2789-2793.1991] [Citation(s) in RCA: 25] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
A polymerase chain reaction (PCR) method was developed for the detection of rodent coronaviruses in biological material by using reverse transcriptase and two primers which flanked an M gene sequence of 375 bp. PCR detected all of 11 different strains of mouse hepatitis virus (MHV) as well as rat sialodacryoadenitis virus but not bovine coronavirus or human coronavirus strains OC43 and 229E. The M gene sequences of bovine coronavirus and human coronavirus OC43 are homologous to that of MHV, but minor differences exist in the primer regions, preventing annealing of the primers. For detecting MHV-Y in tissue samples, PCR was faster than and at least as sensitive as either of the two bioassays (infant mouse bioassay and mouse antibody production test) currently used for MHV diagnostic purposes.
Collapse
Affiliation(s)
- F R Homberger
- Section of Comparative Medicine, Yale University School of Medicine, New Haven, Connecticut 06510
| | | | | |
Collapse
|
29
|
Williams RK, Jiang GS, Snyder SW, Frana MF, Holmes KV. Purification of the 110-kilodalton glycoprotein receptor for mouse hepatitis virus (MHV)-A59 from mouse liver and identification of a nonfunctional, homologous protein in MHV-resistant SJL/J mice. J Virol 1990; 64:3817-23. [PMID: 2164599 PMCID: PMC249677 DOI: 10.1128/jvi.64.8.3817-3823.1990] [Citation(s) in RCA: 100] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The receptor for mouse hepatitis virus strain A59 (MHV-A59) is a 110- to 120-kilodalton (kDa) glycoprotein which is expressed in MHV-susceptible mouse strains on the membranes of hepatocytes, intestinal epithelial cells, and macrophages. SJL/J mice, which are highly resistant to MHV-A59, were previously shown to lack detectable levels of receptor by using either solid-phase virus receptor assays or binding of a monoclonal anti-receptor antibody (MAb) which blocks infection of MHV-susceptible mouse cells. This MAb was used for affinity purification of the receptor glycoprotein from livers of MHV-susceptible Swiss Webster mice. The MHV receptor and an antigenically related protein of 48 to 58 kDa were copurified and then separated by preparative sodium dodecyl sulfate-polyacrylamide gel electrophoresis. The first 15 amino acids of the receptor were sequenced, and a synthetic peptide of this amino acid sequence was prepared. Rabbit antiserum made against this peptide bound to the MHV receptor glycoprotein and the 48- to 58-kDa protein from livers of MHV-susceptible BALB/c mice and Swiss Webster mice and from the intestinal brush border of BALB/c mice. In immunoblots of intestinal brush border and hepatocyte membranes of MHV-resistant SJL/J mice, the antibody against the amino terminus of the receptor identified proteins that are 5 to 10 kDa smaller than the MHV receptor and the 48- to 58-kDa related protein from Swiss Webster or BALB/c mice. Thus, SJL/J mice express a protein which shares some sequence homology with the MHV receptor but which lacks virus-binding activity and is not recognized by the blocking anti-receptor MAb. These results suggest that resistance of SJL/J mice to MHV-A59 may be due to absence or mutation of the virus-binding domain in the nonfunctional receptor homolog in SJL/J mice.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Antibodies, Monoclonal
- Cell Transformation, Viral
- Cells, Cultured
- Disease Susceptibility
- Female
- Hepatitis, Viral, Animal/immunology
- Intestines/immunology
- Liver/microbiology
- Membrane Glycoproteins/genetics
- Membrane Glycoproteins/immunology
- Membrane Glycoproteins/isolation & purification
- Mice
- Mice, Inbred BALB C
- Mice, Inbred Strains
- Microvilli/immunology
- Molecular Weight
- Murine hepatitis virus/physiology
- Receptors, Virus/genetics
- Receptors, Virus/immunology
- Receptors, Virus/isolation & purification
Collapse
Affiliation(s)
- R K Williams
- Department of Pathology, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20814
| | | | | | | | | |
Collapse
|
30
|
Sturman LS, Ricard CS, Holmes KV. Conformational change of the coronavirus peplomer glycoprotein at pH 8.0 and 37 degrees C correlates with virus aggregation and virus-induced cell fusion. J Virol 1990; 64:3042-50. [PMID: 2159562 PMCID: PMC249489 DOI: 10.1128/jvi.64.6.3042-3050.1990] [Citation(s) in RCA: 108] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
