1
|
Rich HE, Bhutia S, Gonzales de Los Santos F, Entrup GP, Warheit-Niemi HI, Gurczynski SJ, Bame M, Douglas MT, Morris SB, Zemans RL, Lukacs NW, Moore BB. RSV enhances Staphylococcus aureus bacterial growth in the lung. Infect Immun 2024; 92:e0030424. [PMID: 39150268 PMCID: PMC11475690 DOI: 10.1128/iai.00304-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Accepted: 07/23/2024] [Indexed: 08/17/2024] Open
Abstract
Patients coinfected with respiratory syncytial virus (RSV) and bacteria have longer hospital stays, higher risk of intensive care unit admission, and worse outcomes. We describe a model of RSV line 19F/methicillin-resistant Staphylococcus aureus (MRSA) USA300 coinfection that does not impair viral clearance, but prior RSV infection enhances USA300 MRSA bacterial growth in the lung. The increased bacterial burden post-RSV correlates with reduced accumulation of neutrophils and impaired bacterial killing by alveolar macrophages. Surprisingly, reduced neutrophil accumulation is likely not explained by reductions in phagocyte-recruiting chemokines or alterations in proinflammatory cytokine production compared with mice infected with S. aureus alone. Neutrophils from RSV-infected mice retain their ability to migrate toward chemokine signals, and neutrophils from the RSV-infected lung are better able to phagocytize and kill S. aureus ex vivo on a per cell basis. In contrast, while alveolar macrophages could ingest USA300 post-RSV, intracellular bacterial killing was impaired. The RSV/S. aureus coinfected lung promotes a state of overactivation in neutrophils, demonstrated by increased production of reactive oxygen species (ROS) that can drive formation of neutrophil extracellular traps (NETs), resulting in cell death. Mice with RSV/S. aureus coinfection had increased extracellular DNA and protein in bronchoalveolar lavage fluid and histological evidence confirmed NETosis in vivo. Taken together, these data highlight that prior RSV infection can prime the overactivation of neutrophils leading to cell death that impairs neutrophil accumulation in the lung. Additionally, alveolar macrophage killing of bacteria is impaired post-RSV. Together, these defects enhance USA300 MRSA bacterial growth in the lung post-RSV.
Collapse
Affiliation(s)
- Helen E. Rich
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan, USA
| | - Simran Bhutia
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan, USA
| | | | - Gabrielle P. Entrup
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan, USA
| | - Helen I. Warheit-Niemi
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan, USA
| | - Stephen J. Gurczynski
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan, USA
| | - Monica Bame
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan, USA
| | - Michael T. Douglas
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Susan B. Morris
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, USA
| | - Rachel L. Zemans
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Nicholas W. Lukacs
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, USA
| | - Bethany B. Moore
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan, USA
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
2
|
Schaerlaekens S, Jacobs L, Stobbelaar K, Cos P, Delputte P. All Eyes on the Prefusion-Stabilized F Construct, but Are We Missing the Potential of Alternative Targets for Respiratory Syncytial Virus Vaccine Design? Vaccines (Basel) 2024; 12:97. [PMID: 38250910 PMCID: PMC10819635 DOI: 10.3390/vaccines12010097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 01/12/2024] [Accepted: 01/13/2024] [Indexed: 01/23/2024] Open
Abstract
Respiratory Syncytial Virus (RSV) poses a significant global health concern as a major cause of lower respiratory tract infections (LRTIs). Over the last few years, substantial efforts have been directed towards developing vaccines and therapeutics to combat RSV, leading to a diverse landscape of vaccine candidates. Notably, two vaccines targeting the elderly and the first maternal vaccine have recently been approved. The majority of the vaccines and vaccine candidates rely solely on a prefusion-stabilized conformation known for its highly neutralizing epitopes. Although, so far, this antigen design appears to be successful for the elderly, our current understanding remains incomplete, requiring further improvement and refinement in this field. Pediatric vaccines still have a long journey ahead, and we must ensure that vaccines currently entering the market do not lose efficacy due to the emergence of mutations in RSV's circulating strains. This review will provide an overview of the current status of vaccine designs and what to focus on in the future. Further research into antigen design is essential, including the exploration of the potential of alternative RSV proteins to address these challenges and pave the way for the development of novel and effective vaccines, especially in the pediatric population.
Collapse
Affiliation(s)
- Sofie Schaerlaekens
- Laboratory for Microbiology, Parasitology and Hygiene, University of Antwerp (UA), Universiteitsplein 1 S.7, 2610 Antwerp, Belgium; (S.S.); (L.J.); (K.S.); (P.C.)
| | - Lotte Jacobs
- Laboratory for Microbiology, Parasitology and Hygiene, University of Antwerp (UA), Universiteitsplein 1 S.7, 2610 Antwerp, Belgium; (S.S.); (L.J.); (K.S.); (P.C.)
| | - Kim Stobbelaar
- Laboratory for Microbiology, Parasitology and Hygiene, University of Antwerp (UA), Universiteitsplein 1 S.7, 2610 Antwerp, Belgium; (S.S.); (L.J.); (K.S.); (P.C.)
- Pediatrics Department, Antwerp University Hospital (UZA), Wilrijkstraat 10, 2650 Edegem, Belgium
| | - Paul Cos
- Laboratory for Microbiology, Parasitology and Hygiene, University of Antwerp (UA), Universiteitsplein 1 S.7, 2610 Antwerp, Belgium; (S.S.); (L.J.); (K.S.); (P.C.)
- Infla-Med Centre of Excellence, University of Antwerp (UA), Universiteitsplein 1 S.7, 2610 Antwerp, Belgium
| | - Peter Delputte
- Laboratory for Microbiology, Parasitology and Hygiene, University of Antwerp (UA), Universiteitsplein 1 S.7, 2610 Antwerp, Belgium; (S.S.); (L.J.); (K.S.); (P.C.)
- Infla-Med Centre of Excellence, University of Antwerp (UA), Universiteitsplein 1 S.7, 2610 Antwerp, Belgium
| |
Collapse
|
3
|
Cheng X, Zhao G, Dong A, He Z, Wang J, Jiang B, Wang B, Wang M, Huai X, Zhang S, Feng S, Qin H, Wang B. A First-in-Human Trial to Evaluate the Safety and Immunogenicity of a G Protein-Based Recombinant Respiratory Syncytial Virus Vaccine in Healthy Adults 18-45 Years of Age. Vaccines (Basel) 2023; 11:vaccines11050999. [PMID: 37243103 DOI: 10.3390/vaccines11050999] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 05/12/2023] [Accepted: 05/15/2023] [Indexed: 05/28/2023] Open
Abstract
BACKGROUND With the enormous morbidity and mortality caused by respiratory syncytial virus (RSV) infections among infants and the elderly, vaccines against RSV infections are in large market demand. METHODS We conducted a first-in-human (FIH), randomized, double-blind, placebo-controlled dose escalation study to evaluate the safety and immunogenicity response of the rRSV vaccine (BARS13) in healthy adults aged 18-45. A total of 60 eligible participants were randomly assigned to receive one of four dose levels or vaccination regimens of BARS13 or placebo at a 4:1 ratio. RESULTS The mean age was 27.40, and 23.3% (14/60) were men. No treatment-emergent adverse events (TEAEs) led to study withdrawal within 30 days after each vaccination. No serious adverse event (SAE) was reported. Most of the treatment-emergent adverse events (TEAEs) recorded were classified as mild. The high-dose repeat group had a serum-specific antibody GMC of 885.74 IU/mL (95% CI: 406.25-1931.17) 30 days after the first dose and 1482.12 IU/mL (706.56-3108.99) 30 days after the second dose, both higher than the GMC in the low-dose repeat group (885.74 IU/mL [406.25-1931.17] and 1187.10 IU/ mL [610.01-2310.13]). CONCLUSIONS BARS13 had a generally good safety and tolerability profile, and no significant difference in terms of adverse reaction severity or frequency was observed between different dose groups. The immune response in repeat-dose recipients shows more potential in further study and has guiding significance for the dose selection of subsequent studies.
Collapse
Affiliation(s)
- Xin Cheng
- Advaccine Biopharmaceuticals Suzhou Co., Ltd., Suzhou 215000, China
| | - Gan Zhao
- Advaccine Biopharmaceuticals Suzhou Co., Ltd., Suzhou 215000, China
| | - Aihua Dong
- Advaccine Biopharmaceuticals Suzhou Co., Ltd., Suzhou 215000, China
| | - Zhonghuai He
- Advaccine Biopharmaceuticals Suzhou Co., Ltd., Suzhou 215000, China
| | - Jiarong Wang
- Advaccine Biopharmaceuticals Suzhou Co., Ltd., Suzhou 215000, China
| | - Brian Jiang
- Advaccine Biopharmaceuticals Suzhou Co., Ltd., Suzhou 215000, China
| | - Bo Wang
- Advaccine Biopharmaceuticals Suzhou Co., Ltd., Suzhou 215000, China
| | - Miaomiao Wang
- Advaccine Biopharmaceuticals Suzhou Co., Ltd., Suzhou 215000, China
| | - Xuefen Huai
- Advaccine Biopharmaceuticals Suzhou Co., Ltd., Suzhou 215000, China
| | - Shijie Zhang
- Advaccine Biopharmaceuticals Suzhou Co., Ltd., Suzhou 215000, China
| | | | - Hong Qin
- Advaccine Biopharmaceuticals Suzhou Co., Ltd., Suzhou 215000, China
| | - Bin Wang
- Advaccine Biopharmaceuticals Suzhou Co., Ltd., Suzhou 215000, China
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Science, Fudan University, Shanghai 200000, China
| |
Collapse
|
4
|
Fisher DG, Gnazzo V, Holthausen DJ, López CB. Non-standard viral genome-derived RNA activates TLR3 and type I IFN signaling to induce cDC1-dependent CD8+ T-cell responses during vaccination in mice. Vaccine 2022; 40:7270-7279. [PMID: 36333225 DOI: 10.1016/j.vaccine.2022.10.052] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 10/13/2022] [Accepted: 10/20/2022] [Indexed: 11/13/2022]
Abstract
There is a critical need to develop vaccine adjuvants that induce robust immune responses able to protect against intracellular pathogens, including viruses. Previously, we described defective viral genome-derived oligonucleotides (DDOs) as novel adjuvants that strongly induce type 1 immune responses, including protective Th1 CD4+ T-cells and effector CD8+ T-cells in mice. Here, we unravel the early innate response required for this type 1 immunity induction. Upon DDO subcutaneous injection, type 1 conventional dendritic cells (cDC1s) accumulate rapidly in the draining lymph node in a Toll-like receptor 3 (TLR3)- and type I interferon (IFN)-dependent manner. cDC1 accumulation in the lymph node is required for antigen-specific CD8+ T-cell responses. Notably, in contrast to poly I:C, DDO administration resulted in type I IFN expression at the injection site, but not in the draining lymph node. Additionally, DDOs induced an inflammatory cytokine profile distinct from that induced by poly I:C. Therefore, DDOs represent a powerful new adjuvant to be used during vaccination against intracellular pathogens.
