1
|
Zhao J, Xiao P, Xin A, Zhu H, Wang H, Xiao J, Gao H. Preliminary evaluation of a novel serotype O foot-and-mouth disease mRNA vaccine. Front Microbiol 2025; 16:1503191. [PMID: 40356647 PMCID: PMC12067417 DOI: 10.3389/fmicb.2025.1503191] [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: 09/28/2024] [Accepted: 04/07/2025] [Indexed: 05/15/2025] Open
Abstract
Foot-and-mouth disease virus (FMDV) is one of the most significant animal pathogens worldwide, severely impacting the health and productivity of pigs, cattle, sheep, and other ungulates. Although the traditional vaccines have played a crucial role in epidemic control, inactivated vaccines face persistent challenges concerning the potential for virus dissemination and pressures from serotype and subtype matching. However, the manufacture of attenuated vaccines is forbidden, and the efficiency of alternative vaccines for immune protection is still inadequate. Consequently, there exists an urgent need for safer and more effective innovative vaccines in animal husbandry. In this study, we aimed to develop a lipid nanoparticle mRNA vaccine based on VP1-3A-3D epitopes from serotype O FMD and to verify its specific expression within cytoplasmic and injection sites. Our findings demonstrated that mRNA transfected into primary spleen cells derived from guinea pigs induced cytokine release, promoted differentiation of both CD4+ T and CD8+ T lymphocytes, and enhanced lymphocyte proliferation rates. Following immunization of mRNA vaccine in guinea pigs, we observed increased differentiation of both CD4+ T and CD8+ T cells, alongside elevated levels of cytokine secretion. Additionally, this vaccination induced the production of specific IgG antibodies as well as neutralizing antibodies. Importantly, our vaccine provided complete protection for all six guinea pigs against a lethal challenge of 100 GPID50, with histopathological scores indicating protection equivalent to that conferred by the inactivated vaccine. The viral load results demonstrated that the vaccine group significantly reduced viral copy numbers in serum and effectively decreased the concentration of the inflammatory cytokine IL-1β. Furthermore, during the pre-immune phase following vaccination with the mRNA vaccine in pigs, heightened cytokine secretion was observed, along with the inhibition of viral replication. Simultaneously, the neutralizing antibody titer in the serum remained stable over 4 months. Immunofluorescence analysis of spleen tissues from both guinea pigs and pigs demonstrated marked activation and increased expression of CD4+ and CD8+ T lymphocytes, as well as macrophages, in the mRNA vaccine group. In summary, this study suggests that the serotype O FMD mRNA vaccine is a promising candidate for further development in the fight against FMDV.
Collapse
Affiliation(s)
- Jingang Zhao
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming, China
- Yunnan Tropical and Subtropical Animal Virus Disease Laboratory, Yunnan Academy of Animal Husbandry and Veterinary Sciences, Kunming, China
| | - Peng Xiao
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming, China
| | - Aiguo Xin
- Yunnan Tropical and Subtropical Animal Virus Disease Laboratory, Yunnan Academy of Animal Husbandry and Veterinary Sciences, Kunming, China
- National Foot-and-Mouth Disease Para-Reference Laboratory (Kunming), Yunnan Academy of Animal Husbandry and Veterinary Sciences, Kunming, China
| | - Heran Zhu
- Yunnan Tropical and Subtropical Animal Virus Disease Laboratory, Yunnan Academy of Animal Husbandry and Veterinary Sciences, Kunming, China
- National Foot-and-Mouth Disease Para-Reference Laboratory (Kunming), Yunnan Academy of Animal Husbandry and Veterinary Sciences, Kunming, China
| | - Hao Wang
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming, China
| | - Jinlong Xiao
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming, China
| | - Hong Gao
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming, China
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming, China
| |
Collapse
|
2
|
Hickman HD, Moutsopoulos NM. Viral infection and antiviral immunity in the oral cavity. Nat Rev Immunol 2025; 25:235-249. [PMID: 39533045 DOI: 10.1038/s41577-024-01100-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/02/2024] [Indexed: 11/16/2024]
Abstract
Individual tissues have distinct antiviral properties garnered through various mechanisms, including physical characteristics, tissue-resident immune cells and commensal organisms. Although the oral mucosa has long been appreciated as a critical barrier tissue that is exposed to a continuous barrage of pathogens, many fundamental aspects of the antiviral immune response in this tissue remain unknown. Several viral pathogens, such as herpesviruses and human papillomaviruses, have been acknowledged both historically and at present for infections in the oral cavity that result in substantial clinical burden. However, recent viral outbreaks, including those with SARS-CoV-2 and mpox, featured oral symptoms even though these viruses are not generally considered oral pathogens. Ensuing studies have shown that the oral cavity is an important locale for viral infection and potential transmission of newly emergent or re-emergent pathogens, highlighting the need for an increased understanding of the mechanisms of antiviral immunity at this site. In this Review, we provide a broad overview of antiviral immune responses in the oral cavity and discuss common viral infections and their manifestations in the oral mucosa. In addition, we present current mouse models for the study of oral viral infections.
