1
|
Drysdale SB, Thwaites RS, Price J, Thakur D, McGinley J, McPherson C, Öner D, Aerssens J, Openshaw PJ, Pollard AJ. What have we learned from animal studies of immune responses to respiratory syncytial virus infection? J Clin Virol 2024; 175:105731. [PMID: 39368446 DOI: 10.1016/j.jcv.2024.105731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 09/18/2024] [Indexed: 10/07/2024]
Abstract
Respiratory syncytial virus (RSV) is a common cause of severe respiratory tract infection at the extremes of age and in vulnerable populations. However, it is difficult to predict the clinical course and most infants who develop severe disease have no pre-existing risk factors. With the recent licencing of RSV vaccines and monoclonal antibodies, it is important to identify high-risk individuals in order to prioritise those who will most benefit from prophylaxis. The immune response to RSV and the mechanisms by which the virus prevents the establishment of immunological memory have been extensively investigated but remain incompletely characterised. In animal models, beneficial and harmful immune responses have both been demonstrated. While only chimpanzees are fully permissive for human RSV replication, most research has been conducted in rodents, or in calves infected with bovine RSV. Based on these studies, components of innate and adaptive immune systems, cytokines, chemokines and metabolites, and specific genetic and transcriptomic signatures are identified as potential predictive indicators of RSV disease severity. These findings may inform the development of future human studies and contribute to the early identification of patients at high risk of severe infection. This narrative review summarises the factors involved in the immune response to RSV infection in these models and highlights the relationship between potential biomarkers and disease severity.
Collapse
Affiliation(s)
- Simon B Drysdale
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, United Kingdom and the NIHR Oxford Biomedical Research Centre, Oxford, UK.
| | - Ryan S Thwaites
- National Heart and Lung Institute, Imperial College London, London, UK
| | | | - Devika Thakur
- St George's University Hospitals NHS Foundation Trust, London, UK
| | - Joseph McGinley
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, United Kingdom and the NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Calum McPherson
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, United Kingdom and the NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Deniz Öner
- Infectious Diseases Translational Biomarkers, Janssen Pharmaceutica NV, Beerse, Belgium
| | - Jeroen Aerssens
- Infectious Diseases Translational Biomarkers, Janssen Pharmaceutica NV, Beerse, Belgium
| | - Peter Jm Openshaw
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Andrew J Pollard
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, United Kingdom and the NIHR Oxford Biomedical Research Centre, Oxford, UK
| |
Collapse
|
2
|
Xu H, Zhou R, Chen Z. Tissue-Resident Memory T Cell: Ontogenetic Cellular Mechanism and Clinical Translation. Clin Exp Immunol 2023; 214:249-259. [PMID: 37586053 PMCID: PMC10719502 DOI: 10.1093/cei/uxad090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 06/22/2023] [Accepted: 08/15/2023] [Indexed: 08/18/2023] Open
Abstract
Mounting evidence has indicated the essential role of tissue-resident memory T (TRM) cells for frontline protection against viral infection and for cancer immune surveillance (Mueller SN, Mackay LK. Tissue-resident memory T cells: local specialists in immune defense. Nat Rev Immunol 2016, 16, 79-89. doi:10.1038/nri.2015.3.). TRM cells are transcriptionally, phenotypically, and functionally distinct from circulating memory T (Tcirm) cells. It is necessary to understand the unique ontogenetic mechanism, migratory regulation, and biological function of TRM cells. In this review, we discuss recent insights into cellular mechanisms and discrete responsiveness in different tissue microenvironments underlying TRM cell development. We also emphasize the translational potential of TRM cells by focusing on their establishment in association with improved protection in mucosal tissues against various types of diseases and effective strategies for eliciting TRM cells in both pre-clinical and clinical studies.
Collapse
Affiliation(s)
- Haoran Xu
- AIDS Institute, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong; Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong; Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
| | - Runhong Zhou
- AIDS Institute, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong; Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong; Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
| | - Zhiwei Chen
- AIDS Institute, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong; Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong; Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
- State Key Laboratory for Emerging Infectious Diseases, University of Hong Kong; Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
| |
Collapse
|
3
|
De C, Pickles RJ, Yao W, Liao B, Boone A, Choi M, Battaglia DM, Askin FB, Whitmire JK, Silvestri G, Garcia JV, Wahl A. Human T cells efficiently control RSV infection. JCI Insight 2023; 8:e168110. [PMID: 37159271 PMCID: PMC10393221 DOI: 10.1172/jci.insight.168110] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 04/26/2023] [Indexed: 05/10/2023] Open
Abstract
Respiratory syncytial virus (RSV) infection causes significant morbidity and mortality in infants, immunocompromised individuals, and older individuals. There is an urgent need for effective antivirals and vaccines for high-risk individuals. We used 2 complementary in vivo models to analyze RSV-associated human lung pathology and human immune correlates of protection. RSV infection resulted in widespread human lung epithelial damage, a proinflammatory innate immune response, and elicited a natural adaptive human immune response that conferred protective immunity. We demonstrated a key role for human T cells in controlling RSV infection. Specifically, primed human CD8+ T cells or CD4+ T cells effectively and independently control RSV replication in human lung tissue in the absence of an RSV-specific antibody response. These preclinical data support the development of RSV vaccines, which also elicit effective T cell responses to improve RSV vaccine efficacy.
Collapse
Affiliation(s)
- Chandrav De
- International Center for the Advancement of Translational Science
- Division of Infectious Diseases, Department of Medicine
- Center for AIDS Research
| | - Raymond J. Pickles
- Department of Microbiology and Immunology, and
- Marsico Lung Institute, University of North Carolina (UNC) at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Wenbo Yao
- International Center for the Advancement of Translational Science
- Division of Infectious Diseases, Department of Medicine
- Center for AIDS Research
| | - Baolin Liao
- International Center for the Advancement of Translational Science
- Division of Infectious Diseases, Department of Medicine
- Center for AIDS Research
- Department of Infectious Diseases, Guangzhou Eighth People’s Hospital, Guangzhou Medical University, Guangzhou, China
| | - Allison Boone
- Department of Microbiology and Immunology, and
- Marsico Lung Institute, University of North Carolina (UNC) at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Mingyu Choi
- International Center for the Advancement of Translational Science
- Division of Infectious Diseases, Department of Medicine
- Center for AIDS Research
| | - Diana M. Battaglia
- International Center for the Advancement of Translational Science
- Division of Infectious Diseases, Department of Medicine
- Center for AIDS Research
| | | | - Jason K. Whitmire
- Department of Microbiology and Immunology, and
- Department of Genetics, and
- Lineberger Comprehensive Cancer Center, UNC at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Guido Silvestri
- Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - J. Victor Garcia
- International Center for the Advancement of Translational Science
- Division of Infectious Diseases, Department of Medicine
- Center for AIDS Research
| | - Angela Wahl
- International Center for the Advancement of Translational Science
- Division of Infectious Diseases, Department of Medicine
- Center for AIDS Research
| |
Collapse
|
4
|
Iordache A, Balica NC, Horhat ID, Morar R, Tischer AA, Milcu AI, Salavat MC, Borugă VM. A Review Regarding the Connections between Allergic Rhinitis and Asthma - Epidemiology, Diagnosis and Treatment. CURRENT HEALTH SCIENCES JOURNAL 2023; 49:5-18. [PMID: 37780190 PMCID: PMC10541080 DOI: 10.12865/chsj.49.01.5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Accepted: 02/15/2023] [Indexed: 10/03/2023]
Abstract
Allergic rhinitis is characterized by an acute or chronic inflammation of the nasal mucosa, being frequently associated with other airway conditions such as sinusitis, serous otitis media, nasal polyposis, sleep disorders and asthma in particular. Among the comorbidities of allergic rhinitis it counts asthma, being a risk factor for this disorder, in which, more than 75% of patients develop asthma (either allergic or nonallergic), whereas the patients with allergic rhinitis can be affected up to 40% by asthma. The classic symptoms for allergic rhinitis involves sneezing, nasal mucosal swelling and watery rhinorrhea; whereas the main symptoms which occurred in patients with asthma are wheezing, breathlessness, chest tightness, coughing, fast heartbeat, confusion, exhaustion or dizziness. Avoiding allergens is the first line of treatment for allergic rhinitis, followed by medication and allergen immunotherapy. Due to the strong connection between allergic rhinitis and asthma, one can affirm that the treatment for allergic rhinitis lead to the improvement of asthma symptoms. One can diagnose asthma by recognizing a certain pattern of respiratory symptoms and expiratory airflow restriction, which varies for each patient. In conclusion, accurate identification of the differences between allergic rhinitis and asthma depends on a thorough history, physical examination, and therapeutic treatments. This article provides an overview of the connection between these disorders, as well as of the diagnosis of these conditions and their current management options.
Collapse
Affiliation(s)
- Andreea Iordache
- PhD Student, Department of ENT, Victor Babeş University of Medicine and Pharmacy, Timişoara, Romania
| | - Nicolae Constantin Balica
- Department of ENT, Victor Babeş University of Medicine and Pharmacy, Timişoara, Romania
- Department of ENT, Emergency City Hospital, Timişoara, Romania
| | - Ioana Delia Horhat
- Department of ENT, Victor Babeş University of Medicine and Pharmacy, Timişoara, Romania
- Department of ENT, Emergency City Hospital, Timişoara, Romania
| | - Raluca Morar
- PhD Student, Department of ENT, Victor Babeş University of Medicine and Pharmacy, Timişoara, Romania
| | | | - Adina Iuliana Milcu
- Department of Surgery, Victor Babeş University of Medicine and Pharmacy, Timişoara, Romania
| | | | - Veronica Mădălina Borugă
- Department of Microbiology, Victor Babeş University of Medicine and Pharmacy, Timişoara, Romania
| |
Collapse
|
5
|
Wellemans V, Benhassou HA, Fuselier E, Bellesort F, Dury S, Lebargy F, Dormoy V, Fichel C, Naour RL, Gounni AS, Lamkhioued B. Role of CCR3 in respiratory syncytial virus infection of airway epithelial cells. iScience 2021; 24:103433. [PMID: 34917892 PMCID: PMC8646169 DOI: 10.1016/j.isci.2021.103433] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 08/29/2021] [Accepted: 11/10/2021] [Indexed: 01/17/2023] Open
Abstract
Respiratory syncytial virus (RSV) infection is the principal cause of severe lower respiratory tract disease and accounts for a significant risk for developing asthma later in life. Clinical studies have shown an increase in airway responsiveness and a concomitant Th2 response in the lungs of RSV-infected patients. These indications suggest that RSV may modulate aspects of the immune response to promote virus replication. Here, we show that CCR3 facilitates RSV infection of airway epithelial cells, an effect that was inhibited by eotaxin-1/CCL11 or upon CCR3 gene silencing. Mechanistically, cellular entry of RSV is mediated by binding of the viral G protein to CCR3 and selective chemotaxis of Th2 cells and eosinophils. In vivo, mice lacking CCR3 display a significant reduction in RSV infection, airway inflammation, and mucus production. Overall, RSV G protein-CCR3 interaction may participate in pulmonary infection and inflammation by enhancing eosinophils' recruitment and less potent antiviral Th2 cells. CCR3 mediates RSV infection of human airway epithelial cells Eotaxin-1 blocks RSV-G binding to CCR3 and significantly decreases RSV infection RSV-G secreted protein (sG) attracts human eosinophils and Th2 cells through CCR3 RSV infection of mice lacking CCR3 exhibited reduced inflammation and mucus secretion
Collapse
Affiliation(s)
| | - Hassan Ait Benhassou
- Laboratoire d'Immunologie et de Biotechnologie, EA7509-IRMAIC, Pôle-Santé, Université de Reims Champagne-Ardenne, Reims, France
| | - Eloise Fuselier
- Laboratoire d'Immunologie et de Biotechnologie, EA7509-IRMAIC, Pôle-Santé, Université de Reims Champagne-Ardenne, Reims, France
| | | | - Sandra Dury
- Laboratoire d'Immunologie et de Biotechnologie, EA7509-IRMAIC, Pôle-Santé, Université de Reims Champagne-Ardenne, Reims, France.,Service des Maladies Respiratoires et Allergiques. Hôpital Maison Blanche, CHU de Reims, Reims, France
| | - François Lebargy
- Laboratoire d'Immunologie et de Biotechnologie, EA7509-IRMAIC, Pôle-Santé, Université de Reims Champagne-Ardenne, Reims, France.,Service des Maladies Respiratoires et Allergiques. Hôpital Maison Blanche, CHU de Reims, Reims, France
| | - Valérian Dormoy
- Inserm UMR-S 1250, Pathologies Pulmonaires et Plasticité Cellulaire (P3Cell). Université de Reims Champagne-Ardenne, Reims, France
| | - Caroline Fichel
- Laboratoire d'Immunologie et de Biotechnologie, EA7509-IRMAIC, Pôle-Santé, Université de Reims Champagne-Ardenne, Reims, France
| | - Richard Le Naour
- Laboratoire d'Immunologie et de Biotechnologie, EA7509-IRMAIC, Pôle-Santé, Université de Reims Champagne-Ardenne, Reims, France
| | - Abdelilah S Gounni
- Department of Immunology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Canada
| | - Bouchaib Lamkhioued
- Laboratoire d'Immunologie et de Biotechnologie, EA7509-IRMAIC, Pôle-Santé, Université de Reims Champagne-Ardenne, Reims, France
| |
Collapse
|
6
|
McGinley J, Thwaites R, Brebner W, Greenan-Barrett L, Aerssens J, Öner D, Bont L, Wildenbeest J, Martinón-Torres F, Nair H, Pollard AJ, Openshaw P, Drysdale S. A Systematic Review and Meta-analysis of Animal Studies Investigating the Relationship Between Serum Antibody, T Lymphocytes, and Respiratory Syncytial Virus Disease. J Infect Dis 2021; 226:S117-S129. [PMID: 34522970 DOI: 10.1093/infdis/jiab370] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 07/15/2021] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Respiratory syncytial virus (RSV) infections occur in human populations around the globe, causing disease of variable severity, disproportionately affecting infants and older adults (>65 years of age). Immune responses can be protective but also contribute to disease. Experimental studies in animals enable detailed investigation of immune responses, provide insights into clinical questions, and accelerate the development of passive and active vaccination. We aimed to review the role of antibody and T-cell responses in relation to RSV disease severity in animals. METHODS Systematic review and meta-analysis of animal studies examining the association between T-cell responses/phenotype or antibody titers and severity of RSV disease. The PubMed, Zoological Record, and Embase databases were screened from January 1980 to May 2018 to identify animal studies of RSV infection that assessed serum antibody titer or T lymphocytes with disease severity as an outcome. Sixty-three studies were included in the final review. RESULTS RSV-specific antibody appears to protect from disease in mice, but such an effect was less evident in bovine RSV. Strong T-cell, Th1, Th2, Th17, CD4/CD8 responses, and weak Treg responses accompany severe disease in mice. CONCLUSIONS Murine studies suggest that measures of T-lymphocyte activity (particularly CD4 and CD8 T cells) may be predictive biomarkers of severity. Further inquiry is merited to validate these results and assess relevance as biomarkers for human disease.