We have obtained biochemical and electron microscopic evidence of conformational changes at pH 8.0 and 37 degrees C in the coronavirus spike glycoprotein E2 (S). The importance of these changes is reflected in the loss of virus infectivity, the aggregation of virions, and increased virus-induced cell fusion at the same pH. Coronavirus (MHV-A59) infectivity is exquisitely sensitive to pH. The virus was quite stable at pH 6.0 and 37 degrees C (half-life, approximately 24 h) but was rapidly and irreversibly inactivated by brief treatment at pH 8.0 and 37 degrees C (half-life, approximately 30 min). Virions treated at pH 8.0 and 37 degrees C formed clumps and large aggregates. With virions treated at pH 8.0 and 37 degrees C, the amino-terminal peptide E2N (or S1) was released from virions and the remaining peptide, E2C (S2), was aggregated. Viral spikes isolated from detergent-treated virions also aggregated at pH 8.0 and 37 degrees C. Loss of virus infectivity and E2 (S) aggregation at pH 8.0 and 37 degrees C were markedly enhanced in the presence of dithiothreitol. On the basis of the effects of dithiothreitol on the reactions of the peplomer, we propose that release of E2N (S1) and aggregation of E2C (S2) may be triggered by rearrangement of intramolecular disulfide bonds. The aggregation of virions and the isolated E2 (S) glycoprotein at pH 8.0 and 37 degrees C or following treatment with guanidine and urea at pH 6.0 and 37 degrees C indicate that an irreversible conformational change has been induced in the peplomer glycoprotein by these conditions. It is interesting that coronavirus-induced cell fusion also occurred under mildly alkaline conditions and at 37 degrees C. Some enveloped viruses, including influenza viruses and alphaviruses, show conformational changes of spike glycoproteins at a low pH, which correlates with fusion and penetration of those viruses in acidified endocytic vesicles. For coronavirus MHV-A59, comparable conformational change of the spike glycoprotein E2 (S) and cell fusion occurred at a mildly alkaline condition, suggesting that coronavirus infection-penetration, like that of paramyxoviruses and lentiviruses, may occur at the plasma membrane, rather than within endocytic vesicles.
Collapse
Affiliation(s)
- L S Sturman
- Wadsworth Center for Laboratories and Research, New York State Department of Health, Albany
| | | | | |
Collapse
|
31
|
Holmes KV, Williams RK, Cardellichio CB, Compton SR, Stephensen CB, Snyder SW, Frana MF, Jiang GS, Smith A, Knobler RL. Is the 110K glycoprotein the only receptor for MHV and does its expression determine species specificity? ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 1990; 276:37-44. [PMID: 1966425 DOI: 10.1007/978-1-4684-5823-7_6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- K V Holmes
- Uniformed Services University of the Health Sciences, Bethesda, MD 20814
| | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Abstract
The resistance of immunized mice to challenge with the same or a different strain of mouse hepatitis virus (MHV) was examined as a model of challenge immunity to coronavirus infection. Genetically susceptible BALB/cByJ mice were given an intranasal immunizing infection of respiratory-type MHV-JHM, MHV-S, or enterotropic MHV-Y. Control mice were sham-immunized with sterile tissue culture fluid. Recovered mice were challenged intranasally with MHV-JHM, MHV-S or sterile tissue culture fluid at 30 days after immunization. Resistance to challenge inoculation was evaluated in groups of mice at 4 and 30 days after challenge. At 4 days, the prevalence of MHV lesions in nose and liver was tabulated and MHV titers in liver were determined. At 30 days, the prevalence of residual brainstem spongiform lesions was tabulated and serum antibody to MHV-JHM and MVH-S was quantified by enzyme immunoassay. Mice immunized with MHV-JHM or MHV-S resisted challenge with the MHV homotype, but MHV-S-immunized mice were fully susceptible to challenge with MVH-JHM. Mice immunized with enterotropic MHV-Y were only partially protected against challenge with antigenically related, but biologically different MHV-S. Serum antibody responses to MHV supported these observations. These data indicate that challenge immunity to coronaviruses is strong, but highly virus strain-specific.