Collapse
Affiliation(s)
- Heather D Hickman
- Viral Immunity and Pathogenesis Unit, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| | - Niki M Moutsopoulos
- Oral Immunity and Infection Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
3
|
Byrne CM, Márquez AC, Cai B, Coombs D, Gantt S. Spatial kinetics and immune control of murine cytomegalovirus infection in the salivary glands. PLoS Comput Biol 2024; 20:e1011940. [PMID: 39150988 DOI: 10.1371/journal.pcbi.1011940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 08/28/2024] [Accepted: 07/30/2024] [Indexed: 08/18/2024] Open
Abstract
Human cytomegalovirus (HCMV) is the most common congenital infection. Several HCMV vaccines are in development, but none have yet been approved. An understanding of the kinetics of CMV replication and transmission may inform the rational design of vaccines to prevent this infection. The salivary glands (SG) are an important site of sustained CMV replication following primary infection and during viral reactivation from latency. As such, the strength of the immune response in the SG likely influences viral dissemination within and between hosts. To study the relationship between the immune response and viral replication in the SG, and viral dissemination from the SG to other tissues, mice were infected with low doses of murine CMV (MCMV). Following intra-SG inoculation, we characterized the viral and immunological dynamics in the SG, blood, and spleen, and identified organ-specific immune correlates of protection. Using these data, we constructed compartmental mathematical models of MCMV infection. Model fitting to data and analysis indicate the importance of cellular immune responses in different organs and point to a threshold of infection within the SG necessary for the establishment and spread of infection.
Collapse
Affiliation(s)
- Catherine M Byrne
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
| | - Ana Citlali Márquez
- British Columbia Centre for Disease Control, Vancouver, British Columbia, Canada
| | - Bing Cai
- British Columbia Children's Hospital Research Institute, Vancouver, British Columbia, Canada
| | - Daniel Coombs
- Department of Mathematics, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Soren Gantt
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, Québec, Canada
| |
Collapse
|
4
|
Pinilla YT, Heinzel C, Caminada LF, Consolaro D, Esen M, Kremsner PG, Held J, Kreidenweiss A, Fendel R. SARS-CoV-2 Antibodies Are Persisting in Saliva for More Than 15 Months After Infection and Become Strongly Boosted After Vaccination. Front Immunol 2021; 12:798859. [PMID: 34956236 PMCID: PMC8695841 DOI: 10.3389/fimmu.2021.798859] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 11/15/2021] [Indexed: 01/15/2023] Open
Abstract
SARS-CoV-2 antibodies in saliva serve as first line of defense against the virus. They are present in the mucosa, more precisely in saliva, after a recovered infection and also following vaccination. We report here the antibody persistence in plasma and in saliva up to 15 months after mild COVID-19. The IgG antibody response was measured every two months in 72 participants using an established and validated in-house ELISA assay. In addition, the virus inhibitory activity of plasma antibodies was assessed in a surrogate virus neutralization test before and after vaccination. SARS-CoV-2-specific antibody concentrations remained stable in plasma and saliva and the response was strongly boosted after one dose COVID-19 vaccination.
Collapse
Affiliation(s)
- Yudi T Pinilla
- Institute of Tropical Medicine, University Hospital Tübingen, Tübingen, Germany.,German Center for Infection Research (DZIF), Partner Site Tübingen, Tübingen, Germany
| | - Constanze Heinzel
- Institute of Tropical Medicine, University Hospital Tübingen, Tübingen, Germany.,German Center for Infection Research (DZIF), Partner Site Tübingen, Tübingen, Germany
| | | | - Douglas Consolaro
- Institute of Tropical Medicine, University Hospital Tübingen, Tübingen, Germany.,German Center for Infection Research (DZIF), Partner Site Tübingen, Tübingen, Germany
| | - Meral Esen
- Institute of Tropical Medicine, University Hospital Tübingen, Tübingen, Germany.,German Center for Infection Research (DZIF), Partner Site Tübingen, Tübingen, Germany.,Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon.,Cluster of Excellence: Exzellenzcluster (EXC) 2124: Controlling Microbes to Fight Infection, Tübingen, Germany
| | - Peter G Kremsner
- Institute of Tropical Medicine, University Hospital Tübingen, Tübingen, Germany.,German Center for Infection Research (DZIF), Partner Site Tübingen, Tübingen, Germany.,Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
| | - Jana Held
- Institute of Tropical Medicine, University Hospital Tübingen, Tübingen, Germany.,German Center for Infection Research (DZIF), Partner Site Tübingen, Tübingen, Germany.,Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
| | - Andrea Kreidenweiss
- Institute of Tropical Medicine, University Hospital Tübingen, Tübingen, Germany.,German Center for Infection Research (DZIF), Partner Site Tübingen, Tübingen, Germany.,Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
| | - Rolf Fendel
- Institute of Tropical Medicine, University Hospital Tübingen, Tübingen, Germany.,German Center for Infection Research (DZIF), Partner Site Tübingen, Tübingen, Germany.,Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
| |
Collapse
|
5
|
