51
|
Ong CWM, Migliori GB, Raviglione M, MacGregor-Skinner G, Sotgiu G, Alffenaar JW, Tiberi S, Adlhoch C, Alonzi T, Archuleta S, Brusin S, Cambau E, Capobianchi MR, Castilletti C, Centis R, Cirillo DM, D'Ambrosio L, Delogu G, Esposito SMR, Figueroa J, Friedland JS, Ho BCH, Ippolito G, Jankovic M, Kim HY, Rosales Klintz S, Ködmön C, Lalle E, Leo YS, Leung CC, Märtson AG, Melazzini MG, Najafi Fard S, Penttinen P, Petrone L, Petruccioli E, Pontali E, Saderi L, Santin M, Spanevello A, van Crevel R, van der Werf MJ, Visca D, Viveiros M, Zellweger JP, Zumla A, Goletti D. Epidemic and pandemic viral infections: impact on tuberculosis and the lung: A consensus by the World Association for Infectious Diseases and Immunological Disorders (WAidid), Global Tuberculosis Network (GTN), and members of the European Society of Clinical Microbiology and Infectious Diseases Study Group for Mycobacterial Infections (ESGMYC). Eur Respir J 2020; 56:2001727. [PMID: 32586885 PMCID: PMC7527651 DOI: 10.1183/13993003.01727-2020] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 06/12/2020] [Indexed: 01/08/2023]
Abstract
Major epidemics, including some that qualify as pandemics, such as severe acute respiratory syndrome (SARS), Middle East respiratory syndrome (MERS), HIV, influenza A (H1N1)pdm/09 and most recently COVID-19, affect the lung. Tuberculosis (TB) remains the top infectious disease killer, but apart from syndemic TB/HIV little is known regarding the interaction of viral epidemics and pandemics with TB. The aim of this consensus-based document is to describe the effects of viral infections resulting in epidemics and pandemics that affect the lung (MERS, SARS, HIV, influenza A (H1N1)pdm/09 and COVID-19) and their interactions with TB. A search of the scientific literature was performed. A writing committee of international experts including the European Centre for Disease Prevention and Control Public Health Emergency (ECDC PHE) team, the World Association for Infectious Diseases and Immunological Disorders (WAidid), the Global Tuberculosis Network (GTN), and members of the European Society of Clinical Microbiology and Infectious Diseases (ESCMID) Study Group for Mycobacterial Infections (ESGMYC) was established. Consensus was achieved after multiple rounds of revisions between the writing committee and a larger expert group. A Delphi process involving the core group of authors (excluding the ECDC PHE team) identified the areas requiring review/consensus, followed by a second round to refine the definitive consensus elements. The epidemiology and immunology of these viral infections and their interactions with TB are discussed with implications for diagnosis, treatment and prevention of airborne infections (infection control, viral containment and workplace safety). This consensus document represents a rapid and comprehensive summary on what is known on the topic.
Collapse
Affiliation(s)
- Catherine Wei Min Ong
- Dept of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Institute for Health Innovation and Technology (iHealthtech), National University of Singapore, Singapore
- These authors contributed equally
- Members of ESGMYC
| | - Giovanni Battista Migliori
- Servizio di Epidemiologia Clinica delle Malattie Respiratorie, Istituti Clinici Scientifici Maugeri IRCCS, Tradate, Italy
- These authors contributed equally
| | - Mario Raviglione
- Centre for Multidisciplinary Research in Health Science, University of Milan, Milan, Italy
- Global Studies Institute, University of Geneva, Geneva, Switzerland
| | | | - Giovanni Sotgiu
- Clinical Epidemiology and Medical Statistics Unit, Dept of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
| | - Jan-Willem Alffenaar
- Sydney Pharmacy School, University of Sydney, Sydney, Australia
- Westmead Hospital, Sydney, Australia
- Marie Bashir Institute of Infectious Diseases and Biosecurity, University of Sydney, Sydney, Australia
- Members of ESGMYC
| | - Simon Tiberi
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- Division of Infection, Royal London Hospital, Barts Health NHS Trust, London, UK
- Members of ESGMYC
| | - Cornelia Adlhoch
- Public Health Emergency Team, European Centre for Disease Prevention and Control, Stockholm, Sweden
- European Centre for Disease Prevention and Control Public Health Emergency team co-authors
| | - Tonino Alonzi
- Translational Research Unit, Epidemiology and Preclinical Research Dept, "L. Spallanzani" National Institute for Infectious Diseases (INMI), IRCCS, Rome, Italy
| | - Sophia Archuleta
- Dept of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Sergio Brusin
- Public Health Emergency Team, European Centre for Disease Prevention and Control, Stockholm, Sweden
- European Centre for Disease Prevention and Control Public Health Emergency team co-authors
| | - Emmanuelle Cambau
- AP-HP-Lariboisiere, Bacteriologie, Laboratory Associated to the National Reference Centre for Mycobacteria, IAME UMR1137, INSERM, University of Paris, Paris, France
- Members of ESGMYC
| | - Maria Rosaria Capobianchi
- Laboratory of Virology, Epidemiology and Preclinical Research Dept, "L. Spallanzani" National Institute for Infectious Diseases (INMI), IRCCS, Rome, Italy
| | - Concetta Castilletti
- Laboratory of Virology, Epidemiology and Preclinical Research Dept, "L. Spallanzani" National Institute for Infectious Diseases (INMI), IRCCS, Rome, Italy
| | - Rosella Centis
- Servizio di Epidemiologia Clinica delle Malattie Respiratorie, Istituti Clinici Scientifici Maugeri IRCCS, Tradate, Italy
| | - Daniela M Cirillo
- Emerging Bacterial Pathogens Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Members of ESGMYC
| | | | - Giovanni Delogu
- Università Cattolica Sacro Cuore, Roma, Italy
- Mater Olbia Hospital, Olbia, Italy
- Members of ESGMYC
| | - Susanna M R Esposito
- Pediatric Clinic, Pietro Barilla Children's Hospital, University of Parma, Parma, Italy
| | | | - Jon S Friedland
- St George's, University of London, London, UK
- Members of ESGMYC
| | - Benjamin Choon Heng Ho
- Tuberculosis Control Unit, Dept of Respiratory and Critical Care Medicine, Tan Tock Seng Hospital, Singapore
| | - Giuseppe Ippolito
- Scientific Direction, "L. Spallanzani" National Institute for Infectious Diseases (INMI), IRCCS, Rome, Italy
| | - Mateja Jankovic
- School of Medicine, University of Zagreb and Clinic for Respiratory Diseases, University Hospital Center Zagreb, Zagreb, Croatia
- Members of ESGMYC
| | - Hannah Yejin Kim
- Sydney Pharmacy School, University of Sydney, Sydney, Australia
- Westmead Hospital, Sydney, Australia
- Marie Bashir Institute of Infectious Diseases and Biosecurity, University of Sydney, Sydney, Australia
| | - Senia Rosales Klintz
- Public Health Emergency Team, European Centre for Disease Prevention and Control, Stockholm, Sweden
- European Centre for Disease Prevention and Control Public Health Emergency team co-authors
| | - Csaba Ködmön
- Public Health Emergency Team, European Centre for Disease Prevention and Control, Stockholm, Sweden
- European Centre for Disease Prevention and Control Public Health Emergency team co-authors
| | - Eleonora Lalle
- Laboratory of Virology, Epidemiology and Preclinical Research Dept, "L. Spallanzani" National Institute for Infectious Diseases (INMI), IRCCS, Rome, Italy