Collapse
Affiliation(s)
- Joseph McGinley
- Oxford Vaccine Group, Paediatrics, University of Oxford, Oxford, United Kingdom
| | | | - Will Brebner
- Oxford Vaccine Group, Paediatrics, University of Oxford, Oxford, United Kingdom
| | | | - Jeroen Aerssens
- Biomarkers Infectious Diseases, Janssen Pharmaceutica NV, Beerse, Belgium
| | - Deniz Öner
- Biomarkers Infectious Diseases, Janssen Pharmaceutica NV, Beerse, Belgium
| | - Louis Bont
- Department of Paediatric Infectious Diseases and Immunology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Joanne Wildenbeest
- Department of Paediatric Infectious Diseases and Immunology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | | | - Harish Nair
- University of Edinburgh, Edinburgh, United Kingdom
| | - Andrew J Pollard
- Oxford Vaccine Group, Paediatrics, University of Oxford, Oxford, United Kingdom
| | | | - Simon Drysdale
- Oxford Vaccine Group, Paediatrics, University of Oxford, Oxford, United Kingdom.,Paediatric Infectious Diseases Unit, St George's University Hospitals NHS Foundation Trust, London, United Kingdom.,Paediatric Infectious Diseases Research Group, Institute for Infection and Immunity, St George's, University of London, London, United Kingdom
| | | |
Collapse
|
7
|
Eichinger KM, Kosanovich JL, Lipp M, Empey KM, Petrovsky N. Strategies for active and passive pediatric RSV immunization. Ther Adv Vaccines Immunother 2021; 9:2515135520981516. [PMID: 33623860 PMCID: PMC7879001 DOI: 10.1177/2515135520981516] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Accepted: 11/20/2020] [Indexed: 12/26/2022] Open
Abstract
Respiratory syncytial virus (RSV) is the leading cause of lower respiratory tract infections in children worldwide, with the most severe disease occurring in very young infants. Despite half a century of research there still are no licensed RSV vaccines. Difficulties in RSV vaccine development stem from a number of factors, including: (a) a very short time frame between birth and first RSV exposure; (b) interfering effects of maternal antibodies; and (c) differentially regulated immune responses in infants causing a marked T helper 2 (Th2) immune bias. This review seeks to provide an age-specific understanding of RSV immunity critical to the development of a successful pediatric RSV vaccine. Historical and future approaches to the prevention of infant RSV are reviewed, including passive protection using monoclonal antibodies or maternal immunization strategies versus active infant immunization using pre-fusion forms of RSV F protein antigens formulated with novel adjuvants such as Advax that avoid excess Th2 immune polarization.
Collapse
Affiliation(s)
- Katherine M. Eichinger
- Department of Pharmacy and Therapeutics, University of Pittsburgh School of Pharmacy, and Clinical and Translational Science Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jessica L. Kosanovich
- Department of Pharmacy and Therapeutics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Madeline Lipp
- Department of Pharmacy and Therapeutics, University of Pittsburgh School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kerry M. Empey
- Department of Pharmacy and Therapeutics, Department of Pharmaceutical Sciences, School of Medicine and Clinical and Translational Science Institute, University of Pittsburgh School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA
| | - Nikolai Petrovsky
- College of Medicine and Public Health, Flinders University, Bedford Park, South Australia 5042, Australia and Vaxine Pty Ltd, Warradale, SA 5046, Australia
| |
Collapse
|
8
|
Eichinger KM, Kosanovich JL, Gidwani SV, Zomback A, Lipp MA, Perkins TN, Oury TD, Petrovsky N, Marshall CP, Yondola MA, Empey KM. Prefusion RSV F Immunization Elicits Th2-Mediated Lung Pathology in Mice When Formulated With a Th2 (but Not a Th1/Th2-Balanced) Adjuvant Despite Complete Viral Protection. Front Immunol 2020; 11:1673. [PMID: 32849580 PMCID: PMC7403488 DOI: 10.3389/fimmu.2020.01673] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 06/23/2020] [Indexed: 12/19/2022] Open
Abstract
Respiratory syncytial virus (RSV) remains the most common cause of lower respiratory tract infections in children worldwide. Development of a vaccine has been hindered by the risk of developing enhanced respiratory disease (ERD) upon natural exposure to the virus. Generation of higher quality neutralizing antibodies with stabilized pre-fusion F protein antigens has been proposed as a strategy to prevent ERD. We sought to test whether there was evidence of ERD in naïve BALB/c mice immunized with an unadjuvanted, stabilized pre-fusion F protein, and challenged with RSV line 19. We further sought to determine the extent to which formulation with a Th2-biased (alum) or a more Th1/Th2-balanced (Advax-SM) adjuvant influenced cellular responses and lung pathology. When exposed to RSV, mice immunized with pre-fusion F protein alone (PreF) exhibited increased airway eosinophilia and mucus accumulation. This was further exacerbated by formulation of PreF with Alum (aluminum hydroxide). Conversely, formulation of PreF with a Th1/Th2-balanced adjuvant, Advax-SM, not only suppressed RSV viral replication, but also inhibited airway eosinophilia and mucus accumulation. This was associated with lower numbers of lung innate lymphocyte cells (ILC2s) and CD4+ T cells producing IL-5+ or IL-13+ and increased IFNγ+ CD4+ and CD8+ T cells, in addition to RSV F-specific CD8+ T cells. These data suggest that in the absence of preimmunity, stabilized PreF antigens may still be associated with aberrant Th2 responses that induce lung pathology in response to RSV infection, and can be prevented by formulation with more Th1/Th2-balanced adjuvants that enhance CD4+ and CD8+ IFNγ+ T cell responses. This may support the use of stabilized PreF antigens with Th1/Th2-balanced adjuvants like, Advax-SM, as safer alternatives to alum in RSV vaccine candidates.
Collapse
Affiliation(s)
- Katherine M Eichinger
- Department of Pharmacy and Therapeutics, University of Pittsburgh School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, United States.,Center for Clinical Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, United States.,Department of Medicine, Division of Internal Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Jessica L Kosanovich
- Department of Pharmacy and Therapeutics, University of Pittsburgh School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, United States.,Center for Clinical Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, United States
| | | | - Aaron Zomback
- Calder Biosciences, New York City, NY, United States
| | - Madeline A Lipp
- Department of Pharmacy and Therapeutics, University of Pittsburgh School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, United States
| | - Timothy N Perkins
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Tim D Oury
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Nikolai Petrovsky
- Vaxine Pty Ltd., Bedford Park, SA, Australia.,College of Medicine and Public Health, Flinders University, Bedford Park, SA, Australia
| | | | | | - Kerry M Empey
- Department of Pharmacy and Therapeutics, University of Pittsburgh School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, United States.,Center for Clinical Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, United States.,Department of Immunology, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
9
|
Schmidt ME, Meyerholz DK, Varga SM. Pre-existing neutralizing antibodies prevent CD8 T cell-mediated immunopathology following respiratory syncytial virus infection. Mucosal Immunol 2020; 13:507-517. [PMID: 31844172 PMCID: PMC7181396 DOI: 10.1038/s41385-019-0243-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Revised: 11/05/2019] [Accepted: 11/23/2019] [Indexed: 02/04/2023]
Abstract
Despite being a leading cause of severe respiratory disease, there remains no licensed respiratory syncytial virus (RSV) vaccine. Neutralizing antibodies reduce the severity of RSV-associated disease, but are not sufficient for preventing reinfection. In contrast, the role of memory CD8 T cells in protecting against a secondary RSV infection is less established. We recently demonstrated that high-magnitude memory CD8 T cells efficiently reduced lung viral titers following RSV infection, but induced fatal immunopathology that was mediated by IFN-γ. To evaluate the ability of RSV-specific neutralizing antibodies to prevent memory CD8 T cell-mediated immunopathology, mice with high-magnitude memory CD8 T cell responses were treated with neutralizing antibodies prior to RSV challenge. Neutralizing antibody treatment significantly reduced morbidity and prevented mortality following RSV challenge compared with IgG-treated controls. Neutralizing antibody treatment restricted early virus replication, which caused a substantial reduction in memory CD8 T cell activation and IFN-γ production, directly resulting in survival. In contrast, therapeutic neutralizing antibody administration did not impact morbidity, mortality, or IFN-γ levels, despite significantly reducing lung viral titers. Therefore, only pre-existing neutralizing antibodies prevent memory CD8 T cell-mediated immunopathology following RSV infection. Overall, our results have important implications for the development of future RSV vaccines.
Collapse
Affiliation(s)
- Megan E Schmidt
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA, 52242, USA
| | - David K Meyerholz
- Department of Pathology, University of Iowa, Iowa City, IA, 52242, USA
| | - Steven M Varga
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA, 52242, USA.
- Department of Pathology, University of Iowa, Iowa City, IA, 52242, USA.