Mandal P, Nagrani LN, Hernandez L, McCormick AL, Dillon CP, Koehler HS, Roback L, Alnemri ES, Green DR, Mocarski ES. Multiple Autonomous Cell Death Suppression Strategies Ensure Cytomegalovirus Fitness. Viruses 2021; 13:v13091707. [PMID: 34578288 PMCID: PMC8473406 DOI: 10.3390/v13091707] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 08/13/2021] [Accepted: 08/20/2021] [Indexed: 12/31/2022] Open
Abstract
Programmed cell death pathways eliminate infected cells and regulate infection-associated inflammation during pathogen invasion. Cytomegaloviruses encode several distinct suppressors that block intrinsic apoptosis, extrinsic apoptosis, and necroptosis, pathways that impact pathogenesis of this ubiquitous herpesvirus. Here, we expanded the understanding of three cell autonomous suppression mechanisms on which murine cytomegalovirus relies: (i) M38.5-encoded viral mitochondrial inhibitor of apoptosis (vMIA), a BAX suppressor that functions in concert with M41.1-encoded viral inhibitor of BAK oligomerization (vIBO), (ii) M36-encoded viral inhibitor of caspase-8 activation (vICA), and (iii) M45-encoded viral inhibitor of RIP/RHIM activation (vIRA). Following infection of bone marrow-derived macrophages, the virus initially deflected receptor-interacting protein kinase (RIPK)3-dependent necroptosis, the most potent of the three cell death pathways. This process remained independent of caspase-8, although suppression of this apoptotic protease enhances necroptosis in most cell types. Second, the virus deflected TNF-mediated extrinsic apoptosis, a pathway dependent on autocrine TNF production by macrophages that proceeds independently of mitochondrial death machinery or RIPK3. Third, cytomegalovirus deflected BCL-2 family protein-dependent mitochondrial cell death through combined TNF-dependent and -independent signaling even in the absence of RIPK1, RIPK3, and caspase-8. Furthermore, each of these cell death pathways dictated a distinct pattern of cytokine and chemokine activation. Therefore, cytomegalovirus employs sequential, non-redundant suppression strategies to specifically modulate the timing and execution of necroptosis, extrinsic apoptosis, and intrinsic apoptosis within infected cells to orchestrate virus control and infection-dependent inflammation. Virus-encoded death suppressors together hold control over an intricate network that upends host defense and supports pathogenesis in the intact mammalian host.
Collapse
Affiliation(s)
- Pratyusha Mandal
- Emory Vaccine Center, Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA 30322, USA; (L.H.); (H.S.K.); (L.R.)
- Correspondence: (P.M.); (E.S.M.); Tel.: +404-727-0563 (P.M.); +404-727-4273 (E.S.M.)
| | | | - Liliana Hernandez
- Emory Vaccine Center, Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA 30322, USA; (L.H.); (H.S.K.); (L.R.)
| | | | | | - Heather S. Koehler
- Emory Vaccine Center, Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA 30322, USA; (L.H.); (H.S.K.); (L.R.)
| | - Linda Roback
- Emory Vaccine Center, Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA 30322, USA; (L.H.); (H.S.K.); (L.R.)
| | - Emad S. Alnemri
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA;
| | - Douglas R. Green
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA;
| | - Edward S. Mocarski
- Emory Vaccine Center, Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA 30322, USA; (L.H.); (H.S.K.); (L.R.)
- Correspondence: (P.M.); (E.S.M.); Tel.: +404-727-0563 (P.M.); +404-727-4273 (E.S.M.)
| |
Collapse
|
6
|
Zhang T, Lu G, Zhao Z, Liu Y, Shen Q, Li P, Chen Y, Yin H, Wang H, Marcella C, Cui B, Cheng L, Ji G, Zhang F. Washed microbiota transplantation vs. manual fecal microbiota transplantation: clinical findings, animal studies and in vitro screening. Protein Cell 2020; 11:251-266. [PMID: 31919742 PMCID: PMC7093410 DOI: 10.1007/s13238-019-00684-8] [Citation(s) in RCA: 175] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 12/03/2019] [Indexed: 02/06/2023] Open
Abstract
Fecal microbiota transplantation (FMT) by manual preparation has been applied to treat diseases for thousands of years. However, this method still endures safety risks and challenges the psychological endurance and acceptance of doctors, patients and donors. Population evidence showed the washed microbiota preparation with microfiltration based on an automatic purification system followed by repeated centrifugation plus suspension for three times significantly reduced FMT-related adverse events. This washing preparation makes delivering a precise dose of the enriched microbiota feasible, instead of using the weight of stool. Intraperitoneal injection in mice with the fecal microbiota supernatant obtained after repeated centrifugation plus suspension for three times induced less toxic reaction than that by the first centrifugation following the microfiltration. The toxic reactions that include death, the change in the level of peripheral white blood cells, and the proliferation of germinal center in secondary lymphoid follicles in spleen were noted. The metagenomic next-generation sequencing (NGS) indicated the increasing types and amount of viruses could be washed out during the washing process. Metabolomics analysis indicated metabolites with pro-inflammatory effects in the fecal microbiota supernatant such as leukotriene B4, corticosterone, and prostaglandin G2 could be removed by repeated washing. Near-infrared absorption spectroscopy could be served as a rapid detection method to control the quality of the washing-process. In conclusion, this study for the first time provides evidence linking clinical findings and animal experiments to support that washed microbiota transplantation (WMT) is safer, more precise and more quality-controllable than the crude FMT by manual.