| | - Yee Sin Leo
- National Centre for Infectious Diseases, Singapore
| | - Chi-Chiu Leung
- Hong Kong Tuberculosis, Chest and Heart Diseases Association, Wanchai, Hong Kong, China
| | - Anne-Grete Märtson
- Dept of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | | | - Saeid Najafi Fard
- Translational Research Unit, Epidemiology and Preclinical Research Dept, "L. Spallanzani" National Institute for Infectious Diseases (INMI), IRCCS, Rome, Italy
| | - Pasi Penttinen
- Public Health Emergency Team, European Centre for Disease Prevention and Control, Stockholm, Sweden
- European Centre for Disease Prevention and Control Public Health Emergency team co-authors
| | - Linda Petrone
- Translational Research Unit, Epidemiology and Preclinical Research Dept, "L. Spallanzani" National Institute for Infectious Diseases (INMI), IRCCS, Rome, Italy
| | - Elisa Petruccioli
- Translational Research Unit, Epidemiology and Preclinical Research Dept, "L. Spallanzani" National Institute for Infectious Diseases (INMI), IRCCS, Rome, Italy
| | | | - Laura Saderi
- Clinical Epidemiology and Medical Statistics Unit, Dept of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
| | - Miguel Santin
- Dept of Infectious Diseases, Bellvitge University Hospital-Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
- Dept of Clinical Science, University of Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain
- Members of ESGMYC
| | - Antonio Spanevello
- Division of Pulmonary Rehabilitation, Istituti Clinici Scientifici Maugeri, IRCCS, Tradate, Italy
- Dept of Medicine and Surgery, Respiratory Diseases, University of Insubria, Varese-Como, Italy
| | - Reinout van Crevel
- Radboudumc Center for Infectious Diseases, Radboud Institute for Health Sciences, Radboudumc, Nijmegen, The Netherlands
- Centre for Tropical Medicine and Global Health, Nuffield Dept of Medicine, University of Oxford, Oxford, UK
- Members of ESGMYC
| | - Marieke J van der Werf
- Public Health Emergency Team, European Centre for Disease Prevention and Control, Stockholm, Sweden
- European Centre for Disease Prevention and Control Public Health Emergency team co-authors
| | - Dina Visca
- Division of Pulmonary Rehabilitation, Istituti Clinici Scientifici Maugeri, IRCCS, Tradate, Italy
- Dept of Medicine and Surgery, Respiratory Diseases, University of Insubria, Varese-Como, Italy
| | - Miguel Viveiros
- Global Health and Tropical Medicine, Institute of Hygiene and Tropical Medicine, NOVA University of Lisbon, Lisbon, Portugal
- Members of ESGMYC
| | | | - Alimuddin Zumla
- Dept of Infection, Division of Infection and Immunity, University College London and NIHR Biomedical Research Centre, UCL Hospitals NHS Foundation Trust, London, UK
| | - Delia Goletti
- Translational Research Unit, Epidemiology and Preclinical Research Dept, "L. Spallanzani" National Institute for Infectious Diseases (INMI), IRCCS, Rome, Italy
- Saint Camillus International University of Health and Medical Sciences, Rome, Italy
- Members of ESGMYC
| |
Collapse
|
52
|
Förster R, Fleige H, Sutter G. Combating COVID-19: MVA Vector Vaccines Applied to the Respiratory Tract as Promising Approach Toward Protective Immunity in the Lung. Front Immunol 2020; 11:1959. [PMID: 32849655 PMCID: PMC7426738 DOI: 10.3389/fimmu.2020.01959] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 07/21/2020] [Indexed: 02/02/2023] Open
Abstract
The lung is the vital target organ of coronavirus disease 2019 (COVID-19) caused by infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). In the majority of patients the most active virus replication seems to be found in the upper respiratory tract, severe cases however suffer from SARS-like disease associated with virus replication in lung tissues. Due to the current lack of suitable anti-viral drugs the induction of protective immunity such as neutralizing antibodies in the lung is the key aim of the only alternative approach—the development and application of SARS-CoV-2 vaccines. However, past experience from experimental animals, livestock, and humans showed that induction of immunity in the lung is limited following application of vaccines at peripheral sides such as skin or muscles. Based on several considerations we therefore propose here to consider the application of a Modified Vaccinia virus Ankara (MVA)-based vaccine to mucosal surfaces of the respiratory tract as a favorable approach to combat COVID-19.
Collapse
Affiliation(s)
- Reinhold Förster
- Institute of Immunology, Hannover Medical School, Hanover, Germany.,Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hanover, Germany
| | - Henrike Fleige
- Institute of Immunology, Hannover Medical School, Hanover, Germany
| | - Gerd Sutter
- Division of Virology, Institute for Infectious Diseases and Zoonoses, Ludwig-Maximilians-Universität München, Munich, Germany.,German Center for Infection Research, Partner Site Munich, Munich, Germany
| |
Collapse
|
53
|
Yao C, Bora SA, Parimon T, Zaman T, Friedman OA, Palatinus JA, Surapaneni NS, Matusov YP, Chiang GC, Kassar AG, Patel N, Green CER, Aziz AW, Suri H, Suda J, Lopez AA, Martins GA, Stripp BR, Gharib SA, Goodridge HS, Chen P. Cell type-specific immune dysregulation in severely ill COVID-19 patients. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2020:2020.07.23.20161182. [PMID: 32743611 PMCID: PMC7386732 DOI: 10.1101/2020.07.23.20161182] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Coronavirus disease 2019 (COVID-19) has quickly become the most serious pandemic since the 1918 flu pandemic. In extreme situations, patients develop a dysregulated inflammatory lung injury called acute respiratory distress syndrome (ARDS) that causes progressive respiratory failure requiring mechanical ventilatory support. Recent studies have demonstrated immunologic dysfunction in severely ill COVID-19 patients. To further delineate the dysregulated immune response driving more severe clinical course from SARS-CoV-2 infection, we used single-cell RNA sequencing (scRNAseq) to analyze the transcriptome of peripheral blood mononuclear cells (PBMC) from hospitalized COVID-19 patients having mild disease (n = 5), developing ARDS (n = 6), and recovering from ARDS (n = 6). Our data demonstrated an overwhelming inflammatory response with select immunodeficiencies within various immune populations in ARDS patients. Specifically, their monocytes had defects in antigen presentation and deficiencies in interferon responsiveness that contrasted the higher interferon signals in lymphocytes. Furthermore, cytotoxic activity was suppressed in both NK and CD8 lymphocytes whereas B cell activation was deficient, which is consistent with the delayed viral clearance in severely ill COVID-19 patients. Finally, we identified altered signaling pathways in the severe group that suggests immunosenescence and immunometabolic changes could be contributing to the dysfunctional immune response. Our study demonstrates that COVID-19 patients with ARDS have an immunologically distinct response when compared to those with a more innocuous disease course and show a state of immune imbalance in which deficiencies in both the innate and adaptive immune response may be contributing to a more severe disease course in COVID-19.