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, 52242, USA.
| |
Collapse
|
10
|
Local heroes or villains: tissue-resident memory T cells in human health and disease. Cell Mol Immunol 2020; 17:113-122. [PMID: 31969685 DOI: 10.1038/s41423-019-0359-1] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 12/17/2019] [Indexed: 12/14/2022] Open
Abstract
Tissue-resident memory T (TRM) cells are increasingly associated with the outcomes of health and disease. TRM cells can mediate local immune protection against infections and cancer, which has led to interest in TRM cells as targets for vaccination and immunotherapies. However, these cells have also been implicated in mediating detrimental pro-inflammatory responses in autoimmune skin diseases such as psoriasis, alopecia areata, and vitiligo. Here, we summarize the biology of TRM cells established in animal models and in translational human studies. We review the beneficial effects of TRM cells in mediating protective responses against infection and cancer and the adverse role of TRM cells in driving pathology in autoimmunity. A further understanding of the breadth and mechanisms of TRM cell activity is essential for the safe design of strategies that manipulate TRM cells, such that protective responses can be enhanced without unwanted tissue damage, and pathogenic TRM cells can be eliminated without losing local immunity.
Collapse
|
11
|
Guvenel A, Jozwik A, Ascough S, Ung SK, Paterson S, Kalyan M, Gardener Z, Bergstrom E, Kar S, Habibi MS, Paras A, Zhu J, Park M, Dhariwal J, Almond M, Wong EH, Sykes A, Del Rosario J, Trujillo-Torralbo MB, Mallia P, Sidney J, Peters B, Kon OM, Sette A, Johnston SL, Openshaw PJ, Chiu C. Epitope-specific airway-resident CD4+ T cell dynamics during experimental human RSV infection. J Clin Invest 2020; 130:523-538. [PMID: 31815739 PMCID: PMC6934186 DOI: 10.1172/jci131696] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 10/08/2019] [Indexed: 01/27/2023] Open
Abstract
BACKGROUNDRespiratory syncytial virus (RSV) is an important cause of acute pulmonary disease and one of the last remaining major infections of childhood for which there is no vaccine. CD4+ T cells play a key role in antiviral immunity, but they have been little studied in the human lung.METHODSHealthy adult volunteers were inoculated i.n. with RSV A Memphis 37. CD4+ T cells in blood and the lower airway were analyzed by flow cytometry and immunohistochemistry. Bronchial soluble mediators were measured using quantitative PCR and MesoScale Discovery. Epitope mapping was performed by IFN-γ ELISpot screening, confirmed by in vitro MHC binding.RESULTSActivated CD4+ T cell frequencies in bronchoalveolar lavage correlated strongly with local C-X-C motif chemokine 10 levels. Thirty-nine epitopes were identified, predominantly toward the 3' end of the viral genome. Five novel MHC II tetramers were made using an immunodominant EFYQSTCSAVSKGYL (F-EFY) epitope restricted to HLA-DR4, -DR9, and -DR11 (combined allelic frequency: 15% in Europeans) and G-DDF restricted to HLA-DPA1*01:03/DPB1*02:01 and -DPA1*01:03/DPB1*04:01 (allelic frequency: 55%). Tetramer labeling revealed enrichment of resident memory CD4+ T (Trm) cells in the lower airway; these Trm cells displayed progressive differentiation, downregulation of costimulatory molecules, and elevated CXCR3 expression as infection evolved.CONCLUSIONSHuman infection challenge provides a unique opportunity to study the breadth of specificity and dynamics of RSV-specific T-cell responses in the target organ, allowing the precise investigation of Trm recognizing novel viral antigens over time. The new tools that we describe enable precise tracking of RSV-specific CD4+ cells, potentially accelerating the development of effective vaccines.TRIAL REGISTRATIONClinicalTrials.gov NCT02755948.FUNDINGMedical Research Council, Wellcome Trust, National Institute for Health Research.
Collapse
Affiliation(s)
| | | | - Stephanie Ascough
- Department of Infectious Disease, Imperial College London, London, United Kingdom
| | - Seng Kuong Ung
- Department of Infectious Disease, Imperial College London, London, United Kingdom
| | - Suzanna Paterson
- Department of Infectious Disease, Imperial College London, London, United Kingdom
| | - Mohini Kalyan
- Department of Infectious Disease, Imperial College London, London, United Kingdom
| | - Zoe Gardener
- Department of Infectious Disease, Imperial College London, London, United Kingdom
| | - Emma Bergstrom
- Department of Infectious Disease, Imperial College London, London, United Kingdom
| | - Satwik Kar
- Department of Infectious Disease, Imperial College London, London, United Kingdom
| | | | | | - Jie Zhu
- National Heart and Lung Institute and
| | | | | | | | | | | | | | | | | | - John Sidney
- Centre for Infectious Disease, Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, California, USA
| | - Bjoern Peters
- Centre for Infectious Disease, Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, California, USA
| | | | - Alessandro Sette
- Centre for Infectious Disease, Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, California, USA
- Department of Medicine, UCSD, La Jolla, California, USA
| | | | | | - Christopher Chiu
- Department of Infectious Disease, Imperial College London, London, United Kingdom
| |
Collapse
|
12
|
Benavente FM, Soto JA, Pizarro-Ortega MS, Bohmwald K, González PA, Bueno SM, Kalergis AM. Contribution of IDO to human respiratory syncytial virus infection. J Leukoc Biol 2019; 106:933-942. [PMID: 31091352 PMCID: PMC7166882 DOI: 10.1002/jlb.4ru0219-051rr] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 04/26/2019] [Accepted: 05/05/2019] [Indexed: 12/18/2022] Open
Abstract
IDO is an enzyme that participates in the degradation of tryptophan (Trp), which is an essential amino acid necessary for vital cellular processes. The degradation of Trp and the metabolites generated by the enzymatic activity of IDO can have immunomodulating effects, notably over T cells, which are particularly sensitive to the absence of Trp and leads to the inhibition of T cell activation, cell death, and the suppression of T cell effector functions. Noteworthy, T cells participate in the cellular immune response against the human respiratory syncytial virus (hRSV) and are essential for viral clearance, as well as the total recovery of the host. Furthermore, inadequate or non‐optimal polarization of T cells is often seen during the acute phase of the disease caused by this pathogen. Here, we discuss the capacity of hRSV to exploit the immunosuppressive features of IDO to reduce T cell function, thus acquiring relevant aspects during the biology of the virus. Additionally, we review studies on the influence of IDO over T cell activation and its relationship with hRSV infection.
Collapse
Affiliation(s)
- Felipe M Benavente
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Jorge A Soto
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Magdalena S Pizarro-Ortega
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Karen Bohmwald
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Pablo A González
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Susan M Bueno
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alexis M Kalergis
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.,Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
13
|
Schmidt ME, Oomens AGP, Varga SM. Vaccination with a Single-Cycle Respiratory Syncytial Virus Is Immunogenic and Protective in Mice. THE JOURNAL OF IMMUNOLOGY 2019; 202:3234-3245. [PMID: 31004010 DOI: 10.4049/jimmunol.1900050] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 04/02/2019] [Indexed: 02/06/2023]
Abstract
Respiratory syncytial virus (RSV) is the leading cause of severe respiratory tract infection in infants and young children, but no vaccine is currently available. Live-attenuated vaccines represent an attractive immunization approach; however, balancing attenuation while retaining sufficient immunogenicity and efficacy has prevented the successful development of such a vaccine. Recently, a recombinant RSV strain lacking the gene that encodes the matrix (M) protein (RSV M-null) was developed. The M protein is required for virion assembly following infection of a host cell but is not necessary for either genome replication or gene expression. Therefore, infection with RSV M-null produces all viral proteins except M but does not generate infectious virus progeny, resulting in a single-cycle infection. We evaluated RSV M-null as a potential vaccine candidate by determining its pathogenicity, immunogenicity, and protective capacity in BALB/c mice compared with its recombinant wild-type control virus (RSV recWT). RSV M-null-infected mice exhibited significantly reduced lung viral titers, weight loss, and pulmonary dysfunction compared with mice infected with RSV recWT. Despite its attenuation, RSV M-null infection induced robust immune responses of similar magnitude to that elicited by RSV recWT. Additionally, RSV M-null infection generated serum Ab and memory T cell responses that were similar to those induced by RSV recWT. Importantly, RSV M-null immunization provided protection against secondary viral challenge by reducing lung viral titers as efficiently as immunization with RSV recWT. Overall, our results indicate that RSV M-null combines attenuation with high immunogenicity and efficacy and represents a promising novel live-attenuated RSV vaccine candidate.
Collapse
Affiliation(s)
- Megan E Schmidt
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA 52242
| | - Antonius G P Oomens
- Center for Veterinary Health Sciences, Oklahoma State University, Stillwater, OK 74078
| | - Steven M Varga
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA 52242; .,Department of Microbiology and Immunology, University of Iowa, Iowa City, IA 52242; and.,Department of Pathology, University of Iowa, Iowa City, IA 52242
| |
Collapse
|
14
|
Schmidt ME, Varga SM. Cytokines and CD8 T cell immunity during respiratory syncytial virus infection. Cytokine 2018; 133:154481. [PMID: 30031680 PMCID: PMC6551303 DOI: 10.1016/j.cyto.2018.07.012] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 07/07/2018] [Indexed: 01/10/2023]
Abstract
Respiratory syncytial virus (RSV) is the leading cause of lower respiratory tract infection and hospitalization in infants. In spite of the enormous clinical burden caused by RSV infections, there remains no efficacious RSV vaccine. CD8 T cells mediate viral clearance as well as provide protection against a secondary RSV infection. However, RSV-specific CD8 T cells may also induce immunopathology leading to exacerbated morbidity and mortality. Many of the crucial functions performed by CD8 T cells are mediated by the cytokines they produce. IFN-γ and TNF are produced by CD8 T cells following RSV infection and contribute to both the acceleration of viral clearance and the induction of immunopathology. To prevent immunopathology, regulatory mechanisms are in place within the immune system to inhibit CD8 T cell effector functions after the infection has been cleared. The actions of a variety of cytokines, including IL-10 and IL-4, play a critical role in the regulation of CD8 T cell effector activity. Herein, we review the current literature on CD8 T cell responses and the functions of the cytokines they produce following RSV infection. Additionally, we discuss the regulation of CD8 T cell activation and effector functions through the actions of various cytokines.
Collapse
Affiliation(s)
- Megan E Schmidt
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA, USA
| | - Steven M Varga
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA, USA; Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, USA; Department of Pathology, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
15
|
Schmidt ME, Varga SM. The CD8 T Cell Response to Respiratory Virus Infections. Front Immunol 2018; 9:678. [PMID: 29686673 PMCID: PMC5900024 DOI: 10.3389/fimmu.2018.00678] [Citation(s) in RCA: 250] [Impact Index Per Article: 41.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 03/20/2018] [Indexed: 12/19/2022] Open
Abstract
Humans are highly susceptible to infection with respiratory viruses including respiratory syncytial virus (RSV), influenza virus, human metapneumovirus, rhinovirus, coronavirus, and parainfluenza virus. While some viruses simply cause symptoms of the common cold, many respiratory viruses induce severe bronchiolitis, pneumonia, and even death following infection. Despite the immense clinical burden, the majority of the most common pulmonary viruses lack long-lasting efficacious vaccines. Nearly all current vaccination strategies are designed to elicit broadly neutralizing antibodies, which prevent severe disease following a subsequent infection. However, the mucosal antibody response to many respiratory viruses is not long-lasting and declines with age. CD8 T cells are critical for mediating clearance following many acute viral infections in the lung. In addition, memory CD8 T cells are capable of providing protection against secondary infections. Therefore, the combined induction of virus-specific CD8 T cells and antibodies may provide optimal protective immunity. Herein, we review the current literature on CD8 T cell responses induced by respiratory virus infections. Additionally, we explore how this knowledge could be utilized in the development of future vaccines against respiratory viruses, with a special emphasis on RSV vaccination.