Collapse
Affiliation(s)
- Ting Zhang
- Medical Center for Digestive Diseases, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, China
- Key Lab of Holistic Integrative Enterology, Nanjing Medical University, Nanjing, 210011, China
| | - Gaochen Lu
- Medical Center for Digestive Diseases, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, China
- Key Lab of Holistic Integrative Enterology, Nanjing Medical University, Nanjing, 210011, China
| | - Zhe Zhao
- Tianjin Key Laboratory of Optoelectronic Detection Technology and Systems, Tianjin, 300387, China
| | - Yafei Liu
- Medical Center for Digestive Diseases, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, China
- Key Lab of Holistic Integrative Enterology, Nanjing Medical University, Nanjing, 210011, China
| | - Quan Shen
- Department of Microbiology, School of Medicine, Jiangsu University, Zhenjiang, 212013, China
| | - Pan Li
- Medical Center for Digestive Diseases, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, China
- Key Lab of Holistic Integrative Enterology, Nanjing Medical University, Nanjing, 210011, China
| | - Yaoyao Chen
- Medical Center for Digestive Diseases, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, China
- Key Lab of Holistic Integrative Enterology, Nanjing Medical University, Nanjing, 210011, China
| | - Haoran Yin
- Tianjin Key Laboratory of Optoelectronic Detection Technology and Systems, Tianjin, 300387, China
| | - Huiquan Wang
- Tianjin Key Laboratory of Optoelectronic Detection Technology and Systems, Tianjin, 300387, China
| | - Cicilia Marcella
- Medical Center for Digestive Diseases, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, China
- Key Lab of Holistic Integrative Enterology, Nanjing Medical University, Nanjing, 210011, China
| | - Bota Cui
- Medical Center for Digestive Diseases, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, China
- Key Lab of Holistic Integrative Enterology, Nanjing Medical University, Nanjing, 210011, China
| | - Lei Cheng
- Biogas Institute of Ministry of Agriculture and Rural Affairs, Chengdu, 610041, China
- Center for Anaerobic Microbial Resources of Sichuan Province, Chengdu, 610041, China
| | - Guozhong Ji
- Medical Center for Digestive Diseases, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, China
- Key Lab of Holistic Integrative Enterology, Nanjing Medical University, Nanjing, 210011, China
| | - Faming Zhang
- Medical Center for Digestive Diseases, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, China.
- Key Lab of Holistic Integrative Enterology, Nanjing Medical University, Nanjing, 210011, China.
- Division of Microbiotherapy, Sir Run Run Shaw Hospital, Nanjing Medical University, Nanjing, 211166, China.
| |
Collapse
|
7
|
Liu G, Zhang F, Wang R, London SD, London L. Salivary gland immunization via Wharton's duct activates differential T-cell responses within the salivary gland immune system. FASEB J 2019; 33:6011-6022. [PMID: 30817215 PMCID: PMC6463922 DOI: 10.1096/fj.201801993r] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 01/15/2019] [Indexed: 12/16/2022]
Abstract
Salivary glands are a major component of the mucosal immune system that confer adaptive immunity to mucosal pathogens. As previously demonstrated, immunization of the submandibular gland with tissue culture-derived murine cytomegalovirus (tcMCMV) or replication-deficient adenoviruses expressing individual murine cytomegalovirus (MCMV) genes protected mice against a lethal MCMV challenge. Here, we report that salivary gland inoculation of BALB/cByJ mice with tcMCMV or recombinant adenoviruses differentially activates T helper (Th)1, -2, and -17 cells in the salivary glands vs. the associated lymph nodes. After inoculation with tcMCMV, lymphocytes from the submandibular gland preferentially express the transcription factor T-cell-specific T-box transcription factor (T-bet), which controls the expression of the hallmark Th1 cytokine, IFN-γ. Lymphocytes from the periglandular lymph nodes (PGLNs) express both T-bet and GATA-binding protein 3 (GATA3), which promotes the secretion of IL-4, -5, and -10 from Th2 cells. In contrast, after inoculation with replication-deficient adenoviruses, lymphocytes from the submandibular gland express T-bet, GATA3, and RAR-related orphan receptor γ, thymus-specific isoform (RORγt) (required for differentiation of Th17 cells) and forkhead box P3 (Foxp3) (required for the differentiation of regulatory T cells). Lymphocytes from the PGLNs were not activated. The differential induction of Th responses in the salivary gland vs. the PGLNs after inoculation with attenuated virus vs. a nominal protein antigen supports the use of the salivary as an alternative mucosal route for administering vaccines.-Liu, G., Zhang, F., Wang, R., London, S. D., London, L. Salivary gland immunization via Wharton's duct activates differential T-cell responses within the salivary gland immune system.