Collapse
Affiliation(s)
- S W Barthold
- Section of Comparative Medicine, Yale University School of Medicine, New Haven, Connecticut
| | | |
Collapse
|
33
|
Hirasawa T, Hirano N, Ohhara S, Mikazuki K, Hayashi Y. Characterization of low-virulent mouse coronavirus isolated from faeces in a mouse colony. ZENTRALBLATT FUR VETERINARMEDIZIN. REIHE B. JOURNAL OF VETERINARY MEDICINE. SERIES B 1988; 35:435-42. [PMID: 2847448 DOI: 10.1111/j.1439-0450.1988.tb00517.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
34
|
La Regina MC, Lonigro J, Woods L, Hall WC, Doyle RE. Comparison of Track XI fluorometric immunoassay with Bio-EnzaBead enzyme-linked immunosorbent assay for detection of serum antibody to mouse hepatitis virus. J Clin Microbiol 1988; 26:573-5. [PMID: 2833531 PMCID: PMC266335 DOI: 10.1128/jcm.26.3.573-575.1988] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
The Track XI system (Microbiological Associates, Bethesda, Md.) was compared with the Bio-EnzaBead assay (Organon Teknika, Durham, N.C.) for the detection of antibody to mouse hepatitis virus (MHV). Strain A/J mice were inoculated intranasally with MHV type 3. Sera were collected at 1, 2, 4, and 9 weeks postinoculation and tested. Individual serum samples were retested twice by each method. The results suggested that the Track XI system was more sensitive and reliable than the Bio-EnzaBead assay in detecting antibody to MHV type 3 in individual serum samples from A/J mice.
Collapse
Affiliation(s)
- M C La Regina
- Department of Comparative Medicine, St. Louis University Medical School, Missouri 63104
| | | | | | | | | |
Collapse
|
35
|
|
36
|
Barthold SW, Beck DS, Smith AL. Mouse hepatitis virus and host determinants of vertical transmission and maternally-derived passive immunity in mice. Arch Virol 1988; 100:171-83. [PMID: 2840871 PMCID: PMC7086610 DOI: 10.1007/bf01487681] [Citation(s) in RCA: 28] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Transmission of mouse hepatitis virus (MHV) in utero following oronasal inoculation of pregnant mice was found to depend upon MHV strain and host genotype. Virulent, polytropic MHV-JHM was recovered from multiple maternal tissues, including liver and uterus, as well as placenta and fetus in susceptible BALB/cByJ mice. Fetuses were infected during all 3 trimesters of pregnancy. Low virulence, polytropic MHV-S infected fetuses in a low percentage of susceptible BALB/cByJ dams. Infection of resistant CD-1 mice with MHV-JHM was limited, with no fetal infection. Enterotropic MHV-Y was largely restricted to intestine of BALB/cByJ and CD-1 dams, with minimal dissemination and no fetal infection. Maternally-derived MHV IgG antibody was detectable in pup sera through 4 weeks of age. Antibody titers were generally lower in second litters of the same dam. Cross-fostering experiments showed that antibody was transferred via colostrum and not in utero, and that pups were capable of absorption through 2 weeks of age. Pups nursing immune dams were protected against MHV challenge at 1 and 2 weeks of age, compared to pups nursing naive dams. Immunity to MHV challenge was cross-protective against both antigenically homotypic and heterotypic strains of MHV.
Collapse
MESH Headings
- Animals
- Antibodies, Viral/analysis
- Colostrum/immunology
- Female
- Fetal Diseases/microbiology
- Hepatitis, Viral, Animal/immunology
- Hepatitis, Viral, Animal/microbiology
- Hepatitis, Viral, Animal/transmission
- Immunity, Maternally-Acquired
- Immunoglobulin G/analysis
- Male
- Maternal-Fetal Exchange
- Mice
- Mice, Inbred BALB C
- Murine hepatitis virus/immunology
- Murine hepatitis virus/isolation & purification
- Pregnancy
- Pregnancy Complications, Infectious/microbiology
- Uterus/microbiology
Collapse
Affiliation(s)
- S W Barthold
- Section of Comparative Medicine, Yale University School of Medicine, New Haven, Connecticut
| | | | | |
Collapse
|
37
|
Abstract
The mechanism of brain infection with mouse hepatitis virus-JHM was studied in BALB/cByJ mice following intranasal inoculation, and found to be a consequence of direct viral spread along olfactory nerves into olfactory bulbs of the brain. Infection was followed sequentially from nose to brain, using microscopy, immunohistochemistry and virus quantification. Lesions, antigen and virus were observed in the olfactory bulb and anterior brain as early as 2 days and posterior brain by 4 days after inoculation. Viral antigen extended through nasal mucosa into submucosa, then coursed along the olfactory nerve perineurium and fibers, through the cribriform plate into the olfactory bulbs. On days 4 and 7, viral antigen was found in the antero-ventral brain, along ventral meninges, olfactory tracts and anterior ramifications of the lateral ventricles. Virus was cleared from nose by 10 days and anterior brain by 20 days, but persisted in posterior brain for 20 days after inoculation. Mice also developed disseminated infection, with viremia and hepatitis. Infection of brain did not correlate with presence of viremia. In contrast to intranasally inoculated mice, orally-inoculated mice did not develop encephalitis, despite evidence of disseminated infection.