Collapse
Affiliation(s)
- Megan E Schmidt
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA, United States
| | - Steven M Varga
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA, United States.,Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, United States.,Department of Pathology, University of Iowa, Iowa City, IA, United States
| |
Collapse
|
16
|
Ascough S, Paterson S, Chiu C. Induction and Subversion of Human Protective Immunity: Contrasting Influenza and Respiratory Syncytial Virus. Front Immunol 2018; 9:323. [PMID: 29552008 PMCID: PMC5840263 DOI: 10.3389/fimmu.2018.00323] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Accepted: 02/06/2018] [Indexed: 12/15/2022] Open
Abstract
Respiratory syncytial virus (RSV) and influenza are among the most important causes of severe respiratory disease worldwide. Despite the clinical need, barriers to developing reliably effective vaccines against these viruses have remained firmly in place for decades. Overcoming these hurdles requires better understanding of human immunity and the strategies by which these pathogens evade it. Although superficially similar, the virology and host response to RSV and influenza are strikingly distinct. Influenza induces robust strain-specific immunity following natural infection, although protection by current vaccines is short-lived. In contrast, even strain-specific protection is incomplete after RSV and there are currently no licensed RSV vaccines. Although animal models have been critical for developing a fundamental understanding of antiviral immunity, extrapolating to human disease has been problematic. It is only with recent translational advances (such as controlled human infection models and high-dimensional technologies) that the mechanisms responsible for differences in protection against RSV compared to influenza have begun to be elucidated in the human context. Influenza infection elicits high-affinity IgA in the respiratory tract and virus-specific IgG, which correlates with protection. Long-lived influenza-specific T cells have also been shown to ameliorate disease. This robust immunity promotes rapid emergence of antigenic variants leading to immune escape. RSV differs markedly, as reinfection with similar strains occurs despite natural infection inducing high levels of antibody against conserved antigens. The immunomodulatory mechanisms of RSV are thus highly effective in inhibiting long-term protection, with disturbance of type I interferon signaling, antigen presentation and chemokine-induced inflammation possibly all contributing. These lead to widespread effects on adaptive immunity with impaired B cell memory and reduced T cell generation and functionality. Here, we discuss the differences in clinical outcome and immune response following influenza and RSV. Specifically, we focus on differences in their recognition by innate immunity; the strategies used by each virus to evade these early immune responses; and effects across the innate-adaptive interface that may prevent long-lived memory generation. Thus, by comparing these globally important pathogens, we highlight mechanisms by which optimal antiviral immunity may be better induced and discuss the potential for these insights to inform novel vaccines.
Collapse
Affiliation(s)
- Stephanie Ascough
- Section of Infectious Diseases and Immunity, Imperial College London, London, United Kingdom
| | - Suzanna Paterson
- Section of Infectious Diseases and Immunity, Imperial College London, London, United Kingdom
| | - Christopher Chiu
- Section of Infectious Diseases and Immunity, Imperial College London, London, United Kingdom
| |
Collapse
|
17
|
Memory CD8 T cells mediate severe immunopathology following respiratory syncytial virus infection. PLoS Pathog 2018; 14:e1006810. [PMID: 29293660 PMCID: PMC5766251 DOI: 10.1371/journal.ppat.1006810] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 01/12/2018] [Accepted: 12/13/2017] [Indexed: 12/20/2022] Open
Abstract
Memory CD8 T cells can provide protection from re-infection by respiratory viruses such as influenza and SARS. However, the relative contribution of memory CD8 T cells in providing protection against respiratory syncytial virus (RSV) infection is currently unclear. To address this knowledge gap, we utilized a prime-boost immunization approach to induce robust memory CD8 T cell responses in the absence of RSV-specific CD4 T cells and antibodies. Unexpectedly, RSV infection of mice with pre-existing CD8 T cell memory led to exacerbated weight loss, pulmonary disease, and lethal immunopathology. The exacerbated disease in immunized mice was not epitope-dependent and occurred despite a significant reduction in RSV viral titers. In addition, the lethal immunopathology was unique to the context of an RSV infection as mice were protected from a normally lethal challenge with a recombinant influenza virus expressing an RSV epitope. Memory CD8 T cells rapidly produced IFN-γ following RSV infection resulting in elevated protein levels in the lung and periphery. Neutralization of IFN-γ in the respiratory tract reduced morbidity and prevented mortality. These results demonstrate that in contrast to other respiratory viruses, RSV-specific memory CD8 T cells can induce lethal immunopathology despite mediating enhanced viral clearance. Memory CD8 T cells have been shown to provide protection against many respiratory viruses. However, the ability of memory CD8 T cells to provide protection against RSV has not been extensively examined. Unexpectedly, mice with pre-existing CD8 T cell memory, in the absence of memory CD4 T cells and antibodies, exhibited exacerbated morbidity and mortality following RSV infection. We demonstrate that the immunopathology is the result of early and excessive production of IFN-γ by memory CD8 T cells in the lung. Our research provides important new insight into the mechanisms of how memory T cells induce immunopathology. In addition, our findings serve as an important cautionary tale against the use of epitope-based T cell vaccines against RSV.
Collapse
|
18
|
Lee JY, Chang J. Recombinant baculovirus-based vaccine expressing M2 protein induces protective CD8 + T-cell immunity against respiratory syncytial virus infection. J Microbiol 2017; 55:900-908. [PMID: 29076066 DOI: 10.1007/s12275-017-7306-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 09/10/2017] [Accepted: 10/11/2017] [Indexed: 12/20/2022]
Abstract
Respiratory syncytial virus (RSV) is an important cause of acute lower respiratory tract disease in infants, young children, immunocompromised individuals, and the elderly. However, despite ongoing efforts to develop an RSV vaccine, there is still no authorized RSV vaccine for humans. Baculovirus has attracted attention as a vaccine vector because of its ability to induce a high level of humoral and cellular immunity, low cytotoxicity against various antigens, and biological safety for humans. In this study, we constructed a recombinant baculovirus- based vaccine expressing the M2 protein of RSV under the control of cytomegalovirus promoter (Bac_RSVM2) to induce CD8+ T-cell responses which play an important role in viral clearance, and investigated its protective efficacy against RSV infection. Immunization with Bac_RSVM2 via intranasal or intramuscular route effectively elicited the specific CD8+ T-cell responses. Most notably, immunization with Bac_RSVM2 vaccine almost completely protected mice from RSV challenge without vaccine-enhanced immunopathology. In conclusion, these results suggest that Bac_RSVM2 vaccine employing the baculovirus delivery platform has promising potential to be developed as a safe and novel RSV vaccine that provides protection against RSV infection.
Collapse
Affiliation(s)
- Jeong-Yoon Lee
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Jun Chang
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760, Republic of Korea.
| |
Collapse
|
19
|
Pulmonary immunity to viruses. Clin Sci (Lond) 2017; 131:1737-1762. [PMID: 28667071 DOI: 10.1042/cs20160259] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 03/31/2017] [Accepted: 04/06/2017] [Indexed: 12/28/2022]
Abstract
Mucosal surfaces, such as the respiratory epithelium, are directly exposed to the external environment and therefore, are highly susceptible to viral infection. As a result, the respiratory tract has evolved a variety of innate and adaptive immune defenses in order to prevent viral infection or promote the rapid destruction of infected cells and facilitate the clearance of the infecting virus. Successful adaptive immune responses often lead to a functional state of immune memory, in which memory lymphocytes and circulating antibodies entirely prevent or lessen the severity of subsequent infections with the same virus. This is also the goal of vaccination, although it is difficult to vaccinate in a way that mimics respiratory infection. Consequently, some vaccines lead to robust systemic immune responses, but relatively poor mucosal immune responses that protect the respiratory tract. In addition, adaptive immunity is not without its drawbacks, as overly robust inflammatory responses may lead to lung damage and impair gas exchange or exacerbate other conditions, such as asthma or chronic obstructive pulmonary disease (COPD). Thus, immune responses to respiratory viral infections must be strong enough to eliminate infection, but also have mechanisms to limit damage and promote tissue repair in order to maintain pulmonary homeostasis. Here, we will discuss the components of the adaptive immune system that defend the host against respiratory viral infections.
Collapse
|
20
|
Shi H, Ren K, Lv B, Zhang W, Zhao Y, Tan RX, Li E. Baicalin from Scutellaria baicalensis blocks respiratory syncytial virus (RSV) infection and reduces inflammatory cell infiltration and lung injury in mice. Sci Rep 2016; 6:35851. [PMID: 27767097 PMCID: PMC5073294 DOI: 10.1038/srep35851] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 10/06/2016] [Indexed: 12/26/2022] Open
Abstract
The roots of Scutellaria baicalensis has been used as a remedy for inflammatory and infective diseases for thousands of years. We evaluated the antiviral activity against respiratory syncytial virus (RSV) infection, the leading cause of childhood infection and hospitalization. By fractionation and chromatographic analysis, we determined that baicalin was responsible for the antiviral activity of S. baicalensis against RSV infection. The concentration for 50% inhibition (IC50) of RSV infection was determined at 19.9 ± 1.8 μM, while the 50% cytotoxic concentration (CC50) was measured at 370 ± 10 μM. We then used a mouse model of RSV infection to further demonstrate baicalin antiviral effect. RSV infection caused significant lung injury and proinflammatory response, including CD4 and CD8 T lymphocyte infiltration. Baicalin treatment resulted in reduction of T lymphocyte infiltration and gene expression of proinflammatory factors, while the treatment moderately reduced RSV titers recovered from the lung tissues. T lymphocyte infiltration and cytotoxic T lymphocyte modulated tissue damage has been identified critical factors of RSV disease. The study therefore demonstrates that baicalin subjugates RSV disease through antiviral and anti-inflammatory effect.
Collapse
Affiliation(s)
- Hengfei Shi
- Medical School and State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China.,Jiangsu Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, China
| | - Ke Ren
- Medical School and State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China.,Jiangsu Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, China
| | - Baojie Lv
- Medical School and State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China.,College of Life Sciences, Nanjing University, Nanjing, China
| | - Wei Zhang
- Nanjing University of Chinese Medicine, Nanjing, China
| | - Ying Zhao
- Medical School and State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China
| | - Ren Xiang Tan
- Medical School and State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China.,College of Life Sciences, Nanjing University, Nanjing, China.,Nanjing University of Chinese Medicine, Nanjing, China
| | - Erguang Li
- Medical School and State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China.,Jiangsu Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, China
| |
Collapse
|
21
|
Jozwik A, Habibi MS, Paras A, Zhu J, Guvenel A, Dhariwal J, Almond M, Wong EHC, Sykes A, Maybeno M, Del Rosario J, Trujillo-Torralbo MB, Mallia P, Sidney J, Peters B, Kon OM, Sette A, Johnston SL, Openshaw PJ, Chiu C. RSV-specific airway resident memory CD8+ T cells and differential disease severity after experimental human infection. Nat Commun 2015; 6:10224. [PMID: 26687547 PMCID: PMC4703893 DOI: 10.1038/ncomms10224] [Citation(s) in RCA: 220] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Accepted: 11/16/2015] [Indexed: 12/30/2022] Open
Abstract
In animal models, resident memory CD8+ T (Trm) cells assist in respiratory virus elimination but their importance in man has not been determined. Here, using experimental human respiratory syncytial virus (RSV) infection, we investigate systemic and local virus-specific CD8+ T-cell responses in adult volunteers. Having defined the immunodominance hierarchy, we analyse phenotype and function longitudinally in blood and by serial bronchoscopy. Despite rapid clinical recovery, we note surprisingly extensive lower airway inflammation with persistent viral antigen and cellular infiltrates. Pulmonary virus-specific CD8+ T cells display a CD69+CD103+ Trm phenotype and accumulate to strikingly high frequencies into convalescence without continued proliferation. While these have a more highly differentiated phenotype, they express fewer cytotoxicity markers than in blood. Nevertheless, their abundance before infection correlates with reduced symptoms and viral load, implying that CD8+ Trm cells in the human lung can confer protection against severe respiratory viral disease when humoral immunity is overcome.