Collapse
Affiliation(s)
- Devin G Fisher
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Victoria Gnazzo
- Department of Molecular Microbiology and Center for Women Infectious Diseases, Washington University School of Medicine, Saint Louis, MO 63110, United States
| | - David J Holthausen
- Department of Molecular Microbiology and Center for Women Infectious Diseases, Washington University School of Medicine, Saint Louis, MO 63110, United States
| | - Carolina B López
- Department of Molecular Microbiology and Center for Women Infectious Diseases, Washington University School of Medicine, Saint Louis, MO 63110, United States.
| |
Collapse
|
5
|
Bigay J, Le Grand R, Martinon F, Maisonnasse P. Vaccine-associated enhanced disease in humans and animal models: Lessons and challenges for vaccine development. Front Microbiol 2022; 13:932408. [PMID: 36033843 PMCID: PMC9399815 DOI: 10.3389/fmicb.2022.932408] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 07/19/2022] [Indexed: 11/13/2022] Open
Abstract
The fight against infectious diseases calls for the development of safe and effective vaccines that generate long-lasting protective immunity. In a few situations, vaccine-mediated immune responses may have led to exacerbated pathology upon subsequent infection with the pathogen targeted by the vaccine. Such vaccine-associated enhanced disease (VAED) has been reported, or at least suspected, in animal models, and in a few instances in humans, for vaccine candidates against the respiratory syncytial virus (RSV), measles virus (MV), dengue virus (DENV), HIV-1, simian immunodeficiency virus (SIV), feline immunodeficiency virus (FIV), severe acute respiratory syndrome coronavirus 1 (SARS-CoV-1), and the Middle East respiratory syndrome coronavirus (MERS-CoV). Although alleviated by clinical and epidemiological evidence, a number of concerns were also initially raised concerning the short- and long-term safety of vaccines against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which is causing the ongoing COVID-19 pandemic. Although the mechanisms leading to this phenomenon are not yet completely understood, the individual and/or collective role of antibody-dependent enhancement (ADE), complement-dependent enhancement, and cell-dependent enhancement have been highlighted. Here, we review mechanisms that may be associated with the risk of VAED, which are important to take into consideration, both in the assessment of vaccine safety and in finding ways to define models and immunization strategies that can alleviate such concerns.
Collapse
Affiliation(s)
| | | | - Frédéric Martinon
- Immunology of Viral Infections and Autoimmune Diseases (IMVA), IDMIT Department, Institut de Biologie François-Jacob (IBJF), University Paris-Sud-INSERM U1184, CEA, Fontenay-Aux-Roses, France
| | | |
Collapse
|
6
|
Manti S, Piedimonte G. An overview on the RSV-mediated mechanisms in the onset of non-allergic asthma. Front Pediatr 2022; 10:998296. [PMID: 36204661 PMCID: PMC9530042 DOI: 10.3389/fped.2022.998296] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 08/19/2022] [Indexed: 12/13/2022] Open
Abstract
Respiratory syncytial virus (RSV) infection is recognized as an important risk factor for wheezing and asthma, since it commonly affects babies during lung development. While the role of RSV in the onset of atopic asthma is widely recognized, its impact on the onset of non-atopic asthma, mediated via other and independent causal pathways, has long been also suspected, but the association is less clear. Following RSV infection, the release of local pro-inflammatory molecules, the dysfunction of neural pathways, and the compromised epithelial integrity can become chronic and influence airway development, leading to bronchial hyperreactivity and asthma, regardless of atopic status. After a brief review of the RSV structure and its interaction with the immune system and neuronal pathways, this review summarizes the current evidence about the RSV-mediated pathogenic pathways in predisposing and inducing airway dysfunction and non-allergic asthma development.
Collapse
Affiliation(s)
- Sara Manti
- Pediatric Pulmonology Unit, Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy.,Pediatric Unit, Department of Human Pathology of Adult and Childhood Gaetano Barresi, University of Messina, Messina, Italy
| | - Giovanni Piedimonte
- Department of Pediatrics, Biochemistry and Molecular Biology, Tulane University, New Orleans, LA, United States
| |
Collapse
|
7
|
Bergeron HC, Tripp RA. Immunopathology of RSV: An Updated Review. Viruses 2021; 13:2478. [PMID: 34960746 PMCID: PMC8703574 DOI: 10.3390/v13122478] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/06/2021] [Accepted: 12/08/2021] [Indexed: 12/14/2022] Open
Abstract
RSV is a leading cause of respiratory tract disease in infants and the elderly. RSV has limited therapeutic interventions and no FDA-approved vaccine. Gaps in our understanding of virus-host interactions and immunity contribute to the lack of biological countermeasures. This review updates the current understanding of RSV immunity and immunopathology with a focus on interferon responses, animal modeling, and correlates of protection.
Collapse
Affiliation(s)
| | - Ralph A. Tripp
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA;
| |
Collapse
|
8
|
Munoz FM, Cramer JP, Dekker CL, Dudley MZ, Graham BS, Gurwith M, Law B, Perlman S, Polack FP, Spergel JM, Van Braeckel E, Ward BJ, Didierlaurent AM, Lambert PH. Vaccine-associated enhanced disease: Case definition and guidelines for data collection, analysis, and presentation of immunization safety data. Vaccine 2021; 39:3053-3066. [PMID: 33637387 PMCID: PMC7901381 DOI: 10.1016/j.vaccine.2021.01.055] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 01/18/2021] [Accepted: 01/20/2021] [Indexed: 12/25/2022]
Abstract
This is a Brighton Collaboration Case Definition of the term "Vaccine Associated Enhanced Disease" to be utilized in the evaluation of adverse events following immunization. The Case Definition was developed by a group of experts convened by the Coalition for Epidemic Preparedness Innovations (CEPI) in the context of active development of vaccines for SARS-CoV-2 vaccines and other emerging pathogens. The case definition format of the Brighton Collaboration was followed to develop a consensus definition and defined levels of certainty, after an exhaustive review of the literature and expert consultation. The document underwent peer review by the Brighton Collaboration Network and by selected Expert Reviewers prior to submission.
Collapse
Affiliation(s)
- Flor M Munoz
- Departments of Pediatrics, Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA.
| | - Jakob P Cramer
- Coalition for Epidemic Preparedness Innovations, CEPI, London, UK
| | - Cornelia L Dekker
- Department of Pediatrics, Stanford University School of Medicine, CA, USA
| | - Matthew Z Dudley
- Institute for Vaccine Safety, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Barney S Graham
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, USA
| | - Marc Gurwith
- Safety Platform for Emergency Vaccines, Los Altos Hills, CA, USA
| | - Barbara Law
- Safety Platform for Emergency Vaccines, Manta, Ecuador
| | - Stanley Perlman
- Department of Microbiology and Immunology, Department of Pediatrics, University of Iowa, USA
| | | | - Jonathan M Spergel
- Division of Allergy and Immunology, Children's Hospital of Philadelphia, Department of Pediatrics, Perelman School of Medicine at University of Pennsylvania, PA, USA
| | - Eva Van Braeckel
- Department of Respiratory Medicine, Ghent University Hospital, and Department of Internal Medicine and Paediatrics, Ghent University, Ghent, Belgium
| | - Brian J Ward
- Research Institute of the McGill University Health Center, Montreal, Quebec, Canada
| | | | | |
Collapse
|
9
|
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
|
10
|
T-Cells and Interferon Gamma Are Necessary for Survival Following Crimean-Congo Hemorrhagic Fever Virus Infection in Mice. Microorganisms 2021; 9:microorganisms9020279. [PMID: 33572859 PMCID: PMC7912317 DOI: 10.3390/microorganisms9020279] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/25/2021] [Accepted: 01/26/2021] [Indexed: 12/11/2022] Open
Abstract
Crimean-Congo hemorrhagic fever (CCHF) is a severe tick-borne febrile illness with wide geographic distribution. In humans, the disease follows infection by the Crimean-Congo hemorrhagic fever virus (CCHFV) and begins as flu-like symptoms that can rapidly progress to hemorrhaging and death. Case fatality rates can be as high as 30%. An important gap in our understanding of CCHF are the host immune responses necessary to control the infection. A better understanding of these responses is needed to direct therapeutic strategies to limit the often-severe morbidity and mortality seen in humans. In this report, we have utilized a mouse model in which mice develop severe disease but ultimately recover. T-cells were robustly activated, differentiated to produce antiviral cytokines, and were critical for survival following CCHFV infection. We further identified a key role for interferon gamma (IFNγ) in survival following CCHFV infection. These results significantly improve our understanding of the host adaptive immune response to severe CCHFV infection.
Collapse
|
11
|
Jang MJ, Kim YJ, Hong S, Na J, Hwang JH, Shin SM, Ahn YM. Positive association of breastfeeding on respiratory syncytial virus infection in hospitalized infants: a multicenter retrospective study. Clin Exp Pediatr 2020; 63:135-140. [PMID: 32024328 PMCID: PMC7170789 DOI: 10.3345/kjp.2019.00402] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 11/11/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Breastfeeding reportedly reduces the overall frequency of infections. Respiratory syncytial virus (RSV), the most common respiratory pathogen in infants, involves recurrent wheezing and has a pathogenic mechanism related to airway structural damage. PURPOSE This study aimed to investigate whether breastfeeding has a beneficial effect against RSV-induced respiratory infection compared to formula feeding among infants in Korea. METHODS We retrospectively reviewed the medical records of infants under 1 year of age who were admitted with RSV infection between January 2016 and February 2018 at the department of pediatrics of 4 hospitals. We investigated the differences in clinical parameters such as cyanosis, chest retraction, combined infection, fever duration, oxygen use, oxygen therapy duration, intensive care unit (ICU) admission, and corticosteroid treatment of exclusive breast milk feeding (BMF), artificial milk formula fed (AMF), and mixed feeding (MF) groups. RESULTS Among the 411 infants included in our study, 94, 161, and 156 were included in the BMF, MF, and AMF groups, respectively. The rates of oxygen therapy were significantly different among the BMF (4.3%), MF (8.1%), and AMF (13.5 %) groups (P=0.042). The odds ratios (ORs) for oxygen therapy was significantly higher in the AMF group than in the BMF group (adjusted OR, 3.807; 95% confidence interval, 1.22-11.90; P=0.021). The ICU admission rate of the BMF group (1.1%) was lower than that of the MF (3.5%) and AMF (4.5%) groups; however, the dissimilarity was not statistically significant (P=0.338). CONCLUSION The severity of RSV infection requiring oxygen therapy was lower in the BMF than the AMF group. This protective role of human milk on RSV infection might decrease the need for oxygen therapy suggesting less airway damage.