Collapse
Affiliation(s)
- Changfu Yao
- Women’s Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Stephanie A Bora
- Women’s Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Tanyalak Parimon
- Women’s Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Tanzira Zaman
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Oren A Friedman
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Joseph A Palatinus
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Nirmala S Surapaneni
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Yuri P Matusov
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Giuliana Cerro Chiang
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Alexander G Kassar
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Nayan Patel
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Chelsi ER Green
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Adam W Aziz
- Women’s Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Harshpreet Suri
- Women’s Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Jo Suda
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Andres A Lopez
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Gislaine A Martins
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
- F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute (IBIRI), Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Division of Gastroenterology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Barry R Stripp
- Women’s Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Sina A Gharib
- Computational Medicine Core at Center for Lung Biology, Division of Pulmonary, Critical Care and Sleep Medicine, University of Washington, Seattle, WA, USA
| | - Helen S Goodridge
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Peter Chen
- Women’s Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| |
Collapse
|
54
|
Ma Q. Polarization of Immune Cells in the Pathologic Response to Inhaled Particulates. Front Immunol 2020; 11:1060. [PMID: 32625201 PMCID: PMC7311785 DOI: 10.3389/fimmu.2020.01060] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 05/01/2020] [Indexed: 12/30/2022] Open
Abstract
Polarization of immune cells is commonly observed in host responses associated with microbial immunity, inflammation, tumorigenesis, and tissue repair and fibrosis. In this process, immune cells adopt distinct programs and perform specialized functions in response to specific signals. Accumulating evidence indicates that inhalation of micro- and nano-sized particulates activates barrier immune programs in the lung in a time- and context-dependent manner, including type 1 and type 2 inflammation, and T helper (Th) 17 cell, regulatory T cell (Treg), innate lymphoid cell (ILC), and myeloid-derived suppressor cell (MDSC) responses, which highlight the polarization of several major immune cell types. These responses facilitate the pulmonary clearance and repair under physiological conditions. When exposure persists and overwhelms the clearance capacity, they foster the chronic progression of inflammation and development of progressive disease conditions, such as fibrosis and cancer. The pulmonary response to insoluble particulates thus represents a distinctive disease process wherein non-infectious, persistent exposures stimulate the polarization of immune cells to orchestrate dynamic inflammatory and immune reactions, leading to pulmonary and pleural chronic inflammation, fibrosis, and malignancy. Despite large variations in particles and their associated disease outcomes, the early response to inhaled particles often follows a common path. The initial reactions entail a barrier immune response dominated by type 1 inflammation that features active phagocytosis by M1 macrophages and recruitment of neutrophils, both of which are fueled by Th1 and proinflammatory cytokines. Acute inflammation is immediately followed by resolution and tissue repair mediated through specialized pro-resolving mediators (SPMs) and type 2 cytokines and cells including M2 macrophages and Th2 lymphocytes. As many particles and fibers cannot be digested by phagocytes, resolution is often extended and incomplete, and type 2 inflammation becomes heightened, which promotes interstitial fibrosis, granuloma formation, and tumorigenesis. Recent studies also reveal the involvement of Th17-, Treg-, ILC-, and MDSC-mediated responses in the pathogenesis caused by inhaled particulates. This review synopsizes the progress in understanding the interplay between inhaled particles and the pulmonary immune functions in disease pathogenesis, with focus on particle-induced polarization of immune cells and its role in the development of chronic inflammation, fibrosis, and cancer in the lung.
Collapse
Affiliation(s)
- Qiang Ma
- Receptor Biology Laboratory, Toxicology and Molecular Biology Branch, Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, Morgantown, WV, United States
| |
Collapse
|
55
|
Rossi GA, Pohunek P, Feleszko W, Ballarini S, Colin AA. Viral infections and wheezing-asthma inception in childhood: is there a role for immunomodulation by oral bacterial lysates? Clin Transl Allergy 2020; 10:17. [PMID: 32509272 PMCID: PMC7255835 DOI: 10.1186/s13601-020-00322-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 05/16/2020] [Indexed: 12/15/2022] Open
Abstract
Severe and recurrent infections of the respiratory tract in early childhood constitute major risk factors for the development of bronchial hyper-responsiveness and obstructive respiratory diseases in later life. In the first years of life, the vast majority of respiratory tract infections (RTI) leading to wheezing and asthma are of a viral origin and severity and recurrence are the consequence of a greater exposure to infectious agents in a period when the immune system is still relatively immature. Therefore, boosting the efficiency of the host immune response against viral infections seems to be a rational preventative approach. In the last decades it has been demonstrated that living in farm environments, i.e. early-life exposure to microbes, may reduce the risk of allergic and infectious disorders, increasing the immune response efficacy. These findings have suggested that treatment with bacterial lysates could promote a nonspecific immunomodulation useful in the prevention of recurrent RTIs and of wheezing inception and persistence. Experimental and clinical studies showing the reduction of RTI frequency and severity in childhood and elucidating the involved mechanisms can support this hypothesis.
Collapse
Affiliation(s)
- Giovanni A. Rossi
- Department of Pediatrics, Pulmonary and Allergy Disease Unit and Cystic Fibrosis Center, G. Gaslini University Hospital, Largo G. Gaslini, 4, 16148 Genoa, Italy
| | - Petr Pohunek
- Dept of Paediatrics, 2nd Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Wojciech Feleszko
- Department of Pediatric Pulmonology and Allergy, The Medical University of Warsaw, Warsaw, Poland
| | - Stefania Ballarini
- Medical Affairs Lead, Infectious Diseases, OM Pharma, a Vifor Pharma Company, Meyrin, Geneva, Switzerland
| | - Andrew A. Colin
- Division of Pediatric Pulmonology, Miller School of Medicine, University of Miami, Miami, FL USA
| |
Collapse
|
56
|
Raeven RHM, Rockx-Brouwer D, Kanojia G, van der Maas L, Bindels THE, Ten Have R, van Riet E, Metz B, Kersten GFA. Intranasal immunization with outer membrane vesicle pertussis vaccine confers broad protection through mucosal IgA and Th17 responses. Sci Rep 2020; 10:7396. [PMID: 32355188 PMCID: PMC7192948 DOI: 10.1038/s41598-020-63998-2] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 04/08/2020] [Indexed: 12/15/2022] Open
Abstract
A vaccine based on outer membrane vesicles of pertussis (omvPV) is protective in a mouse-challenge model and induces a broad antibody and mixed Th1/Th2/Th17 response against multiple antigens following subcutaneous immunization. However, this route did not result in mucosal immunity and did not prevent nasopharyngeal colonization. In this study, we explored the potential of intranasal immunization with omvPV. Only intranasal immunization induced strong mucosal immune responses that encompasses enhanced pulmonary and nasal IgA antibody levels, mainly directed against Vag8 and LPS. Furthermore, high numbers of IgA- and IgG-producing plasma cells were detected as well as lung-resident IgA memory B-cells. Finally, only intranasal immunization induced pulmonary Th1/Th17-related cytokine responses. The magnitude and type of systemic immunity was comparable between both routes and included high systemic IgG antibody levels, strong IgG-producing plasma cell responses, memory B-cells residing in the spleen and systemic Th1/Th2/Th17-related cytokine responses. Importantly, only intranasal immunization prevented colonization in both the lungs and the nasal cavity. In conclusion, intranasal omvPV immunization induces mucosal IgA and Th17-mediated responses without influencing the systemic immunity profile. These responses resulted in prevention of Bordetella pertussis colonization in the respiratory tract, including the nasal cavity, thereby potentially preventing transmission.