Collapse
Affiliation(s)
- Agnieszka Jozwik
- National Heart and Lung Institute, Imperial College London, London W2 1PG, UK
| | | | - Allan Paras
- National Heart and Lung Institute, Imperial College London, London W2 1PG, UK
| | - Jie Zhu
- National Heart and Lung Institute, Imperial College London, London W2 1PG, UK
| | - Aleks Guvenel
- National Heart and Lung Institute, Imperial College London, London W2 1PG, UK
| | - Jaideep Dhariwal
- National Heart and Lung Institute, Imperial College London, London W2 1PG, UK
| | - Mark Almond
- National Heart and Lung Institute, Imperial College London, London W2 1PG, UK
| | - Ernie H. C. Wong
- National Heart and Lung Institute, Imperial College London, London W2 1PG, UK
| | - Annemarie Sykes
- National Heart and Lung Institute, Imperial College London, London W2 1PG, UK
| | - Matthew Maybeno
- Centre for Infectious Disease, Division of Vaccine Discovery, La Jolla Institute of Allergy and Immunology, 9420 Athena Circle, La Jolla, California 92037, USA
| | - Jerico Del Rosario
- National Heart and Lung Institute, Imperial College London, London W2 1PG, UK
| | | | - Patrick Mallia
- National Heart and Lung Institute, Imperial College London, London W2 1PG, UK
| | - John Sidney
- Centre for Infectious Disease, Division of Vaccine Discovery, La Jolla Institute of Allergy and Immunology, 9420 Athena Circle, La Jolla, California 92037, USA
| | - Bjoern Peters
- Centre for Infectious Disease, Division of Vaccine Discovery, La Jolla Institute of Allergy and Immunology, 9420 Athena Circle, La Jolla, California 92037, USA
| | - Onn Min Kon
- National Heart and Lung Institute, Imperial College London, London W2 1PG, UK
| | - Alessandro Sette
- Centre for Infectious Disease, Division of Vaccine Discovery, La Jolla Institute of Allergy and Immunology, 9420 Athena Circle, La Jolla, California 92037, USA
| | | | - Peter J. Openshaw
- National Heart and Lung Institute, Imperial College London, London W2 1PG, UK
| | - Christopher Chiu
- National Heart and Lung Institute, Imperial College London, London W2 1PG, UK
| |
Collapse
|
22
|
Dixon DL. The Role of Human Milk Immunomodulators in Protecting Against Viral Bronchiolitis and Development of Chronic Wheezing Illness. CHILDREN (BASEL, SWITZERLAND) 2015; 2:289-304. [PMID: 27417364 PMCID: PMC4928768 DOI: 10.3390/children2030289] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Revised: 06/21/2015] [Accepted: 06/29/2015] [Indexed: 12/26/2022]
Abstract
Infants who are breastfed are at an immunological advantage when compared with formula fed infants, evidenced by decreased incidence of infections and diminished propensity for long term conditions, including chronic wheeze and/or asthma. Exclusive breastfeeding reduces the duration of hospital admission, risk of respiratory failure and requirement for supplemental oxygen in infants hospitalised with bronchiolitis suggesting a potentially protective mechanism. This review examines the evidence and potential pathways for protection by immunomodulatory factors in human milk against the most common viral cause of bronchiolitis, respiratory syncytial virus (RSV), and subsequent recurrent wheeze in infants. Further investigations into the interplay between respiratory virus infections such as RSV and how they affect, and are affected by, human milk immunomodulators is necessary if we are to gain a true understanding of how breastfeeding protects many infants but not all against infections, and how this relates to long-term protection against conditions such as chronic wheezing illness or asthma.
Collapse
Affiliation(s)
- Dani-Louise Dixon
- Intensive and Critical Care Unit, Flinders Medical Centre, Adelaide 5042, Australia.
- Department of Critical Care Medicine, Flinders University, Adelaide 5001, Australia .
| |
Collapse
|
23
|
Lanari M, Vandini S, Capretti MG, Lazzarotto T, Faldella G. Respiratory syncytial virus infections in infants affected by primary immunodeficiency. J Immunol Res 2014; 2014:850831. [PMID: 25089282 PMCID: PMC4095650 DOI: 10.1155/2014/850831] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Revised: 06/11/2014] [Accepted: 06/12/2014] [Indexed: 12/30/2022] Open
Abstract
Primary immunodeficiencies are rare inherited disorders that may lead to frequent and often severe acute respiratory infections. Respiratory syncytial virus (RSV) is one of the most frequent pathogens during early infancy and the infection is more severe in immunocompromised infants than in healthy infants, as a result of impaired T- and B-cell immune response unable to efficaciously neutralize viral replication, with subsequent increased viral shedding and potentially lethal lower respiratory tract infection. Several authors have reported a severe clinical course after RSV infections in infants and children with primary and acquired immunodeficiencies. Environmental prophylaxis is essential in order to reduce the infection during the epidemic season in hospitalized immunocompromised infants. Prophylaxis with palivizumab, a humanized monoclonal antibody against the RSV F protein, is currently recommended in high-risk infants born prematurely, with chronic lung disease or congenital heart disease. Currently however the prophylaxis is not routinely recommended in infants with primary immunodeficiency, although some authors propose the extension of prophylaxis to this high risk population.
Collapse
Affiliation(s)
- Marcello Lanari
- Pediatrics and Neonatology Unit, Imola Hospital, Via Montericco 4, 40026 Imola, Italy
| | - Silvia Vandini
- DIMEC, Neonatology and Neonatal Intensive Care Unit, St. Orsola-Malpighi Hospital, Via Massarenti 11, University of Bologna, 40138 Bologna, Italy
| | - Maria Grazia Capretti
- DIMEC, Neonatology and Neonatal Intensive Care Unit, St. Orsola-Malpighi Hospital, Via Massarenti 11, University of Bologna, 40138 Bologna, Italy
| | - Tiziana Lazzarotto
- DIMES, Clinical Microbiology Unit, Laboratory of Virology, St. Orsola-Malpighi Hospital, Via Massarenti 9, University of Bologna, Bologna, Italy
| | - Giacomo Faldella
- DIMEC, Neonatology and Neonatal Intensive Care Unit, St. Orsola-Malpighi Hospital, Via Massarenti 11, University of Bologna, 40138 Bologna, Italy
| |
Collapse
|
24
|
Abstract
Respiratory syncytial virus (RSV) is amongst the most important pathogenic infections of childhood and is associated with significant morbidity and mortality. Although there have been extensive studies of epidemiology, clinical manifestations, diagnostic techniques, animal models and the immunobiology of infection, there is not yet a convincing and safe vaccine available. The major histopathologic characteristics of RSV infection are acute bronchiolitis, mucosal and submucosal edema, and luminal occlusion by cellular debris of sloughed epithelial cells mixed with macrophages, strands of fibrin, and some mucin. There is a single RSV serotype with two major antigenic subgroups, A and B. Strains of both subtypes often co-circulate, but usually one subtype predominates. In temperate climates, RSV infections reflect a distinct seasonality with onset in late fall or early winter. It is believed that most children will experience at least one RSV infection by the age of 2 years. There are several key animal models of RSV. These include a model in mice and, more importantly, a bovine model; the latter reflects distinct similarity to the human disease. Importantly, the prevalence of asthma is significantly higher amongst children who are hospitalized with RSV in infancy or early childhood. However, there have been only limited investigations of candidate genes that have the potential to explain this increase in susceptibility. An atopic predisposition appears to predispose to subsequent development of asthma and it is likely that subsequent development of asthma is secondary to the pathogenic inflammatory response involving cytokines, chemokines and their cognate receptors. Numerous approaches to the development of RSV vaccines are being evaluated, as are the use of newer antiviral agents to mitigate disease. There is also significant attention being placed on the potential impact of co-infection and defining the natural history of RSV. Clearly, more research is required to define the relationships between RSV bronchiolitis, other viral induced inflammatory responses, and asthma.
Collapse
Affiliation(s)
- Andrea T. Borchers
- Division of Rheumatology, Allergy and Clinical Immunology, University of California at Davis School of Medicine, 451 Health Sciences Drive, Suite 6501, Davis, CA 95616 USA
| | - Christopher Chang
- Division of Rheumatology, Allergy and Clinical Immunology, University of California at Davis School of Medicine, 451 Health Sciences Drive, Suite 6501, Davis, CA 95616 USA
| | - M. Eric Gershwin
- Division of Rheumatology, Allergy and Clinical Immunology, University of California at Davis School of Medicine, 451 Health Sciences Drive, Suite 6501, Davis, CA 95616 USA
| | - Laurel J. Gershwin
- Department of Pathology, Microbiology and Immunology, University of California, Davis, School of Veterinary Medicine, Davis, CA USA
| |
Collapse
|
25
|
Neonatal antibody responses are attenuated by interferon-γ produced by NK and T cells during RSV infection. Proc Natl Acad Sci U S A 2013; 110:5576-81. [PMID: 23509276 DOI: 10.1073/pnas.1214247110] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Respiratory syncytial virus (RSV) infects most children in the first year of life and is a major single cause of hospitalization in infants and young children. There is no effective vaccine, and antibody generated by primary neonatal infection is poorly protective against reinfection even with antigenically homologous viral strains. Studying the immunological basis of these observations in neonatal mice, we found that antibody responses to infection were low and unaffected by CD4 depletion, in contrast with adult mice, which had stronger CD4-dependent antibody responses. Natural killer cell depletion or codepletion of CD4(+) and CD8(+) cells during neonatal RSV infection caused a striking increase in anti-RSV antibody titer. These cells are major sources of the cytokine IFN-γ, and blocking IFN-γ also enhanced RSV-specific antibody responses in neonates. In addition, infection with a recombinant RSV engineered to produce IFN-γ reduced antibody titer, confirming that IFN-γ plays a pivotal role in inhibition of antibody responses after neonatal infection. These unexpected findings show that the induction of a strong cellular immune response may limit antibody responses in early life and that vaccines that induce IFN-γ-secreting cells might, in some situations, be less protective than those that do not.
Collapse
|
26
|
Modulation of tumor immunity by soluble and membrane-bound molecules at the immunological synapse. Clin Dev Immunol 2013; 2013:450291. [PMID: 23533456 PMCID: PMC3606757 DOI: 10.1155/2013/450291] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2012] [Accepted: 01/15/2013] [Indexed: 12/31/2022]
Abstract
To circumvent pathology caused by infectious microbes and tumor growth, the host immune system must constantly clear harmful microorganisms and potentially malignant transformed cells. This task is accomplished in part by T-cells, which can directly kill infected or tumorigenic cells. A crucial event determining the recognition and elimination of detrimental cells is antigen recognition by the T cell receptor (TCR) expressed on the surface of T cells. Upon binding of the TCR to cognate peptide-MHC complexes presented on the surface of antigen presenting cells (APCs), a specialized supramolecular structure known as the immunological synapse (IS) assembles at the T cell-APC interface. Such a structure involves massive redistribution of membrane proteins, including TCR/pMHC complexes, modulatory receptor pairs, and adhesion molecules. Furthermore, assembly of the immunological synapse leads to intracellular events that modulate and define the magnitude and characteristics of the T cell response. Here, we discuss recent literature on the regulation and assembly of IS and the mechanisms evolved by tumors to modulate its function to escape T cell cytotoxicity, as well as novel strategies targeting the IS for therapy.
Collapse
|
27
|
van Helden MJ, van Kooten PJ, Bekker CP, Gröne A, Topham DJ, Easton AJ, Boog CJ, Busch DH, Zaiss DM, Sijts AJ. Pre-existing virus-specific CD8(+) T-cells provide protection against pneumovirus-induced disease in mice. Vaccine 2012; 30:6382-8. [PMID: 22940382 PMCID: PMC3465553 DOI: 10.1016/j.vaccine.2012.08.027] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2012] [Revised: 07/25/2012] [Accepted: 08/15/2012] [Indexed: 12/23/2022]
Abstract
Pneumoviruses such as pneumonia virus of mice (PVM), bovine respiratory syncytial virus (bRSV) or human (h)RSV are closely related pneumoviruses that cause severe respiratory disease in their respective hosts. It is well-known that T-cell responses are essential in pneumovirus clearance, but pneumovirus-specific T-cell responses also are important mediators of severe immunopathology. In this study we determined whether memory- or pre-existing, transferred virus-specific CD8(+) T-cells provide protection against PVM-induced disease. We show that during infection with a sublethal dose of PVM, both natural killer (NK) cells and CD8(+) T-cells expand relatively late. Induction of CD8(+) T-cell memory against a single CD8(+) T-cell epitope, by dendritic cell (DC)-peptide immunization, leads to partial protection against PVM challenge and prevents Th2 differentiation of PVM-induced CD4 T-cells. In addition, adoptively transferred PVM-specific CD8(+) T-cells, covering the entire PVM-specific CD8(+) T-cell repertoire, provide partial protection from PVM-induced disease. From these data we infer that antigen-specific memory CD8(+) T-cells offer significant protection to PVM-induced disease. Thus, CD8(+) T-cells, despite being a major cause of PVM-associated pathology during primary infection, may offer promising targets of a protective pneumovirus vaccine.