Collapse
Affiliation(s)
- Min Jeong Jang
- Department of Pediatrics, Nowon Eulji Medical Center, Eulji University, Seoul, Korea
| | - Yong Joo Kim
- Department of Pediatrics, Hanyang University Seoul Hospital, Seoul, Korea
| | - Shinhye Hong
- Department of Pediatrics, Hanyang University Seoul Hospital, Seoul, Korea
| | - Jaeyoon Na
- Department of Pediatrics, Hanyang University Seoul Hospital, Seoul, Korea
| | - Jong Hee Hwang
- Department of Pediatrics, Inje University Ilsan Paik Hospital, Goyang, Korea
| | - Son Moon Shin
- Department of Pediatrics, Inje University Busan Paik Hospital, Busan, Korea
| | - Young Min Ahn
- Department of Pediatrics, Nowon Eulji Medical Center, Eulji University, Seoul, Korea
| |
Collapse
|
12
|
Roumanes D, Falsey AR, Quataert S, Secor-Socha S, Lee FEH, Yang H, Bandyopadhyay S, Holden-Wiltse J, Topham DJ, Walsh EE. T-Cell Responses in Adults During Natural Respiratory Syncytial Virus Infection. J Infect Dis 2019; 218:418-428. [PMID: 29920599 DOI: 10.1093/infdis/jiy016] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 06/11/2018] [Indexed: 12/17/2022] Open
Abstract
Background The pathogenesis of respiratory syncytial virus (RSV) in older adults may be due to age-related T-cell immunosenescence. Thus, we evaluated CD4 and CD8 T-cell responses during RSV infection in adults across the age spectrum. Methods Peripheral blood mononuclear cells collected during RSV infection in adults, age 26-96 years, were stimulated with live RSV and peptide pools representing F, M, NP, and G proteins and analyzed by flow cytometry. Results There were no significant age-related differences in frequency of CD4+ T cells synthesizing interferon (IFN)γ, interleukin (IL)2, IL4, IL10, or tumor necrosis factor (TNF)α or in CD8+IFNγ+ T cells. IL4+CD4+ T-cell numbers were low, as were IL13 and IL17 responses. However, in univariate analysis, CD4 T-cell IFNγ, IL2, IL4, IL10, and TNFα responses and CD8+IFNγ+ T cells were significantly increased with more severe illness requiring hospitalization. In multivariate analysis, viral load was also associated with increased T-cell responses. Conclusions We found no evidence of diminished RSV-specific CD4 or CD8 T-cell responses in adults infected with RSV. However, adults with severe disease seemed to have more robust CD4 and CD8 T-cell responses during infection, suggesting that disease severity may have a greater association with T-cell responses than age.
Collapse
Affiliation(s)
- D Roumanes
- Department of Microbiology and Immunology, University of Rochester, New York
| | - A R Falsey
- Department of Medicine, University of Rochester, New York.,Department of Medicine, Rochester General Hospital, New York
| | - S Quataert
- Department of Microbiology and Immunology, University of Rochester, New York
| | - S Secor-Socha
- Department of Microbiology and Immunology, University of Rochester, New York
| | - F E-H Lee
- Department of Medicine, University of Rochester, New York
| | - H Yang
- Computational Biology and Biostatistics, University of Rochester, New York
| | - S Bandyopadhyay
- Computational Biology and Biostatistics, University of Rochester, New York
| | - J Holden-Wiltse
- Computational Biology and Biostatistics, University of Rochester, New York
| | - D J Topham
- Department of Microbiology and Immunology, University of Rochester, New York
| | - E E Walsh
- Department of Medicine, University of Rochester, New York.,Department of Medicine, Rochester General Hospital, New York
| |
Collapse
|
13
|
Kwon YM, Hwang HS, Lee YT, Kim KH, Lee Y, Kim MC, Lee YN, Quan FS, Moore ML, Kang SM. Respiratory Syncytial Virus Fusion Protein-encoding DNA Vaccine Is Less Effective in Conferring Protection against Inflammatory Disease than a Virus-like Particle Platform. Immune Netw 2019; 19:e18. [PMID: 31281715 PMCID: PMC6597443 DOI: 10.4110/in.2019.19.e18] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 03/12/2019] [Accepted: 03/13/2019] [Indexed: 12/17/2022] Open
Abstract
Formalin-inactivated respiratory syncytial virus (RSV) vaccination causes vaccine-enhanced disease (VED) after RSV infection. It is considered that vaccine platforms enabling endogenous synthesis of RSV immunogens would induce favorable immune responses than non-replicating subunit vaccines in avoiding VED. Here, we investigated the immunogenicity, protection, and disease in mice after vaccination with RSV fusion protein (F) encoding plasmid DNA (F-DNA) or virus-like particles presenting RSV F (F-VLP). F-DNA vaccination induced CD8 T cells and RSV neutralizing Abs, whereas F-VLP elicited higher levels of IgG2a isotype and neutralizing Abs, and germinal center B cells, contributing to protection by controlling lung viral loads after RSV challenge. However, mice that were immunized with F-DNA displayed weight loss and pulmonary histopathology, and induced F specific CD8 T cell responses and recruitment of monocytes and plasmacytoid dendritic cells into the lungs. These innate immune parameters, RSV disease, and pulmonary histopathology were lower in mice that were immunized with F-VLP after challenge. This study provides important insight into developing effective and safe RSV vaccines.
Collapse
Affiliation(s)
- Young-Man Kwon
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| | - Hye Suk Hwang
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA.,Department of Microbiology, Chonnam National University Medical School, Hwasun 58128, Korea
| | - Young-Tae Lee
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA.,Green Cross Cell Corp., Yongin 16924, Korea
| | - Ki-Hye Kim
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| | - Youri Lee
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| | - Min-Chul Kim
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA.,Komipharm Co., Ltd., Siheung 15094, Korea
| | - Yu-Na Lee
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA.,Animal and Plant Quarantine Agency, Gimcheon 39660, Korea
| | - Fu-Shi Quan
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA.,Department of Medical Zoology, Kyung Hee University School of Medicine, Seoul 02447, Korea
| | | | - Sang-Moo Kang
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| |
Collapse
|
14
|
Yamaji Y, Sawada A, Yasui Y, Ito T, Nakayama T. Simultaneous Administration of Recombinant Measles Viruses Expressing Respiratory Syncytial Virus Fusion (F) and Nucleo (N) Proteins Induced Humoral and Cellular Immune Responses in Cotton Rats. Vaccines (Basel) 2019; 7:vaccines7010027. [PMID: 30836661 PMCID: PMC6466305 DOI: 10.3390/vaccines7010027] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 02/24/2019] [Accepted: 02/27/2019] [Indexed: 11/16/2022] Open
Abstract
We previously reported that recombinant measles virus expressing the respiratory syncytial virus (RSV) fusion protein (F), MVAIK/RSV/F, induced neutralizing antibodies against RSV, and those expressing RSV-NP (MVAIK/RSV/NP) and M2-1 (MVAIK/RSV/M2-1) induced RSV-specific CD8+/IFN-γ+ cells, but not neutralizing antibodies. In the present study, MVAIK/RSV/F and MVAIK/RSV/NP were simultaneously administered to cotton rats and immune responses and protective effects were compared with MVAIK/RSV/F alone. Sufficient neutralizing antibodies against RSV and RSV-specific CD8+/IFN-γ+ cells were observed after re-immunization with simultaneous administration. After the RSV challenge, CD8+/IFN-γ+ increased in spleen cells obtained from the simultaneous immunization group in response to F and NP peptides. Higher numbers of CD8+/IFN-γ+ and CD4+/IFN-γ+ cells were detected in lung tissues from the simultaneous immunization group after the RSV challenge. No detectable RSV was recovered from lung homogenates in the immunized groups. Mild inflammatory reactions with the thickening of broncho-epithelial cells and the infiltration of inflammatory cells were observed in lung tissues obtained from cotton rats immunized with MVAIK/RSV/F alone after the RSV challenge. No inflammatory responses were observed after the RSV challenge in the simultaneous immunization groups. The present results indicate that combined administration with MVAIK/RSV/F and MVAIK/RSV/NP induces humoral and cellular immune responses and shows effective protection against RSV, suggesting the importance of cellular immunity.
Collapse
Affiliation(s)
- Yoshiaki Yamaji
- Laboratory of Viral Infection II, Kitasato Institute for Life Sciences, Kitasato University, Tokyo 108-8641, Japan.
| | - Akihito Sawada
- Laboratory of Viral Infection II, Kitasato Institute for Life Sciences, Kitasato University, Tokyo 108-8641, Japan.
| | - Yosuke Yasui
- Health Center, Keio University, Kanagawa 223-8521, Japan.
| | - Takashi Ito
- Laboratory of Viral Infection II, Kitasato Institute for Life Sciences, Kitasato University, Tokyo 108-8641, Japan.
| | - Tetsuo Nakayama
- Laboratory of Viral Infection II, Kitasato Institute for Life Sciences, Kitasato University, Tokyo 108-8641, Japan.
| |
Collapse
|
15
|
Han M, Rajput C, Ishikawa T, Jarman CR, Lee J, Hershenson MB. Small Animal Models of Respiratory Viral Infection Related to Asthma. Viruses 2018; 10:E682. [PMID: 30513770 PMCID: PMC6316391 DOI: 10.3390/v10120682] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 11/21/2018] [Accepted: 11/29/2018] [Indexed: 12/20/2022] Open
Abstract
Respiratory viral infections are strongly associated with asthma exacerbations. Rhinovirus is most frequently-detected pathogen; followed by respiratory syncytial virus; metapneumovirus; parainfluenza virus; enterovirus and coronavirus. In addition; viral infection; in combination with genetics; allergen exposure; microbiome and other pathogens; may play a role in asthma development. In particular; asthma development has been linked to wheezing-associated respiratory viral infections in early life. To understand underlying mechanisms of viral-induced airways disease; investigators have studied respiratory viral infections in small animals. This report reviews animal models of human respiratory viral infection employing mice; rats; guinea pigs; hamsters and ferrets. Investigators have modeled asthma exacerbations by infecting mice with allergic airways disease. Asthma development has been modeled by administration of virus to immature animals. Small animal models of respiratory viral infection will identify cell and molecular targets for the treatment of asthma.
Collapse
Affiliation(s)
- Mingyuan Han
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
| | - Charu Rajput
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
| | - Tomoko Ishikawa
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
| | - Caitlin R Jarman
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
| | - Julie Lee
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
| | - Marc B Hershenson
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
| |
Collapse
|
16
|
Eichinger KM, Kosanovich JL, Empey KM. Localization of the T-cell response to RSV infection is altered in infant mice. Pediatr Pulmonol 2018; 53:145-153. [PMID: 29115050 PMCID: PMC5775046 DOI: 10.1002/ppul.23911] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 10/22/2017] [Indexed: 12/23/2022]
Abstract
OBJECTIVES Respiratory syncytial virus (RSV) is a leading cause of lower respiratory tract infections worldwide, causing disproportionate morbidity and mortality in infants and children. Infants with stronger Th1 responses have less severe disease, yet little is known about the infant T-cell response within the air space. Thus, we tested the hypothesis that RSV infected infant mice would have quantitative and qualitative deficiencies in CD4+ and CD8+ T-cell populations isolated from the bronchoalveolar lavage when compared to adults and that local delivery of IFN-γ would increase airway CD4+ Tbet+ and CD8+ Tbet+ T-cell responses. METHODS We compared the localization of T-cell responses in RSV-infected infant and adult mice and investigated the effects of local IFN-γ administration on infant cellular immunity. RESULTS Adult CD8+ CD44HI and CD4+ CD44HI Tbet+ T-cells accumulated in the alveolar space whereas CD4+ CD44HI Tbet+ T-cells were evenly distributed between the infant lung tissue and airway and infant lungs contained higher frequencies of CD8+ T-cells. Delivery of IFN-γ to the infant airway failed to increase the accumulation of T-cells in the airspace and unexpectedly reduced CD4+ CD44HI Tbet+ T-cells. However, intranasal IFN-γ increased RSV F protein-specific CD8+ T-cells in the alveolar space. CONCLUSION Together, these data suggest that quantitative and qualitative defects exist in the infant T-cell response to RSV but early, local IFN-γ exposure can increase the CD8+ RSV-specific T-cell response.