Collapse
Affiliation(s)
- René H M Raeven
- Intravacc (Institute for Translational Vaccinology), Bilthoven, The Netherlands.
| | | | - Gaurav Kanojia
- Intravacc (Institute for Translational Vaccinology), Bilthoven, The Netherlands
| | | | - Tim H E Bindels
- Intravacc (Institute for Translational Vaccinology), Bilthoven, The Netherlands
| | - Rimko Ten Have
- Intravacc (Institute for Translational Vaccinology), Bilthoven, The Netherlands
| | - Elly van Riet
- Intravacc (Institute for Translational Vaccinology), Bilthoven, The Netherlands
| | - Bernard Metz
- Intravacc (Institute for Translational Vaccinology), Bilthoven, The Netherlands
| | - Gideon F A Kersten
- Intravacc (Institute for Translational Vaccinology), Bilthoven, The Netherlands
- Division of Drug Delivery Technology, Leiden Academic Center for Drug Research, Leiden University, Leiden, The Netherlands
| |
Collapse
|
57
|
Comprehensive T cell repertoire characterization of non-small cell lung cancer. Nat Commun 2020; 11:603. [PMID: 32001676 PMCID: PMC6992630 DOI: 10.1038/s41467-019-14273-0] [Citation(s) in RCA: 129] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 12/17/2019] [Indexed: 12/21/2022] Open
Abstract
Immunotherapy targeting T cells is increasingly utilized to treat solid tumors including non-small cell lung cancer (NSCLC). This requires a better understanding of the T cells in the lungs of patients with NSCLC. Here, we report T cell repertoire analysis in a cohort of 236 early-stage NSCLC patients. T cell repertoire attributes are associated with clinicopathologic features, mutational and immune landscape. A considerable proportion of the most prevalent T cells in tumors are also prevalent in the uninvolved tumor-adjacent lungs and appear specific to shared background mutations or viral infections. Patients with higher T cell repertoire homology between the tumor and uninvolved tumor-adjacent lung, suggesting a less tumor-focused T cell response, exhibit inferior survival. These findings indicate that a concise understanding of antigens and T cells in NSCLC is needed to improve therapeutic efficacy and reduce toxicity with immunotherapy, particularly adoptive T cell therapy. Relevant features of T cell repertoire in human cancer remain to be delineated. Here the authors show, by TCR sequencing in a large cohort of lung cancer patients, that while a majority of T cell clones are shared between tumor and adjacent lung tissue, less frequent tumor-unique T cell clones correlate with worse prognosis.
Collapse
|
58
|
Guvenel A, Jozwik A, Ascough S, Ung SK, Paterson S, Kalyan M, Gardener Z, Bergstrom E, Kar S, Habibi MS, Paras A, Zhu J, Park M, Dhariwal J, Almond M, Wong EH, Sykes A, Del Rosario J, Trujillo-Torralbo MB, Mallia P, Sidney J, Peters B, Kon OM, Sette A, Johnston SL, Openshaw PJ, Chiu C. Epitope-specific airway-resident CD4+ T cell dynamics during experimental human RSV infection. J Clin Invest 2020; 130:523-538. [PMID: 31815739 PMCID: PMC6934186 DOI: 10.1172/jci131696] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 10/08/2019] [Indexed: 01/27/2023] Open
Abstract
BACKGROUNDRespiratory syncytial virus (RSV) is an important cause of acute pulmonary disease and one of the last remaining major infections of childhood for which there is no vaccine. CD4+ T cells play a key role in antiviral immunity, but they have been little studied in the human lung.METHODSHealthy adult volunteers were inoculated i.n. with RSV A Memphis 37. CD4+ T cells in blood and the lower airway were analyzed by flow cytometry and immunohistochemistry. Bronchial soluble mediators were measured using quantitative PCR and MesoScale Discovery. Epitope mapping was performed by IFN-γ ELISpot screening, confirmed by in vitro MHC binding.RESULTSActivated CD4+ T cell frequencies in bronchoalveolar lavage correlated strongly with local C-X-C motif chemokine 10 levels. Thirty-nine epitopes were identified, predominantly toward the 3' end of the viral genome. Five novel MHC II tetramers were made using an immunodominant EFYQSTCSAVSKGYL (F-EFY) epitope restricted to HLA-DR4, -DR9, and -DR11 (combined allelic frequency: 15% in Europeans) and G-DDF restricted to HLA-DPA1*01:03/DPB1*02:01 and -DPA1*01:03/DPB1*04:01 (allelic frequency: 55%). Tetramer labeling revealed enrichment of resident memory CD4+ T (Trm) cells in the lower airway; these Trm cells displayed progressive differentiation, downregulation of costimulatory molecules, and elevated CXCR3 expression as infection evolved.CONCLUSIONSHuman infection challenge provides a unique opportunity to study the breadth of specificity and dynamics of RSV-specific T-cell responses in the target organ, allowing the precise investigation of Trm recognizing novel viral antigens over time. The new tools that we describe enable precise tracking of RSV-specific CD4+ cells, potentially accelerating the development of effective vaccines.TRIAL REGISTRATIONClinicalTrials.gov NCT02755948.FUNDINGMedical Research Council, Wellcome Trust, National Institute for Health Research.
Collapse
Affiliation(s)
| | | | - Stephanie Ascough
- Department of Infectious Disease, Imperial College London, London, United Kingdom
| | - Seng Kuong Ung
- Department of Infectious Disease, Imperial College London, London, United Kingdom
| | - Suzanna Paterson
- Department of Infectious Disease, Imperial College London, London, United Kingdom
| | - Mohini Kalyan
- Department of Infectious Disease, Imperial College London, London, United Kingdom
| | - Zoe Gardener
- Department of Infectious Disease, Imperial College London, London, United Kingdom
| | - Emma Bergstrom
- Department of Infectious Disease, Imperial College London, London, United Kingdom
| | - Satwik Kar
- Department of Infectious Disease, Imperial College London, London, United Kingdom
| | | | | | - Jie Zhu
- National Heart and Lung Institute and
| | | | | | | | | | | | | | | | | | - John Sidney
- Centre for Infectious Disease, Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, California, USA
| | - Bjoern Peters
- Centre for Infectious Disease, Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, California, USA
| | | | - Alessandro Sette
- Centre for Infectious Disease, Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, California, USA
- Department of Medicine, UCSD, La Jolla, California, USA
| | | | | | - Christopher Chiu
- Department of Infectious Disease, Imperial College London, London, United Kingdom
| |
Collapse
|
59
|
Warren KJ, Poole JA, Sweeter JM, DeVasure JM, Wyatt TA. An association between MMP-9 and impaired T cell migration in ethanol-fed BALB/c mice infected with respiratory syncytial virus-2A. Alcohol 2019; 80:25-32. [PMID: 30291948 DOI: 10.1016/j.alcohol.2018.09.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 09/14/2018] [Accepted: 09/26/2018] [Indexed: 12/20/2022]
Abstract
Matrix metalloproteinases are important for proper airway matrix structure and wound healing. These enzymes are also implicated in many airway diseases. Previously, chronic ethanol consumption was shown to prolong inflammation and delay viral clearance in respiratory syncytial virus (RSV)-infected mice. We hypothesize that alcohol alters anti-viral immunity by disrupting immune cell chemotaxis in the lung. BALB/c mice were randomly selected to consume 18% alcohol ad libitum for 8 weeks prior to infection with RSV-2A. Bronchoalveolar lavage (BAL) cell populations were measured by flow cytometry, and chemokines were detected by Western blot or ELISA. MMP-9 levels were determined by polymerase chain reaction (PCR) in mouse lungs and in BAL fluid by ELISA. T cells were acquired from the spleens of water-fed, non-infected control mice (CTRL); alcohol-fed, non-infected (ETOH); water-fed, RSV-infected (RSV); or ethanol-fed, RSV-infected (ETOH-RSV) 4 days after RSV infection. T cells were placed in a transmigration system where chemokines had been treated with and without activated MMP-9. Lymphocyte recruitment was significantly reduced in the BAL 4 days after RSV infection in ETOH-RSV mice, whereas chemokine levels were the highest in this group at all experimental time points examined in comparison to RSV (p < 0.05). MMP-9 mRNA and protein were detected at high levels in ETOH-RSV mice compared to RSV. Using ex vivo transmigration to CCL2 and CXCL10, T cell migration was not impaired between any of the treatment groups, yet when CCL2 and CXCL10 were treated with activated MMP-9, significantly fewer T cells migrated across collagen-coated 5-μm membranes (p < 0.05). Immune cell recruitment is necessary for viral clearance. We show that immune cells are decreased in the lungs of ETOH-RSV mice. In contrast to decreased cell recruitment, key inflammatory chemokines were elevated in the lungs of ETOH-RSV mice. These proteins may be prematurely degraded by MMP-9 in the lung, leading to defective immunity and reduced viral clearance.