Collapse
Key Words
- bal, bronchoalveolar lavage
- balf, bal fluid
- dc, dendritic cell
- bm-dc, bone marrow derived dc
- dcp, peptide-loaded dc
- fi, formalin inactivated
- hrsv, human respiratory syncytial virus
- id, infectious dose
- eid, egg id
- i.n., intranasal
- i.p., intraperitoneal
- i.v., intravenous
- mln, mediastinal lymph node
- nk, natural killer
- ns, nonstructural
- p.i., post infection
- pfu, plaque forming units
- pvm, pneunomia virus of mice
- sem, standard error of mean
- pneumoviruses
- pneunomia virus of mice
- nk cell
- cd8+ t-cell
- vaccine
Collapse
Affiliation(s)
- Mary J.G. van Helden
- Division of Immunology, Faculty of Veterinary Medicine, University of Utrecht, Yalelaan 1, 3584 CL Utrecht, The Netherlands
| | - Peter J.S. van Kooten
- Division of Immunology, Faculty of Veterinary Medicine, University of Utrecht, Yalelaan 1, 3584 CL Utrecht, The Netherlands
| | - Cornelis P.J. Bekker
- Division of Immunology, Faculty of Veterinary Medicine, University of Utrecht, Yalelaan 1, 3584 CL Utrecht, The Netherlands
| | - Andrea Gröne
- Division of Pathology, Faculty of Veterinary Medicine, University of Utrecht, Yalelaan 1, 3584 CL Utrecht, The Netherlands
| | - David J. Topham
- D. Smith Center for Vaccine Biology and Immunology, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY 14642, USA
| | - Andrew J. Easton
- School of Life Sciences, Gibbet Hill Campus, University of Warwick, Coventry CV4 7AL, UK
| | - Claire J.P. Boog
- Department of Vaccinology, Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Antonie van Leeuwenhoeklaan 9, 3721 MA Bilthoven, The Netherlands
| | - Dirk H. Busch
- Institute for Medical Microbiology, Immunology and Hygiene, Technische Universität München, Trogerstrasse 30, 81675 München, Germany
| | - Dietmar M.W. Zaiss
- Division of Immunology, Faculty of Veterinary Medicine, University of Utrecht, Yalelaan 1, 3584 CL Utrecht, The Netherlands
| | - Alice J.A.M. Sijts
- Division of Immunology, Faculty of Veterinary Medicine, University of Utrecht, Yalelaan 1, 3584 CL Utrecht, The Netherlands
| |
Collapse
|
28
|
Kim JY, Chang J. Need for a safe vaccine against respiratory syncytial virus infection. KOREAN JOURNAL OF PEDIATRICS 2012; 55:309-15. [PMID: 23049587 PMCID: PMC3454572 DOI: 10.3345/kjp.2012.55.9.309] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/13/2012] [Accepted: 05/15/2012] [Indexed: 11/27/2022]
Abstract
Human respiratory syncytial virus (HRSV) is a major cause of severe respiratory tract illnesses in infants and young children worldwide. Despite its importance as a respiratory pathogen, there is currently no licensed vaccine for HRSV. Following failure of the initial trial of formalin-inactivated virus particle vaccine, continuous efforts have been made for the development of safe and efficacious vaccines against HRSV. However, several obstacles persist that delay the development of HRSV vaccine, such as the immature immune system of newborn infants and the possible Th2-biased immune responses leading to subsequent vaccine-enhanced diseases. Many HRSV vaccine strategies are currently being developed and evaluated, including live-attenuated viruses, subunit-based, and vector-based candidates. In this review, the current HRSV vaccines are overviewed and the safety issues regarding asthma and vaccine-induced pathology are discussed.
Collapse
Affiliation(s)
- Joo-Young Kim
- Division of Life & Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul, Korea
| | | |
Collapse
|
29
|
Kim JY, Chang J. Need for a safe vaccine against respiratory syncytial virus infection. KOREAN JOURNAL OF PEDIATRICS 2012. [DOI: 10.3345/kjp.2012.55.9.359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Affiliation(s)
- Joo-Young Kim
- Division of Life & Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul, Korea
| | - Jun Chang
- Division of Life & Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul, Korea
| |
Collapse
|
30
|
T cells are not required for pathogenesis in the Syrian hamster model of hantavirus pulmonary syndrome. J Virol 2011; 85:9929-44. [PMID: 21775442 DOI: 10.1128/jvi.05356-11] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Andes virus (ANDV) is associated with a lethal vascular leak syndrome in humans termed hantavirus pulmonary syndrome (HPS). In hamsters, ANDV causes a respiratory distress syndrome closely resembling human HPS. The mechanism for the massive vascular leakage associated with HPS is poorly understood; however, T cell immunopathology has been implicated on the basis of circumstantial and corollary evidence. Here, we show that following ANDV challenge, hamster T cell activation corresponds with the onset of disease. However, treatment with cyclophosphamide or specific T cell depletion does not impact the course of disease or alter the number of surviving animals, despite significant reductions in T cell number. These data demonstrate, for the first time, that T cells are not required for hantavirus pathogenesis in the hamster model of human HPS. Depletion of T cells from Syrian hamsters did not significantly influence early events in disease progression. Moreover, these data argue for a mechanism of hantavirus-induced vascular permeability that does not involve T cell immunopathology.
Collapse
|
31
|
Assembly and immunological properties of Newcastle disease virus-like particles containing the respiratory syncytial virus F and G proteins. J Virol 2010; 85:366-77. [PMID: 20980510 DOI: 10.1128/jvi.01861-10] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Human respiratory syncytial virus (RSV) is a serious respiratory pathogen in infants and young children as well as elderly and immunocompromised populations. However, no RSV vaccines are available. We have explored the potential of virus-like particles (VLPs) as an RSV vaccine candidate. VLPs composed entirely of RSV proteins were produced at levels inadequate for their preparation as immunogens. However, VLPs composed of the Newcastle disease virus (NDV) nucleocapsid and membrane proteins and chimera proteins containing the ectodomains of RSV F and G proteins fused to the transmembrane and cytoplasmic domains of NDV F and HN proteins, respectively, were quantitatively prepared from avian cells. Immunization of mice with these VLPs, without adjuvant, stimulated robust, anti-RSV F and G protein antibody responses. IgG2a/IgG1 ratios were very high, suggesting predominantly T(H)1 responses. In contrast to infectious RSV immunization, neutralization antibody titers were robust and stable for 4 months. Immunization with a single dose of VLPs resulted in the complete protection of mice from RSV replication in lungs. Upon RSV intranasal challenge of VLP-immunized mice, no enhanced lung pathology was observed, in contrast to the pathology observed in mice immunized with formalin-inactivated RSV. These results suggest that these VLPs are effective RSV vaccines in mice, in contrast to other nonreplicating RSV vaccine candidates.
Collapse
|
32
|
Fu YH, He JS, Wang XB, Zheng XX, Wu Q, Xie C, Zhang M, Wei W, Tang Q, Song JD, Qu JG, Hong T. A prime–boost vaccination strategy using attenuated Salmonella typhimurium and a replication-deficient recombinant adenovirus vector elicits protective immunity against human respiratory syncytial virus. Biochem Biophys Res Commun 2010; 395:87-92. [DOI: 10.1016/j.bbrc.2010.03.144] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2010] [Accepted: 03/24/2010] [Indexed: 10/19/2022]
|
33
|
Kim S, Jang JE, Yu JR, Chang J. Single mucosal immunization of recombinant adenovirus-based vaccine expressing F1 protein fragment induces protective mucosal immunity against respiratory syncytial virus infection. Vaccine 2010; 28:3801-8. [PMID: 20362203 DOI: 10.1016/j.vaccine.2010.03.032] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2009] [Revised: 03/04/2010] [Accepted: 03/17/2010] [Indexed: 11/24/2022]
Abstract
Respiratory syncytial virus (RSV) is a major cause of severe lower respiratory tract disease in infancy and early childhood. Despite its importance as a pathogen, there is no licensed vaccine against RSV. The fusion (F) protein of RSV is a potentially important target for protective antiviral immune responses. Here, we studied the immune responses elicited by recombinant replication-deficient adenovirus (rAd)-based vaccines expressing the soluble F1 fragment of F protein (amino acids 155-524) in murine model. The expression of secreted F1 fragment by rAd was significantly increased by codon optimization. Strong mucosal IgA response was induced by single intranasal immunization of codon-optimized vaccine, rAd/F1co, but not by rAd/F1wt. A single intranasal immunization with rAd/F1co provided potent protection against subsequent RSV challenge. Interestingly, neither serum Ig nor T-cell response directed to F protein was detected in the rAd/F1co-immune mice, suggesting that protective immunity by rAd/F1co is mainly mediated through mucosal IgA induction. Indeed, co-delivery of cholera toxin B subunit significantly enhanced mucosal IgA responses by the optimized vaccine, which correlates with protective efficacy. Taken together, our data demonstrate that a single intranasal administration of rAd/F1co is sufficient for the protection and represents a promising prophylactic vaccination regimen against RSV infection.
Collapse
Affiliation(s)
- Sol Kim
- Division of Life & Pharmaceutical Sciences, and the Center for Cell Signaling & Drug Discovery Research, Ewha Womans University, 11-1 Dae-Hyun Dong, Seo-Dae-Mun Gu, Seoul 120-750, Republic of Korea
| | | | | | | |
Collapse
|
34
|
Li Y, Dinwiddie DL, Harrod KS, Jiang Y, Kim KC. Anti-inflammatory effect of MUC1 during respiratory syncytial virus infection of lung epithelial cells in vitro. Am J Physiol Lung Cell Mol Physiol 2010; 298:L558-63. [PMID: 20081068 DOI: 10.1152/ajplung.00225.2009] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
MUC1 is a transmembrane glycoprotein expressed on the apical surface of airway epithelial cells and plays an anti-inflammatory role during airway bacterial infection. In this study, we determined whether the anti-inflammatory effect of MUC1 is also operative during the respiratory syncytial virus (RSV) infection. The lung epithelial cell line A549 was treated with RSV, and the production of TNFalpha and the levels of MUC1 protein were monitored temporally during the course of infection by ELISA and Western blot analysis. Small inhibitory RNA (siRNA) transfection was utilized to assess the role of MUC1 in regulating RSV-mediated inflammatory responses by lung epithelial cells. Our results revealed that: 1) following RSV infection, an increase in MUC1 level was preceded by an increase in TNFalpha production and completely inhibited by soluble TNF receptor (TNFR); and 2) knockdown of MUC1 using MUC1 siRNA resulted in a greater increase in TNFalpha level following RSV infection compared with control siRNA treatment. We conclude that the RSV-induced increase in the TNFalpha levels upregulates MUC1 through its interaction with TNFR, which in turn suppresses further increase in TNFalpha by RSV, thus forming a negative feedback loop in the control of RSV-induced inflammation. This is the first demonstration showing that MUC1 can suppress the virus-induced inflammatory responses.
Collapse
Affiliation(s)
- Yusheng Li
- Department of Physiology and Lung Center, Temple University School of Medicine, Philadelphia, Pennsylvania 19140, USA
| | | | | | | | | |
Collapse
|
35
|
Abstract
Respiratory syncytial virus (RSV) is a clinically significant cause of respiratory tract disease, especially among high-risk infants and immunocompromised and elderly adults. Despite the burden of disease, there is no licensed prophylactic RSV vaccine. The initial efforts to develop an RSV vaccine involved formalin-inactivated virus preparations that unexpectedly caused vaccine-enhanced disease in clinical trials in RSV-naive children. Over the last 40 years, cautious and deliberate progress has been made toward RSV vaccine development using various experimental approaches, including live attenuated strains and vector-based and viral protein subunit/DNA-based candidates. The scientific rationale, preclinical testing, and clinical development of each of these approaches are reviewed.