Collapse
Affiliation(s)
- Katherine M Eichinger
- Department of Pharmacy and Therapeutics, University of Pittsburgh School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania.,Center for Clinical Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Jessica L Kosanovich
- Department of Pharmacy and Therapeutics, University of Pittsburgh School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Kerry M Empey
- Department of Pharmacy and Therapeutics, University of Pittsburgh School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania.,Center for Clinical Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania.,Department of Immunology, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania.,Clinical and Translational Science Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
17
|
Virus-like particle vaccine primes immune responses preventing inactivated-virus vaccine-enhanced disease against respiratory syncytial virus. Virology 2017; 511:142-151. [DOI: 10.1016/j.virol.2017.08.022] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 08/08/2017] [Accepted: 08/15/2017] [Indexed: 12/20/2022]
|
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
|
Development of an intradermal DNA vaccine delivery strategy to achieve single-dose immunity against respiratory syncytial virus. Vaccine 2017; 35:2840-2847. [PMID: 28413132 DOI: 10.1016/j.vaccine.2017.04.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 03/22/2017] [Accepted: 04/04/2017] [Indexed: 12/22/2022]
Abstract
Respiratory syncytial virus (RSV) is a massive medical burden in infants, children and the elderly worldwide, and an effective, safe RSV vaccine remains an unmet need. Here we assess a novel vaccination strategy based on the intradermal delivery of a SynCon® DNA-based vaccine encoding engineered RSV-F antigen using a surface electroporation device (SEP) to target epidermal cells, in clinically relevant experimental models. We demonstrate the ability of this strategy to elicit robust immune responses. Importantly we demonstrate complete resistance to pulmonary infection at a single low dose of vaccine in the cotton rat RSV/A challenge model. In contrast to the formalin-inactivated RSV (FI-RSV) vaccine, there was no enhanced lung inflammation upon virus challenge after DNA vaccination. In summary the data presented outline the pre-clinical development of a highly efficacious, tolerable and safe non-replicating vaccine delivery strategy.
Collapse
|
20
|
Wu J, Hu H, Xu L, Qi F, Bai S, Cui Y, Chai R, Wang D, Liu B. Natural helper cells are associated with the exacerbated airway inflammation seen during RSV reinfection of neonatally primed mice. Int Immunopharmacol 2017; 45:156-162. [PMID: 28219838 DOI: 10.1016/j.intimp.2017.02.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2016] [Revised: 02/10/2017] [Accepted: 02/11/2017] [Indexed: 11/26/2022]
Abstract
Infection with respiratory syncytial virus (RSV) in neonatal mice causes more aggressive airway disease when the mice are reinfected with the same virus as adults. However, the underlying mechanisms responsible for this phenomenon are not entirely defined. Natural helper (NH) cells are considered a key factor for virus-induced or exacerbated airway inflammation and airway hyper-responsiveness by producing type 2 cytokines. To confirm whether NH cells are involved in the aggravated lung pathology seen during reinfection, BALB/c mice were initially infected as neonates and reinfected in adulthood. We observed that neonatal RSV infection resulted in an enhanced infiltration of eosinophils and neutrophils in the lungs, in parallel with a significant increase in the levels of IL-5 and IL-13 in bronchoalveolar lavage fluids on day 2 after reinfection. It seems likely that pulmonary NH cells may play a role in the occurrence, since mice first infected at 1wk of age developed an additional increase in the number of NH cells as well as IL-5- and IL-13-producing NH cells in the lungs than those first infected as young adults. In fact, an elevated expression of mRNAs for IL-5 and IL-13 in pulmonary NH cells was detected in mice first infected as neonates. Furthermore, adoptive transfer of NH cells into neonatal mice was able to boost eosinophilic infiltration as well as the production of type 2 cytokines in the lungs after reinfection at adulthood. In contrast, the expression of mRNA for the type 1 cytokine IFN-γ was down-regulated markedly by adoptive transfer of NH cells. Thus, these results suggest that Th2-type NH cells may play a role in the exacerbated airway inflammation seen during RSV reinfection of neonatally primed mice.
Collapse
Affiliation(s)
- Jianqi Wu
- Department of Neurosurgery, the First Affiliated Hospital of China Medical University, Shenyang, China
| | - Haiyan Hu
- Department of Immunology, School of Basic Medical Science, China Medical University, Shenyang, China
| | - Lei Xu
- Department of Immunology, School of Basic Medical Science, China Medical University, Shenyang, China
| | - Feifei Qi
- Department of Immunology, School of Basic Medical Science, China Medical University, Shenyang, China
| | - Song Bai
- Department of Immunology, School of Basic Medical Science, China Medical University, Shenyang, China
| | - Yulin Cui
- Department of Immunology, School of Basic Medical Science, China Medical University, Shenyang, China
| | - Ruonan Chai
- The PLA Center of Respiratory and Allergic Disease Diagnosing Management, the General Hospital of Shenyang Military Command, Shenyang, China
| | - Dandan Wang
- Department of Immunology, School of Basic Medical Science, China Medical University, Shenyang, China
| | - Beixing Liu
- Department of Immunology, School of Basic Medical Science, China Medical University, Shenyang, China.
| |
Collapse
|
21
|
Ramakrishnan B, Viswanathan K, Tharakaraman K, Dančík V, Raman R, Babcock GJ, Shriver Z, Sasisekharan R. A Structural and Mathematical Modeling Analysis of the Likelihood of Antibody-Dependent Enhancement in Influenza. Trends Microbiol 2016; 24:933-943. [PMID: 27751627 DOI: 10.1016/j.tim.2016.09.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 08/15/2016] [Accepted: 09/12/2016] [Indexed: 12/26/2022]
Abstract
Broadly neutralizing monoclonal antibodies (bNAbs) for viral infections, such as HIV, respiratory syncytial virus (RSV), and influenza, are increasingly entering clinical development. For influenza, most neutralizing antibodies target influenza virus hemagglutinin. These bNAbs represent an emerging, promising modality for treatment and prophylaxis of influenza due to their multiple mechanisms of antiviral action and generally safe profile. Preclinical work in other viral diseases, such as dengue, has demonstrated the potential for antibody-based therapies to enhance viral uptake, leading to enhanced viremia and worsening of disease. This phenomenon is referred to as antibody-dependent enhancement (ADE). In the context of influenza, ADE has been used to explain several preclinical and clinical phenomena. Using structural and viral kinetics modeling, we assess the role of ADE in the treatment of influenza with a bNAb.
Collapse
Affiliation(s)
| | | | - Kannan Tharakaraman
- Department of Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 01890, USA
| | - Vlado Dančík
- Center for the Science of Therapeutics, Broad Institute, Cambridge, MA 02142, USA
| | - Rahul Raman
- Department of Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 01890, USA
| | - Gregory J Babcock
- Visterra, Inc. One Kendall Square, Suite B3301, Cambridge, MA 02139, USA
| | - Zachary Shriver
- Visterra, Inc. One Kendall Square, Suite B3301, Cambridge, MA 02139, USA
| | - Ram Sasisekharan
- Department of Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 01890, USA.
| |
Collapse
|
22
|
Zhang Y, Qiao L, Hu X, Zhao K, Zhang Y, Chai F, Pan Z. Baculovirus vectors expressing F proteins in combination with virus-induced signaling adaptor (VISA) molecules confer protection against respiratory syncytial virus infection. Vaccine 2015; 34:252-260. [PMID: 26643933 DOI: 10.1016/j.vaccine.2015.11.027] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Revised: 11/04/2015] [Accepted: 11/12/2015] [Indexed: 11/16/2022]
Abstract
Baculovirus has been exploited for use as a novel vaccine vector. To investigate the feasibility and efficacy of recombinant baculoviruses (rBVs) expressing respiratory syncytial virus (RSV) fusion (F) proteins, four constructs (Bac-tF/64, Bac-CF, Bac-CF/tF64 and Bac-CF/tF64-VISA) were generated. Bac-tF64 displays the F ectodomain (tF) on the envelope of rBVs, whereas Bac-CF expresses full-length F protein in transduced mammalian cells. Bac-CF/tF64 not only displays tF on the envelope but also expresses F in cells. Bac-CF/tF64-VISA comprises Bac-CF/tF64 harboring the virus-induced signaling adaptor (VISA) gene. After administration to BALB/c mice, all four vectors elicited RSV neutralizing antibody (Ab), systemic Ab (IgG, IgG1, and IgG2a), and cytokine responses. Compared with Bac-tF64, mice inoculated with Bac-CF and Bac-CF/tF64 exhibited an increased mixed Th1/Th2 cytokine response, increased ratios of IgG2a/IgG1 antibody responses, and reduced immunopathology upon RSV challenge. Intriguingly, co-expression of VISA reduced Th2 cytokine (IL-4, IL-5, and IL-10) production induced by Bac-CF/tF64, thus relieving lung pathology upon a subsequent RSV challenge. Our results indicated that the Bac-CF/tF64 vector incorporated with the VISA molecule may provide an effective vaccine strategy for protection against RSV.
Collapse
Affiliation(s)
- Yuan Zhang
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Lei Qiao
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Xiao Hu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Kang Zhao
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Yanwen Zhang
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Feng Chai
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Zishu Pan
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China.