Collapse
Affiliation(s)
- S W Barthold
- Section of Comparative Medicine, Yale University School of Medicine, New Haven, CT 06510
| |
Collapse
|
38
|
Smith AL, Barthold SW, Beck DS. Intranasally administered alpha/beta interferon prevents extension of mouse hepatitis virus, strain JHM, into the brains of BALB/cByJ mice. Antiviral Res 1987; 8:239-45. [PMID: 2837142 PMCID: PMC7134092 DOI: 10.1016/s0166-3542(87)80002-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Intranasally administered alpha/beta interferon blocked extension of the coronavirus, mouse hepatitis virus, strain JHM (MHV-JHM), from the nose to the brain of BALB/cByJ mice following intranasal inoculation with the virus. Two hundred units of alpha/beta interferon were administered intranasally to BALB/cByJ mice daily over a five day period. The mice were exposed intranasally to 10(3) median tissue culture infectious doses of MHV-JHM on the third day of interferon treatment. Two days after virus exposure, the proportion of mice with MHV in nasal turbinates was reduced from 10 of 10 in the untreated group to 7 of 10 in the interferon-treated group, and mean titers in virus-containing noses were lower in the interferon-treated group. Five days after virus exposure, the proportion of mice with infectious virus in the brain was significantly lower in the interferon-treated group (1 of 10 mice) than in the untreated group (10 of 10 mice). Systemic infection, as measured by presence and concentration of virus in the spleen, was not affected by intranasal interferon treatment. These results suggest that intranasally administered interferon protects against local extension of MHV-JHM from nose to brain, but not against dissemination of virus to other organs, such as the spleen.
Collapse
Affiliation(s)
- A L Smith
- Section of Comparative Medicine, Yale University School of Medicine, New Haven, Connecticut 06510
| | | | | |
Collapse
|
39
|
Barthold SW, Smith AL. Response of genetically susceptible and resistant mice to intranasal inoculation with mouse hepatitis virus JHM. Virus Res 1987; 7:225-39. [PMID: 3037819 PMCID: PMC7134095 DOI: 10.1016/0168-1702(87)90030-x] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/1986] [Accepted: 01/07/1987] [Indexed: 01/03/2023]
Abstract
Mouse hepatitis virus (MHV)-JHM infection was studied in genetically susceptible (BALB/cByJ) and resistant (SJL/J) mice following intranasal inoculation at 1, 3, 6 or 12 wk of age. Markers of infection included histology, immunohistochemistry, virus quantification and virus serology. All BALB mice developed severe disseminated disease with high mortality due to encephalitis and hepatitis. Peak MHV titers appeared in brain, liver, spleen and intestine on days 3 or 5. Age at inoculation did not influence virus titers in brain, spleen or intestine, but virus titers in liver were inversely proportional to age at inoculation. In 6-wk-old BALB mice, virus was cleared from spleen, intestine and liver by day 30 and from brain by day 60. In intestine, MHV was localized to lymphoid tissue, without fecal excretion. SJL mice of all ages developed remarkably milder disease with low mortality occurring only among mice inoculated at 1 wk of age. SJL mice inoculated at 1 wk had disseminated infection at day 3, but lesions and antigen were cleared from most organs by day 5. Mice inoculated at 3 and 6 wk of age had minimal or no involvement of peripheral organs, and mice inoculated at 12 wk of age had infections restricted to the nose. At day 5, MHV titers in brain, liver, spleen and intestine were significantly lower or undetectable in SJL mice of all ages compared to age-matched BALB mice. In 6-wk-old mice, MHV was cleared from all organs by day 10. Serum antibody titers to MHV were many-fold higher in BALB mice, compared to SJL mice, which mounted only a modest response.