Collapse
Affiliation(s)
- Kristi J Warren
- University of Nebraska Medical Center, Pulmonary, Critical Care, Sleep & Allergy, 985910 Nebraska Medical Center, Omaha, NE, 68198-5910, United States.
| | - Jill A Poole
- University of Nebraska Medical Center, Pulmonary, Critical Care, Sleep & Allergy, 985910 Nebraska Medical Center, Omaha, NE, 68198-5910, United States
| | - Jenea M Sweeter
- University of Nebraska Medical Center, Pulmonary, Critical Care, Sleep & Allergy, 985910 Nebraska Medical Center, Omaha, NE, 68198-5910, United States.
| | - Jane M DeVasure
- University of Nebraska Medical Center, Pulmonary, Critical Care, Sleep & Allergy, 985910 Nebraska Medical Center, Omaha, NE, 68198-5910, United States.
| | - Todd A Wyatt
- University of Nebraska Medical Center, Pulmonary, Critical Care, Sleep & Allergy, 985910 Nebraska Medical Center, Omaha, NE, 68198-5910, United States; Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, 68105, United States; University of Nebraska Medical Center, Department of Environmental, Agricultural, & Occupational Health, Omaha, NE, 68198, United States.
| |
Collapse
|
60
|
Abstract
Respiratory syncytial virus (RSV) can cause severe lower respiratory tract infections especially in infants, immunocompromised individuals and the elderly and is the most common cause of infant hospitalisation in the developed world. The immune responses against RSV are crucial for viral control and clearance but, if dysregulated, can also result in immunopathology and impaired gas exchange. Lung immunity to RSV and other respiratory viruses begins with the recruitment of immune cells from the bloodstream into the lungs. This inflammatory process is controlled largely by chemokines, which are small proteins that are produced in response to innate immune detection of the virus or the infection process. These chemokines serve as chemoattractants for granulocytes, monocytes, lymphocytes and other leukocytes. In this review, we highlight recent advances in the field of RSV infection and disease, focusing on how chemokines regulate virus-induced inflammation.
Collapse
Affiliation(s)
- Rinat Nuriev
- National Heart and Lung Institute, Imperial College London, London, UK.,I. Mechnikov Research Institute for Vaccines and Sera, Moscow, Russian Federation
| | - Cecilia Johansson
- National Heart and Lung Institute, Imperial College London, London, UK
| |
Collapse
|
61
|
Gounder AP, Boon ACM. Influenza Pathogenesis: The Effect of Host Factors on Severity of Disease. THE JOURNAL OF IMMUNOLOGY 2019; 202:341-350. [PMID: 30617115 DOI: 10.4049/jimmunol.1801010] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 09/10/2018] [Indexed: 12/11/2022]
Abstract
Influenza viruses continue to be a major global health threat. Severity and clinical outcome of influenza disease is determined by both viral and host factors. Viral factors have long been the subject of intense research and many molecular determinants have been identified. However, research into the host factors that protect or predispose to severe and fatal influenza A virus infections is lagging. The goal of this review is to highlight the recent insights into host determinants of influenza pathogenesis.
Collapse
Affiliation(s)
- Anshu P Gounder
- Department of Internal Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO 63110.,Department of Molecular Microbiology and Microbial Pathogenesis, Washington University School of Medicine in St. Louis, St. Louis, MO 63110; and
| | - Adrianus C M Boon
- Department of Internal Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO 63110; .,Department of Molecular Microbiology and Microbial Pathogenesis, Washington University School of Medicine in St. Louis, St. Louis, MO 63110; and.,Department of Pathology and Immunology, Washington University School of Medicine in St. Louis, St. Louis, MO 63110
| |
Collapse
|
62
|
Host Single Nucleotide Polymorphisms Modulating Influenza A Virus Disease in Humans. Pathogens 2019; 8:pathogens8040168. [PMID: 31574965 PMCID: PMC6963926 DOI: 10.3390/pathogens8040168] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 09/27/2019] [Accepted: 09/28/2019] [Indexed: 12/14/2022] Open
Abstract
A large number of human genes associated with viral infections contain single nucleotide polymorphisms (SNPs), which represent a genetic variation caused by the change of a single nucleotide in the DNA sequence. SNPs are located in coding or non-coding genomic regions and can affect gene expression or protein function by different mechanisms. Furthermore, they have been linked to multiple human diseases, highlighting their medical relevance. Therefore, the identification and analysis of this kind of polymorphisms in the human genome has gained high importance in the research community, and an increasing number of studies have been published during the last years. As a consequence of this exhaustive exploration, an association between the presence of some specific SNPs and the susceptibility or severity of many infectious diseases in some risk population groups has been found. In this review, we discuss the relevance of SNPs that are important to understand the pathology derived from influenza A virus (IAV) infections in humans and the susceptibility of some individuals to suffer more severe symptoms. We also discuss the importance of SNPs for IAV vaccine effectiveness.
Collapse
|
63
|
Clohisey S, Baillie JK. Host susceptibility to severe influenza A virus infection. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2019; 23:303. [PMID: 31488196 PMCID: PMC6729070 DOI: 10.1186/s13054-019-2566-7] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 08/13/2019] [Indexed: 11/30/2022]
Abstract
Most people exposed to a new flu virus do not notice any symptoms. A small minority develops critical illness. Some of this extremely broad variation in susceptibility is explained by the size of the initial inoculum or the influenza exposure history of the individual; some is explained by generic host factors, such as frailty, that decrease resilience following any systemic insult. Some demographic factors (pregnancy, obesity, and advanced age) appear to confer a more specific susceptibility to severe illness following infection with influenza viruses. As with other infectious diseases, a substantial component of susceptibility is determined by host genetics. Several genetic susceptibility variants have now been reported with varying levels of evidence. Susceptible hosts may have impaired intracellular controls of viral replication (e.g. IFITM3, TMPRS22 variants), defective interferon responses (e.g. GLDC, IRF7/9 variants), or defects in cell-mediated immunity with increased baseline levels of systemic inflammation (obesity, pregnancy, advanced age). These mechanisms may explain the prolonged viral replication reported in critically ill patients with influenza: patients with life-threatening disease are, by definition, abnormal hosts. Understanding these molecular mechanisms of susceptibility may in the future enable the design of host-directed therapies to promote resilience.
Collapse
Affiliation(s)
- Sara Clohisey
- Division of Genetics and Genomics, Roslin Institute, University of Edinburgh, Easter Bush, Edinburgh, EH25 9RG, UK
| | - John Kenneth Baillie
- Division of Genetics and Genomics, Roslin Institute, University of Edinburgh, Easter Bush, Edinburgh, EH25 9RG, UK. .,Intensive Care Unit, Royal Infirmary of Edinburgh, 54 Little France Drive, Edinburgh, EH16 5SA, UK.
| |
Collapse
|
64
|
Feng J, Hu X, Fu M, Dai L, Yu Y, Luo W, Zhao Z, Lu Z, Du Z, Zhou D, Wen B, Jiao J, Xiong X. Enhanced protection against Q fever in BALB/c mice elicited by immunization of chloroform-methanol residue of Coxiella burnetii via intratracheal inoculation. Vaccine 2019; 37:6076-6084. [PMID: 31477436 DOI: 10.1016/j.vaccine.2019.08.041] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 08/07/2019] [Accepted: 08/19/2019] [Indexed: 12/11/2022]
Abstract
Human Q fever is recognized as a worldwide public health problem. It often occurs by inhalation of airborne aerosols contaminated with Coxiella burnetii, a gram-negative intracellular bacterium, mainly from domestic livestock. In this study, we analyzed the possibility to establish mucosal and systemic immunity against C. burnetii infection using a pulmonary delivery of chloroform-methanol residue of C. burnetii (CMR) vaccine. Mice were immunized by the intratracheal inoculation of CMR (IT-CMR) or the subcutaneous injection of CMR (SC-CMR), and the immunized mice were challenged with C. burnetii by the intratracheal route. The levels of IFN-γ, IL-12p70, IL-5, and IL-4 in the IT-CMR group in splenic T cells stimulated ex vivo were significantly higher than in the SC-CMR group. Significantly elevated sIgA to C. burnetii was detected in the bronchoalveolar lavage fluid of mice immunized by IT-CMR but not by SC-CMR, which might have contributed to the significant reduction in C. burnetii load and pathological lesions in the lungs of the mice after the challenge of C. burnetii. These results suggest that compared with SC-CMR in mice, IT-CMR was more efficient to elicit cellular and lung mucosal immune responses against aerosol infection of C. burnetii.