| |
Collapse
|
23
|
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
|
24
|
Pierantoni A, Esposito ML, Ammendola V, Napolitano F, Grazioli F, Abbate A, del Sorbo M, Siani L, D’Alise AM, Taglioni A, Perretta G, Siccardi A, Soprana E, Panigada M, Thom M, Scarselli E, Folgori A, Colloca S, Taylor G, Cortese R, Nicosia A, Capone S, Vitelli A. Mucosal delivery of a vectored RSV vaccine is safe and elicits protective immunity in rodents and nonhuman primates. Mol Ther Methods Clin Dev 2015; 2:15018. [PMID: 26015988 PMCID: PMC4441047 DOI: 10.1038/mtm.2015.18] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Accepted: 03/20/2015] [Indexed: 01/27/2023]
Abstract
Respiratory Syncytial Virus (RSV) is a leading cause of severe respiratory disease in infants and the elderly. No vaccine is presently available to address this major unmet medical need. We generated a new genetic vaccine based on chimpanzee Adenovirus (PanAd3-RSV) and Modified Vaccinia Ankara RSV (MVA-RSV) encoding the F, N, and M2-1 proteins of RSV, for the induction of neutralizing antibodies and broad cellular immunity. Because RSV infection is restricted to the respiratory tract, we compared intranasal (IN) and intramuscular (M) administration for safety, immunogenicity, and efficacy in different species. A single IN or IM vaccination completely protected BALB/c mice and cotton rats against RSV replication in the lungs. However, only IN administration could prevent infection in the upper respiratory tract. IM vaccination with MVA-RSV also protected cotton rats from lower respiratory tract infection in the absence of detectable neutralizing antibodies. Heterologous prime boost with PanAd3-RSV and MVA-RSV elicited high neutralizing antibody titers and broad T-cell responses in nonhuman primates. In addition, animals primed in the nose developed mucosal IgA against the F protein. In conclusion, we have shown that our vectored RSV vaccine induces potent cellular and humoral responses in a primate model, providing strong support for clinical testing.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Alessandra Taglioni
- Cellular Biology and Neurobiology Institute (IBCN) National Research Council of Italy, Rome, Italy
| | - Gemma Perretta
- Cellular Biology and Neurobiology Institute (IBCN) National Research Council of Italy, Rome, Italy
| | | | | | | | | | | | | | | | | | - Riccardo Cortese
- ReiThera Srl, Rome, Italy (former Okairos Srl)
- Keires AG, Basel, Switzerland
| | - Alfredo Nicosia
- ReiThera Srl, Rome, Italy (former Okairos Srl)
- CEINGE, Naples, Italy
- Department of Molecular Medicine and Medical Biotechnology, University Federico II, Naples, Italy
| | | | | |
Collapse
|
25
|
Lambert SL, Aslam S, Stillman E, MacPhail M, Nelson C, Ro B, Sweetwood R, Lei YM, Woo JC, Tang RS. A novel respiratory syncytial virus (RSV) F subunit vaccine adjuvanted with GLA-SE elicits robust protective TH1-type humoral and cellular immunity in rodent models. PLoS One 2015; 10:e0119509. [PMID: 25793508 PMCID: PMC4368639 DOI: 10.1371/journal.pone.0119509] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Accepted: 01/13/2015] [Indexed: 11/18/2022] Open
Abstract
Background Illness associated with Respiratory Syncytial Virus (RSV) remains an unmet medical need in both full-term infants and older adults. The fusion glycoprotein (F) of RSV, which plays a key role in RSV infection and is a target of neutralizing antibodies, is an attractive vaccine target for inducing RSV-specific immunity. Methodology and Principal Findings BALB/c mice and cotton rats, two well-characterized rodent models of RSV infection, were used to evaluate the immunogenicity of intramuscularly administered RSV vaccine candidates consisting of purified soluble F (sF) protein formulated with TLR4 agonist glucopyranosyl lipid A (GLA), stable emulsion (SE), GLA-SE, or alum adjuvants. Protection from RSV challenge, serum RSV neutralizing responses, and anti-F IgG responses were induced by all of the tested adjuvanted RSV sF vaccine formulations. However, only RSV sF + GLA-SE induced robust F-specific TH1-biased humoral and cellular responses. In mice, these F-specific cellular responses include both CD4 and CD8 T cells, with F-specific polyfunctional CD8 T cells that traffic to the mouse lung following RSV challenge. This RSV sF + GLA-SE vaccine formulation can also induce robust RSV neutralizing titers and prime IFNγ-producing T cell responses in Sprague Dawley rats. Conclusions/Significance These studies indicate that a protein subunit vaccine consisting of RSV sF + GLA-SE can induce robust neutralizing antibody and T cell responses to RSV, enhancing viral clearance via a TH1 immune-mediated mechanism. This vaccine may benefit older populations at risk for RSV disease.
Collapse
Affiliation(s)
- Stacie L. Lambert
- Department of Research, MedImmune, Mountain View, California, United States of America
- * E-mail:
| | - Shahin Aslam
- Department of Research, MedImmune, Mountain View, California, United States of America
| | - Elizabeth Stillman
- Department of Research, MedImmune, Mountain View, California, United States of America
| | - Mia MacPhail
- Department of Research, MedImmune, Mountain View, California, United States of America
| | - Christine Nelson
- Department of Research, MedImmune, Mountain View, California, United States of America
| | - Bodrey Ro
- Department of Research, MedImmune, Mountain View, California, United States of America
| | - Rosemary Sweetwood
- Department of Research, MedImmune, Mountain View, California, United States of America
| | - Yuk Man Lei
- Department of Research, MedImmune, Mountain View, California, United States of America
| | - Jennifer C. Woo
- Department of Research, MedImmune, Mountain View, California, United States of America
| | - Roderick S. Tang
- Department of Research, MedImmune, Mountain View, California, United States of America
| |
Collapse
|
26
|
Abstract
Human metapneumovirus (hMPV) and respiratory syncytial virus, its close family member, are two major causes of lower respiratory tract infection in the paediatric population. hMPV is also a common cause of worldwide morbidity and mortality in immunocompromised patients and older adults. Repeated infections occur often, demonstrating a heavy medical burden. However, there is currently no hMPV-specific prevention treatment. This review focuses on the current literature on hMPV vaccine development. We believe that a better understanding of the role(s) of viral proteins in host responses might lead to efficient prophylactic vaccine development.
Collapse
Affiliation(s)
- J Ren
- 1Department of Pediatrics, University of Texas Medical Branch, Galveston, TX, USA
| | - T Phan
- 1Department of Pediatrics, University of Texas Medical Branch, Galveston, TX, USA
| | - X Bao
- 2Institute for Translational Science, University of Texas Medical Branch, Galveston, TX, USA 3Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, USA 1Department of Pediatrics, University of Texas Medical Branch, Galveston, TX, USA
| |
Collapse
|
27
|
Yamaji Y, Nakayama T. Recombinant measles viruses expressing respiratory syncytial virus proteins induced virus-specific CTL responses in cotton rats. Vaccine 2014; 32:4529-4536. [PMID: 24951869 DOI: 10.1016/j.vaccine.2014.06.024] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Revised: 05/27/2014] [Accepted: 06/06/2014] [Indexed: 10/25/2022]
Abstract
Respiratory syncytial virus (RSV) is a common cause of serious lower respiratory tract illnesses in infants. Natural infections with RSV provide limited protection against reinfection because of inefficient immunological responses that do not induce long-term memory. RSV natural infection has been shown to induce unbalanced immune response. The effective clearance of RSV is known to require the induction of a balanced Th1/Th2 immune response, which involves the induction of cytotoxic T lymphocytes (CTL). In our previous study, recombinant AIK-C measles vaccine strains MVAIK/RSV/F and MVAIK/RSV/G were developed, which expressed the RSV fusion (F) protein or glycoprotein (G). These recombinant viruses elicited antibody responses against RSV in cotton rats, and no infectious virus was recovered, but small amounts of infiltration of inflammatory cells were observed in the lungs following RSV challenge. In the present study, recombinant AIK-C measles vaccine strains MVAIK/RSV/M2-1 and MVAIK/RSV/NP were developed, expressing RSV M2-1 or Nucleoprotein (NP), respectively. These viruses exhibited temperature-sensitivity (ts), which was derived from AIK-C, and expressed respective RSV antigens. The intramuscular inoculation of cotton rats with the recombinant measles virus led to the induction of CD8(+) IFN-γ(+) cells. No infectious virus was recovered from a lung homogenate following the challenge. A Histological examination of the lungs revealed a significant reduction in inflammatory reactions without alveolar damage. These results support the recombinant measles viruses being effective vaccine candidates against RSV that induce RSV-specific CTL responses with or without the development of an antibody response.
Collapse
Affiliation(s)
- Yoshiaki Yamaji
- Laboratory of Viral Infection I, Kitasato Institute for Life Sciences, Kitasato University, Shirokane 5-9-1, Minato-ku, Tokyo 108-8641, Japan
| | - Tetsuo Nakayama
- Laboratory of Viral Infection I, Kitasato Institute for Life Sciences, Kitasato University, Shirokane 5-9-1, Minato-ku, Tokyo 108-8641, Japan.
| |
Collapse
|
28
|
Morrison TG, Walsh EE. Subunit and virus-like particle vaccine approaches for respiratory syncytial virus. Curr Top Microbiol Immunol 2014; 372:285-306. [PMID: 24362695 DOI: 10.1007/978-3-642-38919-1_14] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Despite its impact on global health, there is no vaccine available for the prevention of respiratory syncytial virus (RSV) infection. Failure to develop a licensed vaccine is not due to lack of effort, as numerous vaccine candidates have been characterized in preclinical and clinical studies spanning five decades. The vaccine candidates thus far explored can be generally divided into four categories: (1) whole inactivated virus, (2) replication competent, attenuated virus including recombinant viruses, (3) gene-based vectors, and (4) subunit and particulate forms of RSV antigens. The first clinically tested RSV vaccine candidate was a formalin-inactivated purified virus preparation administered to infants and children in the late 1960s. Due to the disastrous outcome of these trials and results of animal models investigating the mechanisms involved, there have been no further studies with inactivated RSV vaccines. Rather, efforts have focused on development of other approaches. In this chapter, we review the history and status of purified proteins, peptides, virus-like particles, virosomes, and nanoparticles and discuss their future potential.
Collapse
Affiliation(s)
- Trudy G Morrison
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA, 01655, USA,
| | | |
Collapse
|
29
|
Sacco RE, McGill JL, Pillatzki AE, Palmer MV, Ackermann MR. Respiratory syncytial virus infection in cattle. Vet Pathol 2013; 51:427-36. [PMID: 24009269 DOI: 10.1177/0300985813501341] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Bovine respiratory syncytial virus (RSV) is a cause of respiratory disease in cattle worldwide. It has an integral role in enzootic pneumonia in young dairy calves and summer pneumonia in nursing beef calves. Furthermore, bovine RSV infection can predispose calves to secondary bacterial infection by organisms such as Mannheimia haemolytica, Pasteurella multocida, and Histophilus somni, resulting in bovine respiratory disease complex, the most prevalent cause of morbidity and mortality among feedlot cattle. Even in cases where animals do not succumb to bovine respiratory disease complex, there can be long-term losses in production performance. This includes reductions in feed efficiency and rate of gain in the feedlot, as well as reproductive performance, milk production, and longevity in the breeding herd. As a result, economic costs to the cattle industry from bovine respiratory disease have been estimated to approach $1 billion annually due to death losses, reduced performance, and costs of vaccinations and treatment modalities. Human and bovine RSV are closely related viruses with similarities in histopathologic lesions and mechanisms of immune modulation induced following infection. Therefore, where appropriate, we provide comparisons between RSV infections in humans and cattle. This review article discusses key aspects of RSV infection of cattle, including epidemiology and strain variability, clinical signs and diagnosis, experimental infection, gross and microscopic lesions, innate and adaptive immune responses, and vaccination strategies.