Collapse
|
40
|
Boyle JF, Weismiller DG, Holmes KV. Genetic resistance to mouse hepatitis virus correlates with absence of virus-binding activity on target tissues. J Virol 1987; 61:185-9. [PMID: 3023696 PMCID: PMC255233 DOI: 10.1128/jvi.61.1.185-189.1987] [Citation(s) in RCA: 127] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
The molecular mechanism of genetic resistance of inbred mouse strains to mouse hepatitis virus, a murine coronavirus, was studied by comparing virus binding to plasma membranes of intestinal epithelium or liver from susceptible BALB/c and resistant SJL/J mice with a new solid-phase assay for virus-binding activity. Virus bound to isolated membranes from susceptible mice, but not to membranes from resistant mice. F1 progeny of SJL/J X BALB/c mice had an intermediate level of virus-binding activity on their enterocyte and hepatocyte membranes. This correlated well with previous studies showing that susceptibility to mouse hepatitis virus strain A59 is controlled by a single autosomal dominant gene (M. S. Smith, R. E. Click, and P. G. W. Plagemann, J. Immunol. 133:428-432). Because virus binding was not prevented by treating membranes with sodium dodecyl sulfate, the virus-binding molecule could be identified by a virus overlay protein blot assay. Virus bound to a single broad band of Mr 100,000 to 110,000 in membranes from hepatocytes or enterocytes of susceptible BALB/c and semisusceptible C3H mice, but no virus-binding band was detected in comparable preparations of resistant SJL/J mouse membranes. Therefore, SJL/J mice may be resistant to mouse hepatitis virus A59 infection because they lack a specific virus receptor which is present on the plasma membranes of target cells from genetically susceptible BALB/c and semisusceptible C3H mice.
Collapse
|
41
|
Lecomte J, Cainelli-Gebara V, Mercier G, Mansour S, Talbot PJ, Lussier G, Oth D. Protection from mouse hepatitis virus type 3-induced acute disease by an anti-nucleoprotein monoclonal antibody. Brief report. Arch Virol 1987; 97:123-30. [PMID: 2825619 PMCID: PMC7086664 DOI: 10.1007/bf01310740] [Citation(s) in RCA: 53] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Fusion of MHV-3-immune splenocytes from MHV-3-resistant A/J murine strain, with NS myeloma cells produced several hybridomas. Among eight hybridoma clones, the 1E7A4H1 clone secreted kappa IgG2a apparently directed against the nucleoprotein of the MHV-3 virion. The monoclonal antibody was able to neutralize the in vitro cytopathic effect of MHV-3 on cultured L2 cells, and was detected by indirect immunofluorescence on MHV-3-infected cultured YAC cells. In addition, it conferred a significant protection against MHV-3-induced acute disease, if injected intraperitoneally to C57BL/6 mice before inoculation with MHV-3.
Collapse
Affiliation(s)
- J Lecomte
- Institut Armand-Frappier, Centre de recherche en virologie, Québec, Canada
| | | | | | | | | | | | | |
Collapse
|
42
|
Carman PS, Ernst PB, Rosenthal KL, Clark DA, Befus DA, Bienenstock J. Natural killer (NK) cell activity against enteric murine coronavirus mediated by intestinal leukocytes. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 1987; 216A:533-7. [PMID: 2825474 DOI: 10.1007/978-1-4684-5344-7_63] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
- P S Carman
- McMaster University, Hamilton, Ontario, Canada
| | | | | | | | | | | |
Collapse
|
43
|
Holmes KV, Boyle JF, Weismiller DG, Compton SR, Williams RK, Stephensen CB, Frana MF. Identification of a receptor for mouse hepatitis virus. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 1987; 218:197-202. [PMID: 2829528 DOI: 10.1007/978-1-4684-1280-2_24] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
- K V Holmes
- Department of Pathology, Uniformed Services University of the Health Sciences, Bethesda, MD 20814
| | | | | | | | | | | | | |
Collapse
|
44
|
Smith AL. SEROLOGIC TESTS FOR DETECTION OF ANTIBODY TO RODENT VIRUSES. VIRAL AND MYCOPLASMAL OF LABORATORY RODENTS 1986. [PMCID: PMC7155483 DOI: 10.1016/b978-0-12-095785-9.50041-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
|
45
|
Barthold SW, Beck DS, Smith AL. Mouse hepatitis virus nasoencephalopathy is dependent upon virus strain and host genotype. Arch Virol 1986; 91:247-56. [PMID: 3022679 PMCID: PMC7087084 DOI: 10.1007/bf01314284] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/1985] [Accepted: 03/21/1986] [Indexed: 01/03/2023]
Abstract
Mouse hepatitis virus (MHV) S induced typical MHV spongiform lesions in brainstem 28 days following intranasal inoculation of adult A/J, BALB/cByJ, CBA/J, C 3 H/HeJ and C 3 H/RV, but not SJL mice. In all but SJL mice, brain lesions occurred at or near the infectious dose level, based on seroconversion by the indirect immunofluorescence assay. During the acute phase of infection (day 5), lesions were limited to the nose and brain in most genotypes. Exceptions were BALB mice, which had mild hepatitis and SJL mice, which had lesions restricted to the nose. No mortality occurred in any genotype. Following intranasal inoculation of adult mice, MHV-1, -3, -A 59, -JHM and -S all caused brain lesions at 28 days after inoculation. MHV-1 and -3 caused lesions that were usually restricted to the anterior olfactory tracts, while MHV-A 59, -S and -JHM also caused more generalized and pronounced lesions involving the midbrain and pons. These studies suggest that avirulent MHV-S given intranasally to most mouse genotypes is a good model for induction of brain infection in the absence of mortality. They also confirm observations made by others in which MHV-JHM, -S and -A 59 are relatively more neurotropic than other MHV strains, such as MHV-1 and -3.