Collapse
Affiliation(s)
- Junxia Feng
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, 20(#) Dong-Dia-Jie Street, Fengtai, Beijing 100071, China
| | - Xueyuan Hu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, 20(#) Dong-Dia-Jie Street, Fengtai, Beijing 100071, China
| | - Mengjiao Fu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, 20(#) Dong-Dia-Jie Street, Fengtai, Beijing 100071, China
| | - Lupeng Dai
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, 20(#) Dong-Dia-Jie Street, Fengtai, Beijing 100071, China
| | - Yonghui Yu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, 20(#) Dong-Dia-Jie Street, Fengtai, Beijing 100071, China
| | - Wenbo Luo
- Anhui Medical University, Mei-Shan Road, Hefei, Anhui 230032, China
| | - Zengming Zhao
- Center for Disease Control and Prevention of Chinese People's Liberation Army, 20(#) Dong-Dia-Jie Street, Fengtai, Beijing 100071, China
| | - Zhiyu Lu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, 20(#) Dong-Dia-Jie Street, Fengtai, Beijing 100071, China
| | - Zongmin Du
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, 20(#) Dong-Dia-Jie Street, Fengtai, Beijing 100071, China
| | - Dongsheng Zhou
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, 20(#) Dong-Dia-Jie Street, Fengtai, Beijing 100071, China
| | - Bohai Wen
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, 20(#) Dong-Dia-Jie Street, Fengtai, Beijing 100071, China
| | - Jun Jiao
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, 20(#) Dong-Dia-Jie Street, Fengtai, Beijing 100071, China.
| | - Xiaolu Xiong
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, 20(#) Dong-Dia-Jie Street, Fengtai, Beijing 100071, China.
| |
Collapse
|
65
|
Graham BS. Immunological goals for respiratory syncytial virus vaccine development. Curr Opin Immunol 2019; 59:57-64. [PMID: 31029910 DOI: 10.1016/j.coi.2019.03.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 03/20/2019] [Accepted: 03/20/2019] [Indexed: 12/26/2022]
Abstract
Defining the immunological goals for respiratory syncytial virus (RSV) vaccination requires understanding of RSV biology and tropism, mechanisms of cell-to-cell spread and immunity, epidemiology, and transmission dynamics. The immunological goals for a particular vaccine would be product-specific based on antigen selection, delivery approach, and target population. There are many ways to achieve immunity against RSV infection involving innate and adaptive responses, humoral, and cellular effector mechanisms, and mucosal and systemic responses. Both protective and pathological immune response patterns have been demonstrated in animal models and humans. In this short commentary, the entire information matrix that may inform the design of particular vaccine candidates cannot be fully reviewed, but the rationale behind the major vaccine approaches in key target populations will be discussed.
Collapse
Affiliation(s)
- Barney S Graham
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892, USA.
| |
Collapse
|
66
|
Nawroth JC, Barrile R, Conegliano D, van Riet S, Hiemstra PS, Villenave R. Stem cell-based Lung-on-Chips: The best of both worlds? Adv Drug Deliv Rev 2019; 140:12-32. [PMID: 30009883 PMCID: PMC7172977 DOI: 10.1016/j.addr.2018.07.005] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 06/06/2018] [Accepted: 07/06/2018] [Indexed: 02/07/2023]
Abstract
Pathologies of the respiratory system such as lung infections, chronic inflammatory lung diseases, and lung cancer are among the leading causes of morbidity and mortality, killing one in six people worldwide. Development of more effective treatments is hindered by the lack of preclinical models of the human lung that can capture the disease complexity, highly heterogeneous disease phenotypes, and pharmacokinetics and pharmacodynamics observed in patients. The merger of two novel technologies, Organs-on-Chips and human stem cell engineering, has the potential to deliver such urgently needed models. Organs-on-Chips, which are microengineered bioinspired tissue systems, recapitulate the mechanochemical environment and physiological functions of human organs while concurrent advances in generating and differentiating human stem cells promise a renewable supply of patient-specific cells for personalized and precision medicine. Here, we discuss the challenges of modeling human lung pathophysiology in vitro, evaluate past and current models including Organs-on-Chips, review the current status of lung tissue modeling using human pluripotent stem cells, explore in depth how stem-cell based Lung-on-Chips may advance disease modeling and drug testing, and summarize practical consideration for the design of Lung-on-Chips for academic and industry applications.
Collapse
Affiliation(s)
| | | | | | - Sander van Riet
- Department of Pulmonology, Leiden University Medical Center, PO Box 9600, 2300 RC, Leiden, the Netherlands
| | - Pieter S Hiemstra
- Department of Pulmonology, Leiden University Medical Center, PO Box 9600, 2300 RC, Leiden, the Netherlands
| | | |
Collapse
|
67
|
Abstract
Pneumonia is a type of acute lower respiratory infection that is common and severe. The outcome of lower respiratory infection is determined by the degrees to which immunity is protective and inflammation is damaging. Intercellular and interorgan signaling networks coordinate these actions to fight infection and protect the tissue. Cells residing in the lung initiate and steer these responses, with additional immunity effectors recruited from the bloodstream. Responses of extrapulmonary tissues, including the liver, bone marrow, and others, are essential to resistance and resilience. Responses in the lung and extrapulmonary organs can also be counterproductive and drive acute and chronic comorbidities after respiratory infection. This review discusses cell-specific and organ-specific roles in the integrated physiological response to acute lung infection, and the mechanisms by which intercellular and interorgan signaling contribute to host defense and healthy respiratory physiology or to acute lung injury, chronic pulmonary disease, and adverse extrapulmonary sequelae. Pneumonia should no longer be perceived as simply an acute infection of the lung. Pneumonia susceptibility reflects ongoing and poorly understood chronic conditions, and pneumonia results in diverse and often persistent deleterious consequences for multiple physiological systems.
Collapse
Affiliation(s)
- Lee J Quinton
- Pulmonary Center, Boston University School of Medicine , Boston, Massachusetts
| | - Allan J Walkey
- Pulmonary Center, Boston University School of Medicine , Boston, Massachusetts
| | - Joseph P Mizgerd
- Pulmonary Center, Boston University School of Medicine , Boston, Massachusetts
| |
Collapse
|
68
|
|
69
|
Casals C, Campanero-Rhodes MA, García-Fojeda B, Solís D. The Role of Collectins and Galectins in Lung Innate Immune Defense. Front Immunol 2018; 9:1998. [PMID: 30233589 PMCID: PMC6131309 DOI: 10.3389/fimmu.2018.01998] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2018] [Accepted: 08/14/2018] [Indexed: 12/16/2022] Open
Abstract
Different families of endogenous lectins use complementary defense strategies against pathogens. They may recognize non-self glycans typically found on pathogens and/or host glycans. The collectin and galectin families are prominent examples of these two lectin categories. Collectins are C-type lectins that contain a carbohydrate recognition domain and a collagen-like domain. Members of this group include surfactant protein A (SP-A) and D (SP-D), secreted by the alveolar epithelium to the alveolar fluid. Lung collectins bind to several microorganisms, which results in pathogen aggregation and/or killing, and enhances phagocytosis of pathogens by alveolar macrophages. Moreover, SP-A and SP-D influence macrophage responses, contributing to resolution of inflammation, and SP-A is essential for tissue-repair functions of macrophages. Galectins also function by interacting directly with pathogens or by modulating the immune system in response to the infection. Direct binding may result in enhanced or impaired infection of target cells, or can have microbicidal effects. Immunomodulatory effects of galectins include recruitment of immune cells to the site of infection, promotion of neutrophil function, and stimulation of the bactericidal activity of infected macrophages. Moreover, intracellular galectins can serve as danger receptors, promoting autophagy of the invading pathogen. This review will focus on the role of collectins and galectins in pathogen clearance and immune response activation in infectious diseases of the respiratory system.