Collapse
Affiliation(s)
- Yoshihiko Murata
- Division of Infectious Diseases, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA.
| |
Collapse
|
36
|
Newcastle disease virus-like particles containing respiratory syncytial virus G protein induced protection in BALB/c mice, with no evidence of immunopathology. J Virol 2009; 84:1110-23. [PMID: 19889768 DOI: 10.1128/jvi.01709-09] [Citation(s) in RCA: 110] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Respiratory syncytial virus (RSV) is the leading cause of serious respiratory infections in children as well as a serious cause of disease in elderly and immunosuppressed populations. There are no licensed vaccines available to prevent RSV disease. We have developed a virus-like particle (VLP) vaccine candidate for protection from RSV. The VLP is composed of the NP and M proteins of Newcastle disease virus (NDV) and a chimeric protein containing the cytoplasmic and transmembrane domains of the NDV HN protein and the ectodomain of the human RSV G protein (H/G). Immunization of mice with 10 or 40 microg total VLP-H/G protein by intraperitoneal or intramuscular inoculation stimulated antibody responses to G protein which were as good as or better than those stimulated by comparable amounts of UV-inactivated RSV. Immunization of mice with two doses or even a single dose of these particles resulted in the complete protection of mice from RSV replication in the lungs. Immunization with these particles induced neutralizing antibodies with modest titers. Upon RSV challenge of VLP-H/G-immunized mice, no enhanced pathology in the lungs was observed, although lungs of mice immunized in parallel with formalin-inactivated RSV (FI-RSV) showed the significant pathology that has previously been documented after immunization with FI-RSV. Thus, the VLP-H/G candidate vaccine was immunogenic in BALB/c mice and prevented replication of RSV in murine lungs, with no evidence of immunopathology. These data support further development of virus-like particle vaccine candidates for protection against RSV.
Collapse
|
37
|
Kovacs-Nolan J, Mapletoft JW, Lawman Z, Babiuk LA, van Drunen Littel-van den Hurk S. Formulation of bovine respiratory syncytial virus fusion protein with CpG oligodeoxynucleotide, cationic host defence peptide and polyphosphazene enhances humoral and cellular responses and induces a protective type 1 immune response in mice. J Gen Virol 2009; 90:1892-1905. [PMID: 19386785 DOI: 10.1099/vir.0.011684-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Respiratory syncytial virus (RSV) is the leading cause of serious respiratory tract disease in children and calves; however, RSV vaccine development has been slow due to early observations that formalin-inactivated vaccines induced Th2-type immune responses and led to disease enhancement upon subsequent exposure. Hence, there is a need for novel adjuvants that will promote a protective Th1-type or balanced immune response against RSV. CpG oligodeoxynucleotides (ODNs), indolicidin, and polyphosphazene were examined for their ability to enhance antigen-specific immune responses and influence the Th-bias when co-formulated with a recombinant truncated bovine RSV (BRSV) fusion protein (DeltaF). Mice immunized with DeltaF co-formulated with CpG ODN, indolicidin, and polyphosphazene (DeltaF/CpG/indol/PP) developed higher levels of DeltaF-specific serum IgG, IgG1 and IgG2a antibodies when compared with DeltaF alone, and displayed an increase in the frequency of gamma interferon-secreting cells and decreased interleukin (IL)-5 production by in vitro restimulated splenocytes, characteristic of a Th1 immune response. These results were observed in both C57BL/6 and BALB/c strains of mice. When evaluated in a BRSV challenge model, mice immunized with DeltaF/CpG/indol/PP developed significantly higher levels of BRSV-neutralizing serum antibodies than mice immunized with the DeltaF protein alone, and displayed significantly less pulmonary IL-4, IL-5, IL-13 and eotaxin and reduced eosinophilia after challenge. These results suggest that co-formulation of DeltaF with CpG ODN, host defence peptide and polyphosphazene may result in a safe and effective vaccine for the prevention of BRSV and may have implications for the development of novel human RSV vaccines.
Collapse
Affiliation(s)
- J Kovacs-Nolan
- Vaccine and Infectious Disease Organization, University of Saskatchewan, Saskatoon, SK S7N 5E3, Canada
| | - J W Mapletoft
- Vaccine and Infectious Disease Organization, University of Saskatchewan, Saskatoon, SK S7N 5E3, Canada
| | - Z Lawman
- Vaccine and Infectious Disease Organization, University of Saskatchewan, Saskatoon, SK S7N 5E3, Canada
| | - L A Babiuk
- University of Alberta, 3-7 University Hall, Edmonton, AB T6G 2J9, Canada
| | | |
Collapse
|
38
|
Olson MR, Varga SM. Fas ligand is required for the development of respiratory syncytial virus vaccine-enhanced disease. THE JOURNAL OF IMMUNOLOGY 2009; 182:3024-31. [PMID: 19234198 DOI: 10.4049/jimmunol.0803585] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Children immunized with a formalin-inactivated respiratory syncytial virus (RSV) vaccine experienced enhanced disease and exhibited pulmonary eosinophilia upon natural RSV infection. BALB/c mice immunized with either formalin-inactivated RSV or a recombinant vaccinia virus (vacv) expressing the RSV attachment (G) protein develop extensive pulmonary eosinophilia after RSV challenge that mimics the eosinophilic response observed in the children during the 1960s vaccine trials. Fas ligand (FasL) is a major immune effector molecule that can contribute to the clearance of respiratory viruses. However, the role of FasL in the development of RSV vaccine-enhanced disease has not been elucidated. RSV challenge of vacvG-immunized gld mice, that lack functional FasL, results in diminished systemic disease as well as pulmonary eosinophilia. The magnitude of the secondary RSV G-specific CD4 T cell response was diminished in gld mice as compared with wild-type controls. Furthermore, we show that CD4 T cells isolated after RSV challenge of vacvG-immunized gld mice exhibit enhanced expression of Annexin V and caspase 3/7 indicating that FasL is important for either the survival or the expansion of virus-specific secondary effector CD4 T cells. Taken together, these data identify a previously undefined role for FasL in the accumulation of secondary effector CD4 T cells and the development of RSV vaccine-enhanced disease.
Collapse
Affiliation(s)
- Matthew R Olson
- Department of Microbiology, University of Iowa, Iowa City, IA 52242, USA
| | | |
Collapse
|
39
|
Fu Y, He J, Zheng X, Wu Q, Zhang M, Wang X, Wang Y, Xie C, Tang Q, Wei W, Wang M, Song J, Qu J, Zhang Y, Wang X, Hong T. Intranasal immunization with a replication-deficient adenoviral vector expressing the fusion glycoprotein of respiratory syncytial virus elicits protective immunity in BALB/c mice. Biochem Biophys Res Commun 2009; 381:528-32. [DOI: 10.1016/j.bbrc.2009.02.075] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2009] [Accepted: 02/16/2009] [Indexed: 10/21/2022]
|
40
|
Reed JL, Brewah YA, Delaney T, Welliver T, Burwell T, Benjamin E, Kuta E, Kozhich A, McKinney L, Suzich J, Kiener PA, Avendano L, Velozo L, Humbles A, Welliver RC, Coyle AJ. Macrophage impairment underlies airway occlusion in primary respiratory syncytial virus bronchiolitis. J Infect Dis 2009; 198:1783-93. [PMID: 18980502 DOI: 10.1086/593173] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Although respiratory syncytial virus (RSV) infection is the most important cause of bronchiolitis in infants, the pathogenesis of RSV disease is poorly described. We studied histopathologic changes in a panel of lung tissue specimens obtained from infants with fatal cases of primary RSV infection. In these tissues, airway occlusion with accumulations of infected, apoptotic cellular debris and serum protein was consistently observed. Similar observations were found after RSV infection in New Zealand black (NZB) mice, which have constitutive deficiencies in macrophage function, but not in BALB/c mice. A deficiency in the number of alveolar macrophages in NZB mice appears to be central to enhanced disease, because depletion of alveolar macrophages in BALB/c mice before RSV exposure resulted in airway occlusion. In mice with insufficient numbers of macrophages, RSV infection yielded an increased viral load and enhanced expression of type I interferon-associated genes at the height of disease. Together, our data suggest that innate, rather than adaptive, immune responses are critical determinants of the severity of RSV bronchiolitis.
Collapse
Affiliation(s)
- Jennifer L Reed
- Respiratory, Inflammation, and Autoimmunity Group, MedImmune, Gaithersburg, Maryland, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Abstract
Respiratory syncytial virus (RSV) is the leading cause of severe respiratory disease in infants and is an important source of morbidity and mortality in the elderly and immunocompromised. This review will discuss the humoral and cellular adaptive immune responses to RSV infection and how these responses are shaped in the immature immune system of the infant and the aged environment of the elderly. Furthermore, we will provide an overview of our current understanding of the role the various arms of the adaptive immune response play in mediating the delicate balance between the successful elimination of the virus from the host and the induction of immunopathology. Efficacious immunization against RSV remains a high priority within the field and we will highlight recent advances made in vaccine design.
Collapse
Affiliation(s)
- Matthew R Olson
- Department of Microbiology, 51 Newton Road, 3−532 Bowen Science Building, University of Iowa, Iowa City, IA 52242, USA Tel.: +1 319 335 8433 Fax: +1 319 335 9006
| | - Steven M Varga
- Department of Microbiology, Interdisciplinary Graduate Program in Immunology, 51 Newton Road, 3−532 Bowen Science Building, University of Iowa, Iowa City, IA 52242, USA Tel.: +1 319 335 7784 Fax: +1 319 335 9006
| |
Collapse
|
42
|
The role of T cells in the enhancement of respiratory syncytial virus infection severity during adult reinfection of neonatally sensitized mice. J Virol 2008; 82:4115-24. [PMID: 18272579 DOI: 10.1128/jvi.02313-07] [Citation(s) in RCA: 97] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Respiratory syncytial virus (RSV) is the major cause of infantile bronchiolitis and hospitalization. Severe RSV disease is associated with the development of wheezing in later life. In a mouse model of the delayed effects of RSV, the age at primary infection determines responses to reinfection in adulthood. During primary RSV infection, neonatal BALB/c mice developed only mild disease and recruited CD8 cells that were defective in gamma interferon production. Secondary reinfection of neonatally primed mice caused enhanced inflammation and profuse lung T-cell recruitment. CD4 cell depletion during secondary RSV challenge attenuated disease (measured by weight loss); depletion of CD8 cells also markedly attenuated disease severity but enhanced lung eosinophilia, and depletion of both CD4 and CD8 cells together completely abrogated weight loss. Depletion of CD8 (but not CD4) cells during primary neonatal infection was protective against weight loss during adult challenge. Therefore, T cells, in particular CD8 T cells, play a central role in the outcome of neonatal infection by enhancing disease during secondary challenge. These findings demonstrate a crucial role for T cells in the regulation of immune responses after neonatal infection.
Collapse
|
43
|
van Drunen Littel-van den Hurk S, Mapletoft JW, Arsic N, Kovacs-Nolan J. Immunopathology of RSV infection: prospects for developing vaccines without this complication. Rev Med Virol 2007; 17:5-34. [PMID: 17004293 DOI: 10.1002/rmv.518] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Respiratory syncytial virus is the most important cause of lower respiratory tract infection in infants and young children. RSV clinical disease varies from rhinitis and otitis media to bronchiolitis and pneumonia. An increased incidence of asthma later in life has been associated with the more severe lower respiratory tract infections. Despite its importance as a pathogen, there is no licensed vaccine against RSV. This is due to a number of factors complicating the development of an effective and safe vaccine. The immunity to natural RSV infection is incomplete as re-infections occur in all age groups, which makes it challenging to design a protective vaccine. Second, the primary target population is the newborn infant, which has a relatively immature immune system and maternal antibodies that can interfere with vaccination. Finally, some vaccines have resulted in a predisposition for exacerbated pulmonary disease in infants, which was attributed to an imbalanced Th2-biased immune response, although the exact cause has not been elucidated. This makes it difficult to proceed with vaccine testing in infants. It is likely that an effective and safe vaccine needs to elicit a balanced immune response, including RSV-specific neutralising antibodies, CD8 T-cells, Th1/Th2 CD4 T-cells and preferably secretory IgA. Subunit vaccines formulated with appropriate adjuvants may be adequate for previously exposed individuals. However, intranasally delivered genetically engineered attenuated or vectored vaccines are currently most promising for newborns, as they are expected to induce a balanced immune response similar to that elicited to natural infection and not be subject to interference from maternal antibodies. Maternal vaccination may be the optimal strategy to protect the very young infants.