Collapse
Affiliation(s)
- R E Sacco
- National Animal Disease Center, Ruminant Diseases and Immunology Research Unit, Ames, IA 50010, USA.
| | | | | | | | | |
Collapse
|
30
|
Iwata H, Bieber K, Tiburzy B, Chrobok N, Kalies K, Shimizu A, Leineweber S, Ishiko A, Vorobyev A, Zillikens D, Köhl J, Westermann J, Seeger K, Manz R, Ludwig RJ. B Cells, Dendritic Cells, and Macrophages Are Required To Induce an Autoreactive CD4 Helper T Cell Response in Experimental Epidermolysis Bullosa Acquisita. THE JOURNAL OF IMMUNOLOGY 2013; 191:2978-88. [DOI: 10.4049/jimmunol.1300310] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
31
|
Regulatory T cells prevent Th2 immune responses and pulmonary eosinophilia during respiratory syncytial virus infection in mice. J Virol 2013; 87:10946-54. [PMID: 23926350 PMCID: PMC3807299 DOI: 10.1128/jvi.01295-13] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
During viral infection, inflammation and recovery are tightly controlled by competing proinflammatory and regulatory immune pathways. Respiratory syncytial virus (RSV) is the leading global cause of infantile bronchiolitis, which is associated with recurrent wheeze and asthma diagnosis in later life. Th2-driven disease has been well described under some conditions for RSV-infected mice. In the present studies, we used the Foxp3DTR mice (which allow specific conditional depletion of Foxp3+ T cells) to investigate the functional effects of regulatory T cells (Tregs) during A2-strain RSV infection. Infected Treg-depleted mice lost significantly more weight than wild-type mice, indicating enhanced disease. This enhancement was characterized by increased cellularity in the bronchoalveolar lavage (BAL) fluid and notable lung eosinophilia not seen in control mice. This was accompanied by abundant CD4+ and CD8+ T cells exhibiting an activated phenotype and induction of interleukin 13 (IL-13)- and GATA3-expressing Th2-type CD4+ T cells that remained present in the airways even 14 days after infection. Therefore, Treg cells perform vital anti-inflammatory functions during RSV infection, suppressing pathogenic T cell responses and inhibiting lung eosinophilia. These findings provide additional evidence that dysregulation of normal immune responses to viral infection may contribute to severe RSV disease.
Collapse
|
32
|
Benefit and harm from immunity to respiratory syncytial virus: implications for treatment. Curr Opin Infect Dis 2013; 25:687-94. [PMID: 23086186 DOI: 10.1097/qco.0b013e32835a1d92] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
PURPOSE OF REVIEW Human respiratory syncytial virus (RSV) infection is a major cause of morbidity in children and of morbidity and mortality in elderly or immunocompromised adults. Given prophylactically, antibody can protect against infection, but natural levels are poorly protective. Vaccination may enhance disease, and there is no well tolerated and effective vaccine or antiviral treatment. Despite over 50 years of research, therapy remains nonspecific and supportive. RECENT FINDINGS Experimental human challenge in adult volunteers is beginning to elucidate the dynamics of viral shedding and causes of disease, but investigations of naturally infected children remain logistically challenging. RSV was known to bind several surface ligands, but the recent demonstration that nucleolin acts as a receptor for the RSV fusion protein was unexpected. Recent studies increasingly emphasize the relevance of innate immune responses and the dysregulation of inflammation as key factors in causing the pathological effects of infection. Studies in both human infants and mice indicate that interleukin-17 plays a role in some forms of RSV disease and regulatory T cells may be important in controlling inflammation. SUMMARY Improved understanding of the human immune response to RSV infection continues to be needed in order to accelerate the development of vaccines and new treatments for bronchiolitis.
Collapse
|
33
|
Neonatal calf infection with respiratory syncytial virus: drawing parallels to the disease in human infants. Viruses 2013; 4:3731-53. [PMID: 23342375 PMCID: PMC3528288 DOI: 10.3390/v4123731] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Respiratory syncytial virus (RSV) is the most common viral cause of childhood acute lower respiratory tract infections. It is estimated that RSV infections result in more than 100,000 deaths annually worldwide. Bovine RSV is a cause of enzootic pneumonia in young dairy calves and summer pneumonia in nursing beef calves. Furthermore, bovine RSV plays a significant role in bovine respiratory disease complex, the most prevalent cause of morbidity and mortality among feedlot cattle. Infection of calves with bovine RSV shares features in common with RSV infection in children, such as an age-dependent susceptibility. In addition, comparable microscopic lesions consisting of bronchiolar neutrophilic infiltrates, epithelial cell necrosis, and syncytial cell formation are observed. Further, our studies have shown an upregulation of pro-inflammatory mediators in RSV-infected calves, including IL-12p40 and CXCL8 (IL-8). This finding is consistent with increased levels of IL-8 observed in children with RSV bronchiolitis. Since rodents lack IL-8, neonatal calves can be useful for studies of IL-8 regulation in response to RSV infection. We have recently found that vitamin D in milk replacer diets can be manipulated to produce calves differing in circulating 25-hydroxyvitamin D3. The results to date indicate that although the vitamin D intracrine pathway is activated during RSV infection, pro-inflammatory mediators frequently inhibited by the vitamin D intacrine pathway in vitro are, in fact, upregulated or unaffected in lungs of infected calves. This review will summarize available data that provide parallels between bovine RSV infection in neonatal calves and human RSV in infants.
Collapse
|
34
|
|
35
|
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
|
36
|
Bayon JCL, Lina B, Rosa-Calatrava M, Boivin G. Recent developments with live-attenuated recombinant paramyxovirus vaccines. Rev Med Virol 2012; 23:15-34. [DOI: 10.1002/rmv.1717] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2011] [Revised: 03/14/2012] [Accepted: 03/22/2012] [Indexed: 12/30/2022]
Affiliation(s)
- Jean-Christophe Le Bayon
- Laboratoire de Virologie et Pathologie Humaine, VirPath EMR 4610/Equipe VirCell, Université de Lyon; Université Claude Bernard Lyon 1 - Hospices Civils de Lyon, Faculté de médecine RTH Laennec; Lyon France
- Research Center in Infectious Diseases; CHUQ-CHUL and Université Laval; Québec City QC Canada
| | - Bruno Lina
- Laboratoire de Virologie et Pathologie Humaine, VirPath EMR 4610/Equipe VirCell, Université de Lyon; Université Claude Bernard Lyon 1 - Hospices Civils de Lyon, Faculté de médecine RTH Laennec; Lyon France
- Laboratoire de Virologie, Centre de Biologie et de Pathologie Est; Hospices Civils de Lyon; Lyon Bron Cedex France
| | - Manuel Rosa-Calatrava
- Laboratoire de Virologie et Pathologie Humaine, VirPath EMR 4610/Equipe VirCell, Université de Lyon; Université Claude Bernard Lyon 1 - Hospices Civils de Lyon, Faculté de médecine RTH Laennec; Lyon France
| | - Guy Boivin
- Research Center in Infectious Diseases; CHUQ-CHUL and Université Laval; Québec City QC Canada
| |
Collapse
|
37
|
Interleukin-27 inhibits vaccine-enhanced pulmonary disease following respiratory syncytial virus infection by regulating cellular memory responses. J Virol 2012; 86:4505-17. [PMID: 22301139 DOI: 10.1128/jvi.07091-11] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Respiratory syncytial virus (RSV) is the most important cause of lower respiratory tract disease in young children. In the 1960s, infants vaccinated with formalin-inactivated RSV developed a more severe disease characterized by excessive inflammatory immunopathology in lungs upon natural RSV infection. The fear of causing the vaccine-enhanced disease (VED) is an important obstacle for development of safe and effective RSV vaccines. The recombinant vaccine candidate G1F/M2 immunization also led to VED. It has been proved that cellular memory induced by RSV vaccines contributed to VED. Interleukin-27 (IL-27) and IL-23 regulate Th1, Th17, and/or Th2 cellular immune responses. In this study, mice coimmunized with pcDNA3-IL-27 and G1F/M2 were fully protected and, importantly, did not develop vaccine-enhanced inflammatory responses and immunopathology in lungs after RSV challenge, which was correlated with moderate Th1-, suppressed Th2-, and Th17-like memory responses activated by RSV. In contrast, G1F/M2- or pcDNA3-IL-23+G1F/M2-immunized mice, in which robust Th2- and Th17-like memory responses were induced, developed enhanced pulmonary inflammation and severe immunopathology. Mice coimmunized with G1F/M2 and the two cytokine plasmids exhibited mild inflammatory responses as well as remarkable Th1-, suppressed Th2-, and Th17-like memory responses. These results suggested that Th1-, Th2-, and Th17-like memory responses and, in particular, excessive Th2- and Th17-like memory responses were closely associated with VED; IL-27 may inhibit VED following respiratory syncytial virus infection by regulating cellular memory responses.
Collapse
|
38
|
Shao HY, Lin YW, Yu SL, Lin HY, Chitra E, Chang YC, Sia C, Chong P, Hsu MT, Wei OL, Chow YH. Immunoprotectivity of HLA-A2 CTL peptides derived from respiratory syncytial virus fusion protein in HLA-A2 transgenic mouse. PLoS One 2011; 6:e25500. [PMID: 21980478 PMCID: PMC3183052 DOI: 10.1371/journal.pone.0025500] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2011] [Accepted: 09/06/2011] [Indexed: 11/18/2022] Open
Abstract
Identification of HLA-restricted CD8+ T cell epitopes is important to study RSV-induced immunity and illness. We algorithmically analyzed the sequence of the fusion protein (F) of respiratory syncytial virus (RSV) and generated synthetic peptides that can potentially bind to HLA-A*0201. Four out of the twenty-five 9-mer peptides tested: peptides 3 (F33–41), 13 (F214–222), 14 (F273–281), and 23 (F559–567), were found to bind to HLA-A*0201 with moderate to high affinity and were capable of inducing IFN-γ and IL-2 secretion in lymphocytes from HLA-A*0201 transgenic (HLA-Tg) mice pre-immunized with RSV or recombinant adenovirus expressing RSV F. HLA-Tg mice were immunized with these four peptides and were found to induce both Th1 and CD8+ T cell responses in in vitro secondary recall. Effector responses induced by these peptides were observed to confer differential protection against live RSV challenge. These peptides also caused better recovery of body weight loss induced by RSV. A significant reduction of lung viral load was observed in mice immunized with peptide 23, which appeared to enhance the levels of inflammatory chemokines (CCL17, CCL22, and IL-18) but did not increase eosinophil infiltration in the lungs. Whereas, significant reduction of infiltrated eosinophils induced by RSV infection was found in mice pre-immunized with peptide 13. Our results suggest that HLA-A2-restricted epitopes of RSV F protein could be useful for the development of epitope-based RSV vaccine.