Collapse
Affiliation(s)
- Cristina Casals
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain.,Departamento de Bioquímica y Biología Molecular, Universidad Complutense de Madrid, Madrid, Spain
| | - María A Campanero-Rhodes
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain.,Instituto de Química Física Rocasolano, CSIC, Madrid, Spain
| | - Belén García-Fojeda
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain.,Departamento de Bioquímica y Biología Molecular, Universidad Complutense de Madrid, Madrid, Spain
| | - Dolores Solís
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain.,Instituto de Química Física Rocasolano, CSIC, Madrid, Spain
| |
Collapse
|
70
|
Mansoor KA, Qadan F, Schmidt M, Qinna NA, Badr M, Matalka KZ. A Functional Food Mixture "Protector" Reinforces the Protective Immune Parameters against Viral Flu Infection in Mice. Nutrients 2018; 10:nu10060743. [PMID: 29890620 PMCID: PMC6024812 DOI: 10.3390/nu10060743] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 05/29/2018] [Accepted: 06/06/2018] [Indexed: 11/24/2022] Open
Abstract
Background: Viral influenza infection causes serious health issues especially when an outbreak occurs. Although influenza virus vaccines are available and each year manufactures modify the vaccine depending on the expected mutated strain, it is still far from satisfactory, mainly in young children and older adults. Therefore, a product that can support and shape the immune system to protect against viral flu infections is highly essential. Methods: A functional food water-soluble mixture of pomegranate, red grape, dates, olive fruit, figs, and ginger extracts, termed herein “Protector”, was prepared and tested in stimulating/modulating the production of specific cytokines, and hemagglutinin inhibition (HAI) antibodies following viral flu vaccination in mice. Results: A single intraperitoneal or multiple oral administration for 1–7 days of “Protector” significantly increased the production of interferon (IFN)-γ and interleukin (IL)-12 in blood, spleen, and lungs of mice. When “Protector” was orally administered for one week following a single vaccine injection (primary immunization) or for two weeks (one week apart) following double vaccine injections (secondary immunization), mice significantly produced higher titers of HAI antibodies. This increase in HAI antibodies was associated with Pillow-inducing significant and different changes in vaccine-induced IFN-γ, IL-12, IL-6 and IL-22 following primary and secondary immunizations. Conclusions: “Protector” administration reinforces the protective immune parameters against viral flu infection. Therefore, after performing preclinical toxicology studies and ensuring its safety, “Protector” should be considered a potential product to be tested in clinical trials to conclude its efficacy in reducing the devastating effects of flu infection in humans and its outbreaks.
Collapse
Affiliation(s)
- Kenza A Mansoor
- Faculty of Pharmacy & Medical Sciences, University of Petra, Amman, Jordan.
| | - Fadi Qadan
- Herbresearch Germany, Wartbergweg 15, D-86874 Mattsies, Germany.
| | - Mathias Schmidt
- Herbresearch Germany, Wartbergweg 15, D-86874 Mattsies, Germany.
| | - Nidal A Qinna
- University of Petra Pharmaceutical Center (UPPC), Amman, Jordan.
| | - Mujtaba Badr
- University of Petra Pharmaceutical Center (UPPC), Amman, Jordan.
| | | |
Collapse
|
71
|
Mohn KGI, Zhou F. Clinical Expectations for Better Influenza Virus Vaccines-Perspectives from the Young Investigators' Point of View. Vaccines (Basel) 2018; 6:E32. [PMID: 29861454 PMCID: PMC6027204 DOI: 10.3390/vaccines6020032] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 05/23/2018] [Accepted: 05/23/2018] [Indexed: 01/20/2023] Open
Abstract
The influenza virus is one of a few viruses that is capable of rendering an otherwise healthy person acutly bedridden for several days. This impressive knock-out effect, without prodromal symptoms, challenges our immune system. The influenza virus undergoes continuous mutations, escaping our pre-existing immunity and causing epidemics, and its segmented genome is subject to reassortment, resulting in novel viruses with pandemic potential. The personal and socieoeconomic burden from influenza is high. Vaccination is the most cost-effective countermeasure, with several vaccines that are available. The current limitations in vaccine effectivness, combined with the need for yearly updating of vaccine strains, is a driving force for research into developing new and improved influenza vaccines. The lack of public concern about influenza severity, and misleading information concerning vaccine safety contribute to low vaccination coverage even in high-risk groups. The success of future influeza vaccines will depend on an increased public awarness of the disease, and hence, the need for vaccination-aided through improved rapid diagnositics. The vaccines must be safe and broadly acting, with new, measurable correlates of protection and robust post-marketing safety studies, to improve the confidence in influenza vaccines.
Collapse
Affiliation(s)
- Kristin G-I Mohn
- Influenza Centre, Department of Clinical Science, University of Bergen, Bergen 5021, Norway.
- Emergency Care clinic, Haukeland University Hospital, Bergen 5021, Norway.
| | - Fan Zhou
- Influenza Centre, Department of Clinical Science, University of Bergen, Bergen 5021, Norway.
- K.G. Jebsen Centre for Influenza Vaccine Research, Department of Clinical Science, University of Bergen, Bergen 5021, Norway.
| |
Collapse
|
72
|
Raeven RH, Brummelman J, Pennings JLA, van der Maas L, Helm K, Tilstra W, van der Ark A, Sloots A, van der Ley P, van Eden W, Jiskoot W, van Riet E, van Els CA, Kersten GF, Han WG, Metz B. Molecular and cellular signatures underlying superior immunity against Bordetella pertussis upon pulmonary vaccination. Mucosal Immunol 2018; 11:979-993. [PMID: 28930286 DOI: 10.1038/mi.2017.81] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Accepted: 08/15/2017] [Indexed: 02/04/2023]
Abstract
Mucosal immunity is often required for protection against respiratory pathogens but the underlying cellular and molecular mechanisms of induction remain poorly understood. Here, systems vaccinology was used to identify immune signatures after pulmonary or subcutaneous immunization of mice with pertussis outer membrane vesicles. Pulmonary immunization led to improved protection, exclusively induced mucosal immunoglobulin A (IgA) and T helper type 17 (Th17) responses, and in addition evoked elevated systemic immunoglobulin G (IgG) antibody levels, IgG-producing plasma cells, memory B cells, and Th17 cells. These adaptive responses were preceded by unique local expression of genes of the innate immune response related to Th17 (e.g., Rorc) and IgA responses (e.g., Pigr) in addition to local and systemic secretion of Th1/Th17-promoting cytokines. This comprehensive systems approach identifies the effect of the administration route on the development of mucosal immunity, its importance in protection against Bordetella pertussis, and reveals potential molecular correlates of vaccine immunity to this reemerging pathogen.