Collapse
Affiliation(s)
- Guangliang Liu
- Department of Oral Biology and Pathology, School of Dental Medicine, Stony Brook University, Stony Brook, New York, USA
| | - Fangfang Zhang
- Department of Oral Biology and Pathology, School of Dental Medicine, Stony Brook University, Stony Brook, New York, USA
| | - Ruixue Wang
- Department of Oral Biology and Pathology, School of Dental Medicine, Stony Brook University, Stony Brook, New York, USA
| | - Steven D. London
- Department of Oral Biology and Pathology, School of Dental Medicine, Stony Brook University, Stony Brook, New York, USA
| | - Lucille London
- Department of Oral Biology and Pathology, School of Dental Medicine, Stony Brook University, Stony Brook, New York, USA
| |
Collapse
|
8
|
Brisse E, Imbrechts M, Mitera T, Vandenhaute J, Wouters CH, Snoeck R, Andrei G, Matthys P. Lytic viral replication and immunopathology in a cytomegalovirus-induced mouse model of secondary hemophagocytic lymphohistiocytosis. Virol J 2017; 14:240. [PMID: 29258535 PMCID: PMC5738214 DOI: 10.1186/s12985-017-0908-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 12/08/2017] [Indexed: 12/28/2022] Open
Abstract
Background Hemophagocytic lymphohistiocytosis (HLH) is a rare immunological disorder caused by unbridled activation of T cells and macrophages, culminating in a life-threatening cytokine storm. A genetic and acquired subtype are distinguished, termed primary and secondary HLH, respectively. Clinical manifestations of both forms are frequently preceded by a viral infection, predominantly with herpesviruses. The exact role of the viral infection in the development of the hemophagocytic syndrome remains to be further elucidated. Methods We utilized a recently developed murine model of cytomegalovirus-associated secondary HLH and dissected the respective contributions of lytic viral replication and immunopathology in its pathogenesis. Results HLH-like disease only developed in cytomegalovirus-susceptible mouse strains unable to clear the virus, but the severity of symptoms was not correlated to the infectious viral titer. Lytic viral replication and sustained viremia played an essential part in the pathogenesis since abortive viral infection was insufficient to induce a full-blown HLH-like syndrome. Nonetheless, a limited set of symptoms, in particular anemia, thrombocytopenia and elevated levels of soluble CD25, appeared less dependent of the viral replication but rather mediated by the host’s immune response, as corroborated by immunosuppressive treatment of infected mice with dexamethasone. Conclusion Both virus-mediated pathology and immunopathology cooperate in the pathogenesis of full-blown virus-associated secondary HLH and are closely entangled. A certain level of viremia appears necessary to elicit the characteristic HLH-like symptoms in the model.
Collapse
Affiliation(s)
- Ellen Brisse
- Laboratory of Immunobiology, Rega Institute, Department of Microbiology and Immunology, KU Leuven, Leuven, Belgium
| | - Maya Imbrechts
- Laboratory of Immunobiology, Rega Institute, Department of Microbiology and Immunology, KU Leuven, Leuven, Belgium
| | - Tania Mitera
- Laboratory of Immunobiology, Rega Institute, Department of Microbiology and Immunology, KU Leuven, Leuven, Belgium
| | - Jessica Vandenhaute
- Laboratory of Immunobiology, Rega Institute, Department of Microbiology and Immunology, KU Leuven, Leuven, Belgium
| | - Carine H Wouters
- Laboratory of Pediatric Immunology, Department of Microbiology and Immunology, University Hospital Gasthuisberg, KU Leuven, Leuven, Belgium
| | - Robert Snoeck
- Laboratory of Virology and Chemotherapy, Rega Institute, Department of Microbiology and Immunology, KU Leuven, Leuven, Belgium
| | - Graciela Andrei
- Laboratory of Virology and Chemotherapy, Rega Institute, Department of Microbiology and Immunology, KU Leuven, Leuven, Belgium
| | - Patrick Matthys
- Laboratory of Immunobiology, Rega Institute, Department of Microbiology and Immunology, KU Leuven, Leuven, Belgium.
| |
Collapse
|
9
|
Ponzio TA, Sanders JW. The salivary gland as a target for enhancing immunization response. TROPICAL DISEASES TRAVEL MEDICINE AND VACCINES 2017; 3:4. [PMID: 28883974 PMCID: PMC5531011 DOI: 10.1186/s40794-017-0047-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 02/13/2017] [Indexed: 12/02/2022]
Abstract
Background An organism’s immune response to a vaccine is dependent on a number of factors, including the site of immunization. While muscle is the most common site for vaccine administration, other sites, including the salivary gland, are poised to confer stronger and broader immunoprotection. Findings Studies exploring the salivary gland as an immunization site have involved protein antigens, as well as live pathogens and DNA vaccines. While intraductal instillation of protein antigens into the salivary gland may result in a relatively transient increase in antibody production, DNA or attenuated pathogen vaccination appear to confer a lasting widespread mucosal immune response that includes robust salivary and enteric IgA, as well as high levels of circulating IgG. Furthermore, vaginal and lung antibodies are also seen. For enteric pathogens, a common class of pathogen encountered by travelers, this type of immune response provides for a level of redundant protection against foreign microbes with mucosal targets. Conclusion The strength of immune response conferred by salivary gland vaccination is generally stronger than that seen in response to the same vaccine at a comparison site. For example, where other routes fail, immunization of the salivary gland has been shown to confer protection in lethal challenge models of infectious pathogens. A host of vaccines currently under development suffer from immunogenicity challenges, adding to the widespread interest and search for novel routes and adjuvants. With its capability to facilitate a strong and broad immune response, the salivary gland warrants consideration as an immunization site, especially for vaccines with immunogenicity challenges, as well as vaccines that would benefit from combined systemic and mucosal immunity.