Collapse
Affiliation(s)
- Hsiao-Yun Shao
- Institute of Infectious Disease and Vaccinology, National Health Research Institutes, Miaoli County, Taiwan, Republic of China
| | - Yi-Wen Lin
- Institute of Infectious Disease and Vaccinology, National Health Research Institutes, Miaoli County, Taiwan, Republic of China
- Graduate Program of Biotechnology in Medicine, Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, Taiwan
| | - Shu-Ling Yu
- Institute of Infectious Disease and Vaccinology, National Health Research Institutes, Miaoli County, Taiwan, Republic of China
| | - Hsiang-Yin Lin
- Institute of Infectious Disease and Vaccinology, National Health Research Institutes, Miaoli County, Taiwan, Republic of China
| | - Ebenezer Chitra
- Institute of Infectious Disease and Vaccinology, National Health Research Institutes, Miaoli County, Taiwan, Republic of China
| | - Yung-Chen Chang
- Institute of Infectious Disease and Vaccinology, National Health Research Institutes, Miaoli County, Taiwan, Republic of China
| | - Charles Sia
- Institute of Infectious Disease and Vaccinology, National Health Research Institutes, Miaoli County, Taiwan, Republic of China
| | - Pele Chong
- Institute of Infectious Disease and Vaccinology, National Health Research Institutes, Miaoli County, Taiwan, Republic of China
| | - Ming-Tao Hsu
- Institute of Infectious Disease and Vaccinology, National Health Research Institutes, Miaoli County, Taiwan, Republic of China
| | - Olivia L. Wei
- The Graduate Division of Biological and Biomedical Sciences (GDBBS), Emory University, Atlanta, Georgia, United States of America
| | - Yen-Hung Chow
- Institute of Infectious Disease and Vaccinology, National Health Research Institutes, Miaoli County, Taiwan, Republic of China
- * E-mail:
| |
Collapse
|
39
|
Krause A, Xu Y, Ross S, Wu W, Joh J, Worgall S. Absence of vaccine-enhanced RSV disease and changes in pulmonary dendritic cells with adenovirus-based RSV vaccine. Virol J 2011; 8:375. [PMID: 21801372 PMCID: PMC3166937 DOI: 10.1186/1743-422x-8-375] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2011] [Accepted: 07/29/2011] [Indexed: 01/22/2023] Open
Abstract
The development of a vaccine against respiratory syncytial virus (RSV) has been hampered by the risk for vaccine-enhanced RSV pulmonary disease induced by immunization with formalin-inactivated RSV (FIRSV). This study focuses on the evaluation of vaccine-enhanced pulmonary disease following immunization with AdF.RGD, an integrin-targeted adenovirus vector that expresses the RSV F protein and includes an RGD (Arg-Gly-Asp) motif. Immunization of BALB/c mice with AdF.RGD, resulted in anti-RSV protective immunity and induced increased RSV-specific IFN-γ T cell responses compared to FIRSV. RSV infection 5 wk after immunization with FIRSV induced pulmonary inflammatory responses in the lung, that was not observed with AdF.RGD. Additionally, In the FIRSV-immunized mice following infection with RSV, pulmonary DC increased and Tregs decreased. This suggests that distinct responses of pulmonary DC and Tregs are a features of vaccine-enhanced RSV disease and that immunization with an RGD-modified Ad vaccine does not trigger vaccine-enhanced disease.
Collapse
Affiliation(s)
- Anja Krause
- Department of Genetic Medicine, Weill Medical College of Cornell University, New York, New York, USA
| | | | | | | | | | | |
Collapse
|
40
|
Effects of respiratory syncytial virus infection and major basic protein derived from eosinophils in pulmonary alveolar epithelial cells (A549). Cell Biol Int 2011; 35:467-74. [PMID: 20977431 DOI: 10.1042/cbi20100255] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
RSV (respiratory syncytial virus)-induced pneumonia and bronchiolitis may be associated with hyperresponsive conditions, including asthma. Eosinophilic proteins such as MBP (major basic protein) may also be associated with the pathophysiology of asthma. To elucidate the roles of RSV infection and MBP in the pathogenesis of pneumonia with hyperresponsiveness, we investigated the effects of RSV infection and MBP on A549 (alveolar epithelial) cells. CPE (cytopathic effects) in A549 cells were observed by microscopy. Apoptosis and cell death was evaluated by flow cytometric analysis and modified MTT [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide] assay. We also measured 15 types of cytokines and chemokines in A549 cell supernatants. Although RSV alone did not affect the CPE of A549, high concentrations of MBP resulted in cell death within 24 h. Combinations of RSV and MBP synergistically induced cell death. In A549 cells infected with RSV alone, the release of GM-CSF (granulocyte-macrophage colony-stimulating factor) was significantly enhanced compared with control cells (no infection). In the cells treated with MBP alone, the production of IL (interleukin)-2, 4, 5, 7, 10, 12, 13, 17, IFN (interferon)-γ, GM-CSF, G-CSF (granulocyte colony-stimulating factor) and MIP (macrophage inflammatory protein)-1β was significantly increased compared with control cells. Notably, the levels of GM-CSF and IL-17 in RSV/MBP-treated cells were significantly higher than those treated with MBP alone. These results suggest that MBP synergistically enhanced the release of various cytokines/chemokines and the cell death of RSV-infected A549 cells, indicating that MBP may be closely associated with the pathophysiology of allergic reactions in bronchiolitis/pneumonia due to RSV.
Collapse
|
41
|
Munir S, Hillyer P, Le Nouën C, Buchholz UJ, Rabin RL, Collins PL, Bukreyev A. Respiratory syncytial virus interferon antagonist NS1 protein suppresses and skews the human T lymphocyte response. PLoS Pathog 2011; 7:e1001336. [PMID: 21533073 PMCID: PMC3080852 DOI: 10.1371/journal.ppat.1001336] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2010] [Accepted: 03/23/2011] [Indexed: 12/17/2022] Open
Abstract
We recently demonstrated that the respiratory syncytial virus (RSV) NS1 protein, an antagonist of host type I interferon (IFN-I) production and signaling, has a suppressive effect on the maturation of human dendritic cells (DC) that was only partly dependent on released IFN-I. Here we investigated whether NS1 affects the ability of DC to activate CD8+ and CD4+ T cells. Human DC were infected with RSV deletion mutants lacking the NS1 and/or NS2 genes and assayed for the ability to activate autologous T cells in vitro, which were analyzed by multi-color flow cytometry. Deletion of the NS1, but not NS2, protein resulted in three major effects: (i) an increased activation and proliferation of CD8+ T cells that express CD103, a tissue homing integrin that directs CD8+ T cells to mucosal epithelial cells of the respiratory tract and triggers cytolytic activity; (ii) an increased activation and proliferation of Th17 cells, which have recently been shown to have anti-viral effects and also indirectly attract neutrophils; and (iii) decreased activation of IL-4-producing CD4+ T cells--which are associated with enhanced RSV disease--and reduced proliferation of total CD4+ T cells. Except for total CD4+ T cell proliferation, none of the T cell effects appeared to be due to increased IFN-I signaling. In the infected DC, deletion of the NS1 and NS2 genes strongly up-regulated the expression of cytokines and other molecules involved in DC maturation. This was partly IFN-I-independent, and thus might account for the T cell effects. Taken together, these data demonstrate that the NS1 protein suppresses proliferation and activation of two of the protective cell populations (CD103+ CD8+ T cells and Th17 cells), and promotes proliferation and activation of Th2 cells that can enhance RSV disease.
Collapse
Affiliation(s)
- Shirin Munir
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Philippa Hillyer
- Center for Biologics Evaluation and Research, US Food and Drug Administration, Bethesda, Maryland, United States of America
| | - Cyril Le Nouën
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Ursula J. Buchholz
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Ronald L. Rabin
- Center for Biologics Evaluation and Research, US Food and Drug Administration, Bethesda, Maryland, United States of America
| | - Peter L. Collins
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Alexander Bukreyev
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail:
| |
Collapse
|
42
|
Kruijsen D, Schijf MA, Lukens MV, van Uden NO, Kimpen JL, Coenjaerts FE, van Bleek GM. Local innate and adaptive immune responses regulate inflammatory cell influx into the lungs after vaccination with formalin inactivated RSV. Vaccine 2011; 29:2730-41. [PMID: 21316502 DOI: 10.1016/j.vaccine.2011.01.087] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2010] [Revised: 01/21/2011] [Accepted: 01/27/2011] [Indexed: 11/29/2022]
Abstract
Inactivated respiratory syncytial virus (RSV) vaccines tend to predispose for immune mediated enhanced disease, characterized by Th2 responses and airway hypersensitivity reactions. We show in a C57BL/6 mouse model that the early innate response elicited by the challenge virus (RSV versus influenza virus) influences the outcome of the Th1/Th2 balance in the lung after intramuscular priming with inactivated vaccine. Priming of CD4(+)/IFN-γ(+) T cells by mature dendritic cells administered intravenously and/or priming of a virus specific CD8(+) T cell response ameliorated the Th2-mediated inflammatory response in the lung, suggesting that vaccination procedures are feasible that prevent vaccine induced immune pathology.
Collapse
Affiliation(s)
- Debby Kruijsen
- Department of Pediatrics, The Wilhelmina Children's Hospital, University Medical Centre, Utrecht, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
43
|
Melendi GA, Bridget D, Monsalvo AC, Laham FF, Acosta P, Delgado MF, Polack FP, Irusta PM. Conserved cysteine residues within the attachment G glycoprotein of respiratory syncytial virus play a critical role in the enhancement of cytotoxic T-lymphocyte responses. Virus Genes 2010; 42:46-54. [PMID: 21053062 PMCID: PMC5454483 DOI: 10.1007/s11262-010-0545-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2010] [Accepted: 10/18/2010] [Indexed: 01/21/2023]
Abstract
The cytotoxic T-lymphocyte (CTL) response plays an important role in the control of respiratory syncytial virus (RSV) replication and the establishment of a Th1-CD4+ T cell response against the virus. Despite lacking Major Histocompatibility Complex I (MHC I)-restricted epitopes, the attachment G glycoprotein of RSV enhances CTL activity toward other RSV antigens, and this effect depends on its conserved central region. Here, we report that RSV-G can also improve CTL activity toward antigens from unrelated pathogens such as influenza, and that a mutant form of RSV-G lacking four conserved cysteine residues at positions 173, 176, 182, and 186 fails to enhance CTL responses. Our results indicate that these conserved residues are essential for the wide-spectrum pro-CTL activity displayed by the protein.
Collapse
|
44
|
Transforming growth factor beta is a major regulator of human neonatal immune responses following respiratory syncytial virus infection. J Virol 2010; 84:12895-902. [PMID: 20926560 DOI: 10.1128/jvi.01273-10] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Respiratory syncytial virus (RSV) is a major cause of morbidity and mortality. Previous studies have suggested that T-cell responses may contribute to RSV immunopathology, which could be driven by dendritic cells (DCs). DCs are productively infected by RSV, and during RSV infections, there is an increase of DCs in the lungs with a decrease in the blood. Pediatric populations are particularly susceptible to severe RSV infections; however, DC responses to RSV from pediatric populations have not been examined. In this study, primary isolated DCs from cord blood and adult peripheral blood were compared after RSV infection. Transcriptional profiling and biological network analysis identified transforming growth factor beta (TGF-β) and associated signaling molecules as differentially regulated in the two age groups. TGF-β1 was decreased in RSV-infected adult-blood DCs but increased in RSV-infected cord blood DCs. Coculture of adult RSV-infected DCs with autologous T cells induced secretion of gamma interferon (IFN-γ), interleukin 12p70 (IL-12p70), IL-2, and tumor necrosis factor alpha (TNF-α). Conversely, coculture of cord RSV-infected DCs and autologous T cells induced secretion of IL-4, IL-6, IL-1β, and IL-17. Addition of purified TGF-β1 to adult DC-T-cell cocultures reduced secretion of IFN-γ, IL-12p70, IL-2, and TNF-α, while addition of a TGF-β chemical inhibitor to cord DC-T-cell cocultures increased secretion of IL-12p70. These data suggest that TGF-β acts as a major regulator of RSV DC-T-cell responses, which could contribute to immunopathology during infancy.