Collapse
MESH Headings
- Adult
- Animals
- Antibody Formation
- Child, Preschool
- Female
- Humans
- Immunity, Active
- Immunity, Cellular
- Immunity, Innate
- Immunity, Maternally-Acquired
- Infant
- Infant, Newborn
- Pregnancy
- Respiratory Syncytial Virus Infections/immunology
- Respiratory Syncytial Virus Infections/prevention & control
- Respiratory Syncytial Virus Vaccines/administration & dosage
- Respiratory Syncytial Virus Vaccines/adverse effects
- Respiratory Syncytial Virus Vaccines/immunology
- Respiratory Syncytial Virus, Human/immunology
- Vaccination/methods
- Vaccination/trends
- Vaccines, Subunit/administration & dosage
- Vaccines, Subunit/immunology
- Vaccines, Synthetic/administration & dosage
- Vaccines, Synthetic/immunology
Collapse
|
44
|
Kong X, Zhang W, Lockey RF, Auais A, Piedimonte G, Mohapatra SS. Respiratory syncytial virus infection in Fischer 344 rats is attenuated by short interfering RNA against the RSV-NS1 gene. GENETIC VACCINES AND THERAPY 2007; 5:4. [PMID: 17270047 PMCID: PMC1805440 DOI: 10.1186/1479-0556-5-4] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/27/2006] [Accepted: 02/01/2007] [Indexed: 11/21/2022]
Abstract
Background Respiratory syncytial virus (RSV) causes severe bronchiolitis and is a risk factor for asthma. Since there is no commercially available vaccine against RSV, a short interfering RNA against the RSV-NS1gene (siNS1) was developed and its potential for decreasing RSV infection and infection-associated inflammation in rats was tested. Methods Plasmids encoding siNS1 or an unrelated siRNA were complexed with a chitosan nanoparticle delivery agent and administered intranasally. Control animals received a plasmid for a non-specific siRNA. After expression of the plasmid in lung cells for 24 hours, the rats were intranasally infected with RSV. Results Prophylaxis with siNS1 significantly reduced lung RSV titers and airway hyperreactivity to methacholine challenge compared to the control group. Lung sections from siNS1-treated rats showed a sizable reduction in goblet cell hyperplasia and in lung infiltration by inflammatory cells, both characteristics of asthma. Also, bronchoalveolar lavage samples from siNS1-treated animals had fewer eosinophils. Treatment of rats with siNS1 prior to RSV exposure was effective in reducing virus titers in the lung and in preventing the inflammation and airway hyperresponsiveness associated with the infection that has been linked to development of asthma. Conclusion The use of siNS1 prophylaxis may be an effective method for preventing RSV bronchiolitis and potentially reducing the later development of asthma associated with severe respiratory infections.
Collapse
Affiliation(s)
- Xiaoyuan Kong
- University of South Florida College of Medicine, Florida, USA
| | - Weidong Zhang
- University of South Florida College of Medicine, Florida, USA
| | | | - Alexander Auais
- Batchelor Children's Institute, University of Miami, Miami, Florida, USA
| | | | | |
Collapse
|
45
|
Abstract
Originally considered as only a paediatric pathogen, respiratory syncytial virus (RSV) has recently been shown to be a significant cause of respiratory illness among elderly and high-risk adults. Approximately 170,000 hospitalizations and 10,000 deaths associated with RSV occur annually in people over the age of 65 years in the United States. Although rhinorrhoea and wheezing are common symptoms among adults, the clinical syndrome associated with RSV is not distinctive and thus laboratory methods are required for specific diagnosis. Presently, the combination of reverse transcription PCR and enzyme immunoassay serology offers the best sensitivity and specificity for diagnosis of RSV. Treatment options are limited at present, with inhaled ribavirin being the only licensed drug for use in hospitalized children. Vaccines against RSV remain an unachieved goal. Promising new agents that inhibit the virus-cell fusion, cell-cell fusion, or viral gene expression are currently in development.
Collapse
Affiliation(s)
- Yoshihiko Murata
- Infectious Diseases Unit, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester Genera Hospital, New York, USA
| | - Ann R Falsey
- Infectious Diseases Unit, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester Genera Hospital, New York, USA
| |
Collapse
|
46
|
Fan CF, Mei XG. Co-immunization of BALB/c mice with recombinant immunogens containing G protein fragment and chimeric CTL epitope of respiratory syncytial virus induces enhanced cellular immunity and high level of antibody response. Vaccine 2005; 23:4453-61. [PMID: 15935522 DOI: 10.1016/j.vaccine.2005.03.044] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2004] [Accepted: 03/29/2005] [Indexed: 12/01/2022]
Abstract
With the goal to develop effective immunogens against infection of respiratory syncytial virus (RSV), vectors co-expressing chimeric CTL epitope or G protein fragment of RSV with carrier protein DsbA (disulfide bond isomerase) were constructed. The capacity of the expressed recombinant immunogens to induce cellular and humoral immunities were evaluated. It was demonstrated that the presence of G protein fragment was able to enhance the CTL activities induced by the chimeric CTL epitope, though G protein fragment alone had no effect on induction of CTL response. In contrast, the level of antibody response to RSV and neutralization titer in co-immunization with G protein fragment plus chimeric CTL epitope was lower than that in immunization with G protein fragment alone. The challenge experiments indicated that co-immunization further reduced RSV titers both in lung tissue and nasal track, indicating the combination of humoral and cellular immunities is more effective. This data imply that the combination of the two protein immunogens would be a viable strategy for a RSV vaccine.
Collapse
Affiliation(s)
- Chang-Fa Fan
- Beijing Institute of Pharmacology and Toxicology, 27 Taiping Road, 100850 Beijing, PR China.
| | | |
Collapse
|
47
|
Openshaw PJM, Tregoning JS. Immune responses and disease enhancement during respiratory syncytial virus infection. Clin Microbiol Rev 2005; 18:541-55. [PMID: 16020689 PMCID: PMC1195968 DOI: 10.1128/cmr.18.3.541-555.2005] [Citation(s) in RCA: 217] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Respiratory syncytial virus (RSV) is one of the commonest and most troublesome viruses of infancy. It causes most cases of bronchiolitis, which is associated with wheezing in later childhood. In primary infection, the peak of disease typically coincides with the development of specific T- and B-cell responses, which seem, in large part, to be responsible for disease. Animal models clearly show that a range of immune responses can enhance disease severity, particularly after vaccination with formalin-inactivated RSV. Prior immune sensitization leads to exuberant chemokine production, an excessive cellular influx, and an overabundance of cytokines during RSV challenge. Under different circumstances, specific mediators and T-cell subsets and antibody-antigen immune complex deposition are incriminated as major factors in disease. Animal models of immune enhancement permit a deep understanding of the role of specific immune responses in RSV disease, assist in vaccine design, and indicate which immunomodulatory therapy might be beneficial to children with bronchiolitis.
Collapse
Affiliation(s)
- Peter J M Openshaw
- Department of Respiratory Medicine, National Heart and Lung and Wright Fleming Institutes, Faculty of Medicine, Imperial College London, Paddington, London W2 1PG, United Kingdom.
| | | |
Collapse
|
48
|
Chemokines and Chemokine Receptors in Pulmonary Disease. CURRENT TOPICS IN MEMBRANES 2005. [DOI: 10.1016/s1063-5823(04)55008-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
49
|
Heidema J, de Bree GJ, de Graaff PMA, van Maren WWC, Hoogerhout P, Out TA, Kimpen JLL, van Bleek GM. Human CD8(+) T cell responses against five newly identified respiratory syncytial virus-derived epitopes. J Gen Virol 2004; 85:2365-2374. [PMID: 15269378 DOI: 10.1099/vir.0.80131-0] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
CD8(+) T lymphocytes play a major role in the clearance of respiratory syncytial virus (RSV) infections. To be able to study the primary CTL response in RSV-infected children, epitopes presented by a set of commonly used HLA alleles (HLA-A1, -A3, -B44 and -B51) were searched for. Five epitopes were characterized derived from the matrix (M), non-structural (NS2) and second matrix (M2) proteins of RSV. All epitopes were shown to be processed and presented by RSV-infected antigen-presenting cells. HLA-A1 tetramers for one of these epitopes derived from the M protein were constructed and used to quantify and phenotype the memory CD8(+) T cell pool in a panel of healthy adult donors. In about 60 % of the donors, CD8(+) T cells specific for the M protein could be identified. These cells belonged to the memory T cell subset characterized by expression of CD27 and CD28, and down-regulation of CCR7 and CD45RA. The frequency of tetramer-positive cells varied between 0.4 and 3 per 10(4) CD8(+) T cells in PBMC of healthy asymptomatic adult donors.
Collapse
Affiliation(s)
- Jojanneke Heidema
- Division of Pediatrics, Wilhelmina Children's Hospital, University Medical Center, KE 04.133.1, PO Box 85500, 3508 AB Utrecht, The Netherlands
| | - Godelieve J de Bree
- Division of Pulmonology and the Department of Experimental Immunology, Academic Medical Center, Amsterdam, The Netherlands
| | - Patricia M A de Graaff
- Division of Pediatrics, Wilhelmina Children's Hospital, University Medical Center, KE 04.133.1, PO Box 85500, 3508 AB Utrecht, The Netherlands
| | - Wendy W C van Maren
- Division of Pediatrics, Wilhelmina Children's Hospital, University Medical Center, KE 04.133.1, PO Box 85500, 3508 AB Utrecht, The Netherlands
| | | | - Theo A Out
- Division of Clinical Immunology, Academic Medical Center, Amsterdam, The Netherlands
| | - Jan L L Kimpen
- Division of Pediatrics, Wilhelmina Children's Hospital, University Medical Center, KE 04.133.1, PO Box 85500, 3508 AB Utrecht, The Netherlands
| | - Grada M van Bleek
- Division of Pediatrics, Wilhelmina Children's Hospital, University Medical Center, KE 04.133.1, PO Box 85500, 3508 AB Utrecht, The Netherlands
| |
Collapse
|
50
|
Davies JK, Koh MBC, Lowdell MW. Antiviral immunity and T-regulatory cell function are retained after selective alloreactive T-cell depletion in both the HLA-identical and HLA-mismatched settings. Biol Blood Marrow Transplant 2004; 10:259-68. [PMID: 15077224 DOI: 10.1016/j.bbmt.2003.12.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Nonselective T-cell depletion reduces the incidence of severe graft-versus-host disease after allogeneic hematopoietic stem cell transplantation, but the cost is delayed and disordered antigen-specific immune reconstitution and increased infection. We use a method of selective depletion of alloreactive T cells expressing the activation marker CD69 after coculture with stimulator cells in a modified or standard mixed lymphocyte reaction. The technique has been shown to reduce alloreactivity while retaining third-party responses in vitro and, in a mismatched murine model, led to donor T-cell engraftment with a virtual absence of graft-versus-host disease and increased survival. We show in a human HLA-mismatched and unrelated HLA-identical setting that this technique retains >80% of specific cellular antiviral activity by cytomegalovirus-tetramer analysis and cytomegalovirus/Epstein-Barr virus peptide-stimulated interferon-gamma ELISpot assay. Furthermore, CD4(+) CD25(+) T-regulatory cells are not removed by this method of selective allodepletion and retain their function in suppressing allogeneic proliferative responses. Preservation of antiviral cytotoxic T lymphocytes in selectively allodepleted stem cell grafts would lead to improved antiviral immunity after transplantation. The retention of immunosuppressive CD4(+) CD25(+) T-regulatory cells could lead to more ordered immune reconstitution and further suppress alloreactive responses after transplantation.
Collapse
Affiliation(s)
- Jeffrey K Davies
- Department of Haematology, Royal Free & University College Medical School, London, United Kingdom, UK.
| | | | | |
Collapse
|