Collapse
|
46
|
|
47
|
Frana MF, Behnke JN, Sturman LS, Holmes KV. Proteolytic cleavage of the E2 glycoprotein of murine coronavirus: host-dependent differences in proteolytic cleavage and cell fusion. J Virol 1985; 56:912-20. [PMID: 2999444 PMCID: PMC252664 DOI: 10.1128/jvi.56.3.912-920.1985] [Citation(s) in RCA: 174] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Cell fusion induced by infection with mouse hepatitis virus strain A59 (MHV-A59) varied markedly in extent and time course in four different murine cell lines. When inoculated at a multiplicity of 3 to 5 PFU per cell, the Sac-, L2, and DBT cell lines began to fuse by 7 h, were fused into confluent syncytia by 9 to 12 h, and peeled from the substrate by 10 to 14 h. These virulent virus-cell interactions were in striking contrast to the moderate interaction of MHV-A59 with the 17 Cl 1 cell line, in which only small syncytia were observed 18 h postinoculation, and greater than 50% of the cells remained unfused by 24 h. The yield of infectious virus produced by 17 Cl 1 cells was 10-fold higher than the yields from the other three cell lines. The processing of the nucleocapsid protein, the membrane glycoprotein E1, and the peplomeric glycoprotein E2 were found to differ significantly in the four cell lines. Since the E2 glycoprotein is responsible for virus-induced cell fusion, we attempted to correlate differences in cellular processing of E2 with differences in fusion of infected cells. The predominant intracellular form of E2 in all cell lines was the 180K species. Pulse-chase experiments showed that a small portion of the 17 Cl 1 cell-associated 180K E2 was cleaved by 1 h after synthesis to yield 90K E2, shown in the preceding paper to consist of two different glycoproteins called 90A and 90B (L. S. Sturman, C. S. Ricard, and K. V. Holmes, J. Virol. 56:904-911, 1985). This cleavage occurred shortly before the release of virions from cells, as shown by pulse-chase experiments. After budding at intracellular membranes, virions released into the medium by the four cell lines contained different ratios of 180K to 90K E2. Virions from Sac- cells, which contained 100% 90K E2, fused L2 cells rapidly without requiring virus replication, whereas virions from 17 Cl 1 cells, which had 50% 90K E2, required trypsin activation to induce rapid fusion (Sturman et al., J. Virol. 56:904-911, 1985). The addition of protease inhibitors to the medium markedly delayed L2 cell fusion induced by MHV infection. The extent of coronavirus-induced cell fusion does not depend solely upon the percent cleavage of the E2 glycoprotein by cellular proteases, since extensive fusion was induced by infection of L2 and DBT cells but not 17 Cl 1 cells, although all three cell lines cleaved E2 to the same extent.(ABSTRACT TRUNCATED AT 400 WORDS)
Collapse
|
48
|
Chatterjee NK, Haley TM, Nejman C. Functional alterations in pancreatic beta cells as a factor in virus-induced hyperglycemia in mice. J Biol Chem 1985. [DOI: 10.1016/s0021-9258(17)38946-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
|
49
|
|
50
|
Mouse Hepatitis Virus Infection, Intestine, Mouse. ACTA ACUST UNITED AC 1985. [DOI: 10.1007/978-3-642-96910-2_55] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
|