Collapse
Affiliation(s)
- Todd A Ponzio
- Naval Medical Research Center, 503 Robert Grant Ave., Silver Spring, MD 20910 USA.,Wake Forest University School of Medicine, Winston-Salem, USA
| | - John W Sanders
- Wake Forest University School of Medicine, Medical Center Blvd., Winston-Salem, NC 27157 USA.,Hefner Veterans Affairs Medical Center, Salisbury, NC UK
| |
Collapse
|
10
|
Alston CI, Dix RD. Murine cytomegalovirus infection of mouse macrophages stimulates early expression of suppressor of cytokine signaling (SOCS)1 and SOCS3. PLoS One 2017; 12:e0171812. [PMID: 28182772 PMCID: PMC5300177 DOI: 10.1371/journal.pone.0171812] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 01/26/2017] [Indexed: 12/13/2022] Open
Abstract
Human cytomegalovirus (HCMV) is a species-specific β-herpesvirus that infects for life up to 80% of the world’s population and causes severe morbidity in at-risk immunocompromised populations. Suppressors of cytokine signaling (SOCS)1 and SOCS3 are host proteins that act as inducible negative feedback regulators of cytokine signaling and have been implicated in several ocular diseases and viral infections. We recently found in our mouse model of experimental cytomegalovirus retinitis that subretinally-injected murine cytomegalovirus (MCMV) stimulates ocular SOCS1 and SOCS3 during retrovirus-induced immune suppression of murine AIDS (MAIDS), and that infiltrating macrophages are prominent cellular sources of retinal SOCS1 and SOCS3 expression. Herein we investigate possible virologic mechanisms whereby MCMV infection may stimulate SOCS1 and/or SOCS3 expression in cell culture. We report that infection of IC-21 mouse macrophages with MCMV propagated through the salivary glands of BALB/c mice, but not from tissue culture in C57BL/6 fibroblasts, transiently stimulates SOCS1 and SOCS3 mRNA transcripts, but not SOCS5 mRNA. Viral tegument proteins are insufficient for this stimulation, as replication-deficient UV-inactivated MCMV fails to stimulate SOCS1 or SOCS3 in IC-21 macrophages. By contrast, infection of murine embryonic fibroblasts (MEFs) with either productive MCMV or UV-inactivated MCMV significantly stimulates SOCS1 and SOCS3 mRNA expression early after infection. Treatment of MCMV-infected IC-21 mouse macrophages with the antiviral drug ganciclovir significantly decreases MCMV-stimulated SOCS3 expression at 3 days post-infection. These data suggest cell type-specific, different roles for viral immediate early or early gene expression and/or viral tegument proteins in the early stimulation of SOCS1 and SOCS3 during MCMV infection. Furthermore, putative biphasic stimulation of SOCS3 during late MCMV infection of IC-21 mouse macrophages may occur by divergent virologic mechanisms.
Collapse
Affiliation(s)
- Christine I. Alston
- Viral Immunology Center, Department of Biology, Georgia State University, Atlanta, Georgia, United States of America
- Department of Ophthalmology, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Richard D. Dix
- Viral Immunology Center, Department of Biology, Georgia State University, Atlanta, Georgia, United States of America
- Department of Ophthalmology, Emory University School of Medicine, Atlanta, Georgia, United States of America
- * E-mail:
| |
Collapse
|
11
|
The Salivary Gland Acts as a Sink for Tissue-Resident Memory CD8(+) T Cells, Facilitating Protection from Local Cytomegalovirus Infection. Cell Rep 2015; 13:1125-1136. [PMID: 26526997 DOI: 10.1016/j.celrep.2015.09.082] [Citation(s) in RCA: 115] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Revised: 09/14/2015] [Accepted: 09/25/2015] [Indexed: 11/20/2022] Open
Abstract
Tissue-resident memory T cells (TRM) reside in barrier tissues and provide local immediate protective immunity. Here, we show that the salivary gland (SG) most-effectively induces CD8(+) and CD4(+) TRM cells against murine cytomegalovirus (MCMV), which persists in and spreads from this organ. TRM generation depended on local antigen for CD4(+), but not CD8(+), TRM cells, highlighting major differences in T cell subset-specific demands for TRM development. CMV-specific CD8(+) T cells fail to control virus replication upon primary infection in the SG due to CMV-induced MHC I downregulation in glandular epithelial cells. Using intraglandular infection, we challenge this notion and demonstrate that memory CD8(+) T cells confer immediate protection against locally introduced MCMV despite active viral immune evasion, owing to early viral tropism to cells that largely withstand MHC I downregulation. Thus, we unravel a yet-unappreciated role for memory CD8(+) T cells in protecting mucosal tissues against CMV infection.