Collapse
|
45
|
DeVincenzo JP, Wilkinson T, Vaishnaw A, Cehelsky J, Meyers R, Nochur S, Harrison L, Meeking P, Mann A, Moane E, Oxford J, Pareek R, Moore R, Walsh E, Studholme R, Dorsett P, Alvarez R, Lambkin-Williams R. Viral load drives disease in humans experimentally infected with respiratory syncytial virus. Am J Respir Crit Care Med 2010; 182:1305-14. [PMID: 20622030 DOI: 10.1164/rccm.201002-0221oc] [Citation(s) in RCA: 250] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
RATIONALE Respiratory syncytial virus (RSV) is the leading cause of childhood lower respiratory infection, yet viable therapies are lacking. Two major challenges have stalled antiviral development: ethical difficulties in performing pediatric proof-of-concept studies and the prevailing concept that the disease is immune-mediated rather than being driven by viral load. OBJECTIVES The development of a human experimental wild-type RSV infection model to address these challenges. METHODS Healthy volunteers (n = 35), in five cohorts, received increasing quantities (3.0-5.4 log plaque-forming units/person) of wild-type RSV-A intranasally. MEASUREMENTS AND MAIN RESULTS Overall, 77% of volunteers consistently shed virus. Infection rate, viral loads, disease severity, and safety were similar between cohorts and were unrelated to quantity of RSV received. Symptoms began near the time of initial viral detection, peaked in severity near when viral load peaked, and subsided as viral loads (measured by real-time polymerase chain reaction) slowly declined. Viral loads correlated significantly with intranasal proinflammatory cytokine concentrations (IL-6 and IL-8). Increased viral load correlated consistently with increases in multiple different disease measurements (symptoms, physical examination, and amount of nasal mucus). CONCLUSIONS Viral load appears to drive disease manifestations in humans with RSV infection. The observed parallel viral and disease kinetics support a potential clinical benefit of RSV antivirals. This reproducible model facilitates the development of future RSV therapeutics.
Collapse
Affiliation(s)
- John P DeVincenzo
- Department of Pediatrics, University of Tennessee College of Medicine, Memphis, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Stevens WW, Sun J, Castillo JP, Braciale TJ. Pulmonary eosinophilia is attenuated by early responding CD8(+) memory T cells in a murine model of RSV vaccine-enhanced disease. Viral Immunol 2009; 22:243-51. [PMID: 19594395 DOI: 10.1089/vim.2009.0016] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Vaccination with formalin-inactivated respiratory syncytial virus (RSV) vaccine results in enhanced respiratory tract inflammation and injury following subsequent RSV infection. RSV vaccine-enhanced disease can also be produced in mice by prior vaccination with a vaccinia virus vector containing the RSV G protein, followed by intranasal infectious RSV challenge, a process characterized by induction of a potent memory CD4(+) T-cell response to challenge infection with some features characteristic of Th-2 CD4(+) T-cell responses, including increased eosinophil accumulation in pulmonary inflammatory infiltrates. The adaptive immune response to the RSV G protein in immunized BALB/c mice is characterized by a weak or absent primary and secondary recall CD8(+) T-cell response. These and related results have led to the hypothesis that the failure of the infected animals to mount an effective CD8(+) memory T-cell (CD8(+) Tm) response in this model could account for the pulmonary eosinophilia associated with the development of enhanced disease, and that CD8(+) T cells may control the development of eosinophilia. In this study, we investigated how and when the generation of a CD8(+) Tm response to RSV infection might affect the development of pulmonary eosinophilia in this model of vaccine-enhanced disease. By defining the CD8(+) T-cell response kinetics and monitoring lung parenchymal eosinophil accumulation, we show that the establishment of an RSV-specific CD8(+) Tm response in the infected lungs early after challenge infection (i.e., within the first 3 d of RSV infection) is necessary and sufficient to control pulmonary eosinophilia development. Additionally, our work suggests that the mechanism by which CD8(+) T cells regulate this process is not by modulating the differentiation or development of the CD4(+) Tm response. Rather, we demonstrate that IL-10 produced by early responding CD8(+) Tm cells may regulate the pulmonary eosinophilia development observed in RSV vaccine-enhanced disease.
Collapse
Affiliation(s)
- Whitney W Stevens
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, Virginia 22908, USA
| | | | | | | |
Collapse
|
47
|
Shao HY, Yu SL, Sia C, Chen Y, Chitra E, Chen IH, Venkatesan N, Leng CH, Chong P, Chow YH. Immunogenic properties of RSV-B1 fusion (F) protein gene-encoding recombinant adenoviruses. Vaccine 2009; 27:5460-71. [PMID: 19622401 DOI: 10.1016/j.vaccine.2009.07.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2009] [Revised: 06/16/2009] [Accepted: 07/01/2009] [Indexed: 12/29/2022]
Abstract
Two recombinant adenoviruses designated rAd-F0DeltaTM and rAd-F0 carrying the transmembrane truncated and full length of the F gene of the RSV-B1 strain, respectively, were engineered. Comparative immunogenicity studies in BALB/c mice showed that each vector was capable of inducing RSV-B1-specific antibodies that cross-reacted with the RSV-long and RSV-A2 viruses. The anti-RSV-B1 antibodies were neutralizing, and exhibited strong cross-neutralizing activity against the RSV-long and RSV-A2 isolates as well. Analysis of the cellular responses revealed that animals immunized with rAd-F0DeltaTM and rAd-F0 elicited CD4(+) T-cell responses of the Th1 and Th2 phenotypes, as well as F protein-specific CTLs. Production of Th2 cytokines (IL-4, IL-5 and IL-13) by splenocytes of the rAd-F0DeltaTM and rAd-F0 immunized mice was markedly lower than those released by animals administered with heat-inactivated RSV-B1 (HIRSV-B1). Comparison of the overall humoral and cellular responses suggests that rAd-F0DeltaTM is significantly more immunogenic than rAd-F0. The anti-viral immunity generated by both recombinant adenovirus vectors has conferred protection against live RSV-B1 challenge as judged by the lower viral load recovered in the lungs, a faster rate of recovery of body weight loss, and a lower count of eosinophils as compared to eosinophilia in mice immunized with HIRSV-B1. Results from these studies suggest that rAd-F0DeltaTM or rAd-F0 possess immunogenic properties that meet the requirements expected from potential RSV vaccine candidates.
Collapse
Affiliation(s)
- Hsiao-Yun Shao
- Vaccine R&D Center, National Health Research Institutes, Zhunan Town, Miaoli County 350, Taiwan, ROC
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Oshansky CM, Zhang W, Moore E, Tripp RA. The host response and molecular pathogenesis associated with respiratory syncytial virus infection. Future Microbiol 2009; 4:279-97. [PMID: 19327115 DOI: 10.2217/fmb.09.1] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Since the isolation of respiratory syncytial virus (RSV) in 1956, its significance as an important human pathogen in infants, the elderly and the immunocompromised has been established. Many important mechanisms contributing to RSV infection, replication and disease pathogenesis have been uncovered; however, there is still insufficient knowledge in these and related areas, which must be addressed to facilitate the development of safe and effective vaccines and therapeutic treatments. A better understanding of the molecular pathogenesis of RSV infection, particularly the host-cell response and transcription profiles to RSV infection, is required to advance disease intervention strategies. Substantial information is accumulating regarding how RSV proteins modulate molecular signaling and regulation of cytokine and chemokine responses to infection, molecular signals regulating programmed cell death, and innate and adaptive immune responses to infection. This review discusses RSV manipulation of the host response to infection and related disease pathogenesis.
Collapse
Affiliation(s)
- Christine M Oshansky
- Department of Infectious Diseases, University of Georgia, Athens, GA 30602, USA.
| | | | | | | |
Collapse
|
49
|
Shirato K, Taguchi F. Mast cell degranulation is induced by A549 airway epithelial cell infected with respiratory syncytial virus. Virology 2009; 386:88-93. [PMID: 19195674 DOI: 10.1016/j.virol.2009.01.011] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2008] [Revised: 11/10/2008] [Accepted: 01/07/2009] [Indexed: 10/21/2022]
Abstract
Respiratory syncytial virus (RSV), a major causative agent of respiratory tract infections, influences allergic diseases. Mast cells, important effector cells in allergic disease, also express chemokine (C-X(3)-C motif) receptor 1 (CX(3)CR1). The RSV attachment glycoprotein (G protein) is structurally similar to CX(3)C ligand 1 (CX(3)CL1), the CX(3)CR1 ligand, suggesting that RSV directly interacts with and affects mast cell function, including degranulation. In this paper, the effect of RSV infection on mast cell function was studied using the human mast cell line (HMC-1). The results showed that RSV infection and replication was inefficient in HMC-1 cells than in human epithelial A549 cells. Additionally, HMC-1 degranulation occurred only in coculture with RSV-infected A549 cells, with up-regulation of TNFalpha secretion. However, direct RSV inoculation and incubation with RSV-infected A549 cell culture medium failed to induce HMC-1 degranulation, suggesting that virus-infected cells are critical for degranulation during RSV infection; however, degranulation does not occur by direct RSV infection into mast cells.
Collapse
Affiliation(s)
- Kazuya Shirato
- Laboratory of Acute Respiratory Viral Diseases and Cytokines, Department of Virology III, National Institute of Infectious Diseases, Murayama Branch 4-7-1 Gakuen, Musashimurayama, Tokyo 208-0011 Japan.
| | | |
Collapse
|
50
|
Regulatory T cells promote early influx of CD8+ T cells in the lungs of respiratory syncytial virus-infected mice and diminish immunodominance disparities. J Virol 2009; 83:3019-28. [PMID: 19153229 DOI: 10.1128/jvi.00036-09] [Citation(s) in RCA: 109] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
In addition to regulating autoimmunity and antitumor immunity, CD4(+) CD25(+) FoxP3(+) natural regulatory T (Treg) cells are global regulators of adaptive immune responses. Depletion of these cells with the anti-CD25 antibody PC61 prior to primary respiratory syncytial virus (RSV) infection was partial but had several effects on the RSV-specific CD8(+) response in a hybrid mouse model. Mediastinal lymph node and spleen epitope-specific CD8(+) T-cell responses were enhanced in Treg-cell-depleted mice at all time points following infection, but responses of Treg-cell-depleted lung show a strikingly different pattern than lymphoid organ responses, with an initial delay in the CD8(+) T-cell response. The delay in the CD8(+) T-cell response correlated with a delay both in the early phase of viral clearance and in illness in Treg-cell-depleted mice compared to isotype-treated controls. The lungs of Treg-cell-depleted mice were shown to have increased lung chemokine and cytokine levels 7 days postinfection despite lower CD8(+) T-cell responses. Following the early delay in the lung response, CD8(+) T-cell responses at later infection time points were enhanced and increased the severity of illness in depleted mice. Finally, decreasing regulatory T-cell control of the CD8(+) T-cell response had a greater effect on response of the dominant K(d)-restricted M2 epitope consisting of amino acids 82 to 90 (K(d)M2(82-90)) than on the subdominant D(b)M(187-195) epitope response, indicating that regulatory T cells modulate immunodominance disparities in epitope-specific CD8(+) T-cell responses following primary RSV infection.
Collapse
|