Collapse
|
12
|
Liu G, Zhang F, Wang R, London L, London SD. Protective MCMV immunity by vaccination of the salivary gland via Wharton's duct: replication-deficient recombinant adenovirus expressing individual MCMV genes elicits protection similar to that of MCMV. FASEB J 2014; 28:1698-710. [PMID: 24391133 DOI: 10.1096/fj.13-244178] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Salivary glands, a major component of the mucosal immune system, confer antigen-specific immunity to mucosally acquired pathogens. We investigated whether a physiological route of inoculation and a subunit vaccine approach elicited MCMV-specific and protective immunity. Mice were inoculated by retrograde perfusion of the submandibular salivary glands via Wharton's duct with tcMCMV or MCMV proteins focused to the salivary gland via replication-deficient adenovirus expressing individual MCMV genes (gB, gH, IE1; controls: saline and replication deficient adenovirus without MCMV inserts). Mice were evaluated for MCMV-specific antibodies, T-cell responses, germinal center formation, and protection against a lethal MCMV challenge. Retrograde perfusion with tcMCMV or adenovirus expressed MCMV proteins induced a 2- to 6-fold increase in systemic and mucosal MCMV-specific antibodies, a 3- to 6-fold increase in GC marker expression, and protection against a lethal systemic challenge, as evidenced by up to 80% increased survival, decreased splenic pathology, and decreased viral titers from 10(6) pfu to undetectable levels. Thus, a focused salivary gland immunization via a physiological route with a protein antigen induced systemic and mucosal protective immune responses. Therefore, salivary gland immunization can serve as an alternative mucosal route for administering vaccines, which is directly applicable for use in humans.
Collapse
Affiliation(s)
- Guangliang Liu
- 1Stony Brook University, School of Dental Medicine, Department of Oral Biology and Pathology, Stony Brook, NY 11794, USA.
| | | | | | | | | |
Collapse
|
13
|
Liu LL, Li XF, Qin WQ, Liu XL, Li G, Shu SN, Fang F. Is IL-17 an accomplice contributing to salivary gland damage during CMV infection? Future Virol 2013. [DOI: 10.2217/fvl.13.78] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Aim: This study aimed to explore the potential role of IL-17 in the pathological damage to the salivary gland during CMV infection. Materials & methods: For all studies, 4-week-old female BALB/c mice were used. Mouse embryo fibroblast cells were isolated from embryos of pregnant mice. The Smith murine CMV (MCMV) strain was propagated and obtained from homogenates of the salivary glands. BALB/c mice were randomly divided into two groups. Mice in one group were infected with Smith MCMV to establish disseminated infection; mice in the other group were controls. Four mice of each group were randomly chosen to be harvested on days 3, 7, 14 and 28. Viral titers in salivary gland tissues were determined using a standard plaque assay; IL-17+ T cells in the spleen were analyzed by flow cytometry; the expressions of IL-17 and IL-17 receptor (IL-17R) mRNA was measured by reverse transcription PCR; and the pathology of the salivary glands in infected mice were assessed by hematoxylin and eosin staining. Results: The viral titers in salivary gland tissues, which were much higher than those of other organs, elevated on day 7 and peaked on day 14, then gradually declined on day 28. The percentages of CD4+IL-17+ T cells were increased in MCMV-infected mice compared with controls, with the main increase of CD4+IL-17+ T cells being Th17 cells (p < 0.05). In salivary gland tissues, the expression of IL-17 and IL-17R mRNA increased significantly and achieved the highest level on day 14, which was significantly higher than that of the controls (p < 0.05). Compared with controls, serious inflammatory cellular infiltrates in the salivary glands were present on day 7 after MCMV infection, and on day 14, many of the cellular infiltrate became larger and coalesced with neighboring foci. We found that IL-17 and IL-17R were enriched in the salivary glands when the tissues were mainly injured. Conclusion: The results illustrated that the inflammatory factors, IL-17 and IL-17R, possibly contributed to the process of viral infection and played a part in inducing pathological damage of the salivary gland.
Collapse
Affiliation(s)
- Ling-ling Liu
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430030, China
| | - Xu-fang Li
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430030, China
| | - Wen-qing Qin
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430030, China
| | - Xing-lou Liu
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430030, China
| | - Ge Li
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430030, China
| | - Sai-nan Shu
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430030, China
| | - Feng Fang
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430030, China
| |
Collapse
|
14
|
Melnick M, Abichaker G, Htet K, Sedghizadeh P, Jaskoll T. Small molecule inhibitors of the host cell COX/AREG/EGFR/ERK pathway attenuate cytomegalovirus-induced pathogenesis. Exp Mol Pathol 2011; 91:400-10. [PMID: 21565184 DOI: 10.1016/j.yexmp.2011.04.014] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2011] [Accepted: 04/18/2011] [Indexed: 11/17/2022]
Abstract
As with other herpesviruses, human cytomegalovirus (hCMV) has the ability to establish lifelong persistence and latent infection following primary exposure, salivary glands (SMGs) being the primary site of both. In the immunocompromised patient, hCMV is a common cause of opportunistic infections, and subsequent morbidity and mortality. Elucidating the molecular pathogenesis of CMV-induced disease is critical to the development of more effective and safer drug therapies. In the present study, we used a novel mouse postnatal SMG organ culture model of mCMV-induced dysplasia to investigate a candidate signaling network suggested by our prior studies (COX-2/AREG/EGFR/ERK). The objective was to employ small molecule inhibitors to target several key steps in the autocrine loop, and in this way ameliorate pathology. Our results indicate that upregulation of ERK phosphorylation is necessary for initial mCMV-induced pathogenesis, and that ErbB receptor family phosphorylation and downstream signaling are highly relevant targets for drug discovery.
Collapse
Affiliation(s)
- Michael Melnick
- Laboratory for Developmental Genetics, USC, Los Angeles, CA 90089-0641, USA.
| | | | | | | | | |
Collapse
|
15
|
Grewal JS, Pilgrim MJ, Grewal S, Kasman L, Werner P, Bruorton ME, London SD, London L. Salivary glands act as mucosal inductive sites via the formation of ectopic germinal centers after site-restricted MCMV infection. FASEB J 2011; 25:1680-96. [PMID: 21307334 DOI: 10.1096/fj.10-174656] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
We investigated the hypothesis that salivary gland inoculation stimulates formation of ectopic germinal centers (GCs), transforming the gland into a mucosal inductive site. Intraglandular infection of mice with murine cytomegalovirus (MCMV; control: UV-inactivated MCMV) induces salivary gland ectopic follicles comprising cognate interactions between CD4(+) and B220(+) lymphocytes, IgM(+) and isotype-switched IgG(+) and IgA(+) B cells, antigen presenting cells, and follicular dendritic cells. B cells coexpressed the GC markers GCT (57%) and GL7 (52%), and bound the lectin peanut agglutinin. Lymphoid follicles were characterized by a 2- to 3-fold increase in mRNA for CXCL13 (lymphoid neogenesis), syndecan-1 (plasma cells), Blimp-1 (plasma cell development/differentiation), and a 2- to 6-fold increase for activation-induced cytidine deaminase, PAX5, and the nonexcised rearranged DNA of an IgA class-switch event, supporting somatic hypermutation and class-switch recombination within the salivary follicles. Intraglandular inoculation also provided protection against a systemic MCMV challenge, as evidenced by decreased viral titers (10(5) plaque-forming units to undetectable), and restoration of normal salivary flow rates from a 6-fold decrease. Therefore, these features suggest that the salivary gland participates in oral mucosal immunity via generation of ectopic GCs, which function as ectopic mucosal inductive sites.
Collapse
Affiliation(s)
- Jasvir S Grewal
- Department of Oral Biology and Pathology, School of Dental Medicine, Stony Brook University, Stony Brook, New York 11794, USA
| | | | | | | | | | | | | | | |
Collapse
|
16
|
A spread-deficient cytomegalovirus for assessment of first-target cells in vaccination. J Virol 2010; 84:7730-42. [PMID: 20463067 DOI: 10.1128/jvi.02696-09] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Human cytomegalovirus (HCMV) is a human pathogen that causes severe disease primarily in the immunocompromised or immunologically immature individual. To date, no vaccine is available. We describe use of a spread-deficient murine CMV (MCMV) as a novel approach for betaherpesvirus vaccination. To generate a spread-deficient MCMV, the conserved, essential gene M94 was deleted. Immunization with MCMV-DeltaM94 is apathogenic and protective against wild-type challenge even in highly susceptible IFNalphabetaR(-/-) mice. MCMV-DeltaM94 was able to induce a robust CD4(+) and CD8(+) T-cell response as well as a neutralizing antibody response comparable to that induced by wild-type infection. Endothelial cells were identified as activators of CD8(+) T cells in vivo. Thus, the vaccination with a spread-deficient betaherpesvirus is a safe and protective strategy and allows the linkage between cell tropism and immunogenicity. Furthermore, genomes of MCMV-DeltaM94 were present in lungs 12 months after infection, revealing first-target cells as sites of genome maintenance.
Collapse
|
17
|
Kasman LM, London LL, London SD, Pilgrim MJ. A mouse model linking viral hepatitis and salivary gland dysfunction. Oral Dis 2009; 15:587-95. [PMID: 19656314 DOI: 10.1111/j.1601-0825.2009.01600.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
OBJECTIVE Viral hepatitis is known to cause xerostomia in humans, but this has not been reported in an animal model. We report a severe, acute, highly reproducible saliva deficiency occurring in BALB/c mice as a result of experimental viral hepatitis. MATERIALS AND METHODS BALB/c mice, splenectomized or carrying genetic mutations to detect immunological contributions to the saliva deficiency syndrome, were infected intraperitoneally with a non-lethal dose of murine cytomegalovirus. Pilocarpine-stimulated saliva volumes were determined between 0 and 15 days after infection. Salivary gland, liver, spleen, and sera were analyzed for the presence of virus, cytokines, inflammatory infiltrates, and tissue damage. RESULTS Saliva deficiency was detectable 2 days after cytomegalovirus infection, peaked at 88% below normal by day 7, and resolved partially in all mice by 15 days postinfection as sialoadenitis increased. Neither salivary gland viral titers, sialoadenitis, splenectomy, nor systemic inflammatory markers correlated with hyposalivation severity. Elevated liver enzymes did correlate with hyposalivation, and mice genetically resistant to murine cytomegalovirus-induced hepatitis were significantly protected. CONCLUSIONS Murine cytomegalovirus-induced salivary gland dysfunction is biphasic, with an acute hepatitis-associated phase and a later sialoadenitis-associated phase. Acute murine cytomegalovirus infection of BALB/c mice may provide a model for investigation of hepatitis-associated xerostomia.
Collapse
Affiliation(s)
- L M Kasman
- Department of Microbiology and Immunology, Medical University of South Carolina, BSB-201, PO Box 250504, 173 Ashley Ave, Charleston, SC 29403, USA.
| | | | | | | |
Collapse
|