Collapse
Affiliation(s)
- R Hm Raeven
- Institute for Translational Vaccinology (Intravacc), Bilthoven, The Netherlands.,Division of Drug Delivery Technology, Cluster BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden, The Netherlands
| | - J Brummelman
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands.,Department of Infectious Diseases and Immunology, Utrecht University, Utrecht, The Netherlands
| | - J L A Pennings
- Centre for Health Protection, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - L van der Maas
- Institute for Translational Vaccinology (Intravacc), Bilthoven, The Netherlands
| | - K Helm
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - W Tilstra
- Institute for Translational Vaccinology (Intravacc), Bilthoven, The Netherlands
| | - A van der Ark
- Institute for Translational Vaccinology (Intravacc), Bilthoven, The Netherlands
| | - A Sloots
- Institute for Translational Vaccinology (Intravacc), Bilthoven, The Netherlands
| | - P van der Ley
- Institute for Translational Vaccinology (Intravacc), Bilthoven, The Netherlands
| | - W van Eden
- Department of Infectious Diseases and Immunology, Utrecht University, Utrecht, The Netherlands
| | - W Jiskoot
- Division of Drug Delivery Technology, Cluster BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden, The Netherlands
| | - E van Riet
- Institute for Translational Vaccinology (Intravacc), Bilthoven, The Netherlands
| | - C Acm van Els
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - G Fa Kersten
- Institute for Translational Vaccinology (Intravacc), Bilthoven, The Netherlands.,Division of Drug Delivery Technology, Cluster BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden, The Netherlands
| | - W Gh Han
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - B Metz
- Institute for Translational Vaccinology (Intravacc), Bilthoven, The Netherlands
| |
Collapse
|
73
|
Jia J, Conlon TM, Sarker RS, Taşdemir D, Smirnova NF, Srivastava B, Verleden SE, Güneş G, Wu X, Prehn C, Gao J, Heinzelmann K, Lintelmann J, Irmler M, Pfeiffer S, Schloter M, Zimmermann R, Hrabé de Angelis M, Beckers J, Adamski J, Bayram H, Eickelberg O, Yildirim AÖ. Cholesterol metabolism promotes B-cell positioning during immune pathogenesis of chronic obstructive pulmonary disease. EMBO Mol Med 2018; 10:e8349. [PMID: 29674392 PMCID: PMC5938615 DOI: 10.15252/emmm.201708349] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 03/08/2018] [Accepted: 03/14/2018] [Indexed: 12/30/2022] Open
Abstract
The development of chronic obstructive pulmonary disease (COPD) pathogenesis remains unclear, but emerging evidence supports a crucial role for inducible bronchus-associated lymphoid tissue (iBALT) in disease progression. Mechanisms underlying iBALT generation, particularly during chronic CS exposure, remain to be defined. Oxysterol metabolism of cholesterol is crucial to immune cell localization in secondary lymphoid tissue. Here, we demonstrate that oxysterols also critically regulate iBALT generation and the immune pathogenesis of COPD In both COPD patients and cigarette smoke (CS)-exposed mice, we identified significantly upregulated CH25H and CYP7B1 expression in airway epithelial cells, regulating CS-induced B-cell migration and iBALT formation. Mice deficient in CH25H or the oxysterol receptor EBI2 exhibited decreased iBALT and subsequent CS-induced emphysema. Further, inhibition of the oxysterol pathway using clotrimazole resolved iBALT formation and attenuated CS-induced emphysema in vivo therapeutically. Collectively, our studies are the first to mechanistically interrogate oxysterol-dependent iBALT formation in the pathogenesis of COPD, and identify a novel therapeutic target for the treatment of COPD and potentially other diseases driven by the generation of tertiary lymphoid organs.
Collapse
Affiliation(s)
- Jie Jia
- Comprehensive Pneumology Center (CPC), Institute of Lung Biology and Disease, Helmholtz Zentrum München, Munich, Germany
- Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Thomas M Conlon
- Comprehensive Pneumology Center (CPC), Institute of Lung Biology and Disease, Helmholtz Zentrum München, Munich, Germany
- Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Rim Sj Sarker
- Comprehensive Pneumology Center (CPC), Institute of Lung Biology and Disease, Helmholtz Zentrum München, Munich, Germany
- Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Demet Taşdemir
- Department of Chest Diseases, School of Medicine, University of Gaziantep, Gaziantep, Turkey
| | - Natalia F Smirnova
- Comprehensive Pneumology Center (CPC), Institute of Lung Biology and Disease, Helmholtz Zentrum München, Munich, Germany
- Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Barkha Srivastava
- Comprehensive Pneumology Center (CPC), Institute of Lung Biology and Disease, Helmholtz Zentrum München, Munich, Germany
- Member of the German Center for Lung Research (DZL), Munich, Germany
| | | | - Gizem Güneş
- Comprehensive Pneumology Center (CPC), Institute of Lung Biology and Disease, Helmholtz Zentrum München, Munich, Germany
- Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Xiao Wu
- Joint Mass Spectrometry Centre, Comprehensive Molecular Analytics, Helmholtz Zentrum München, Munich, Germany
| | - Cornelia Prehn
- Institute of Experimental Genetics, Genome Analysis Center, Helmholtz Zentrum München, Munich, Germany
- German Center for Diabetes Research (DZD), Munich, Germany
| | - Jiaqi Gao
- Comprehensive Pneumology Center (CPC), Institute of Lung Biology and Disease, Helmholtz Zentrum München, Munich, Germany
- Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Katharina Heinzelmann
- Comprehensive Pneumology Center (CPC), Institute of Lung Biology and Disease, Helmholtz Zentrum München, Munich, Germany
- Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Jutta Lintelmann
- Joint Mass Spectrometry Centre, Comprehensive Molecular Analytics, Helmholtz Zentrum München, Munich, Germany
| | - Martin Irmler
- Institute of Experimental Genetics, Helmholtz Zentrum München, Munich, Germany
| | - Stefan Pfeiffer
- Research Unit Comparative Microbiome Analysis, Helmholtz Zentrum München, Munich, Germany
| | - Michael Schloter
- Research Unit Comparative Microbiome Analysis, Helmholtz Zentrum München, Munich, Germany
| | - Ralf Zimmermann
- Joint Mass Spectrometry Centre, Comprehensive Molecular Analytics, Helmholtz Zentrum München, Munich, Germany
- University of Rostock, Rostock, Germany
| | - Martin Hrabé de Angelis
- German Center for Diabetes Research (DZD), Munich, Germany
- Institute of Experimental Genetics, Helmholtz Zentrum München, Munich, Germany
- Chair of Experimental Genetics, Technische Universität München, Freising-Weihenstephan, Germany
| | - Johannes Beckers
- German Center for Diabetes Research (DZD), Munich, Germany
- Institute of Experimental Genetics, Helmholtz Zentrum München, Munich, Germany
- Chair of Experimental Genetics, Technische Universität München, Freising-Weihenstephan, Germany
| | - Jerzy Adamski
- Institute of Experimental Genetics, Genome Analysis Center, Helmholtz Zentrum München, Munich, Germany
- German Center for Diabetes Research (DZD), Munich, Germany
- Chair of Experimental Genetics, Technische Universität München, Freising-Weihenstephan, Germany
| | - Hasan Bayram
- Department of Chest Diseases, School of Medicine, University of Gaziantep, Gaziantep, Turkey
- School of Medicine, Koç University, Istanbul, Turkey
| | - Oliver Eickelberg
- Comprehensive Pneumology Center (CPC), Institute of Lung Biology and Disease, Helmholtz Zentrum München, Munich, Germany
- Member of the German Center for Lung Research (DZL), Munich, Germany
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado, Denver, CO, USA
| | - Ali Önder Yildirim
- Comprehensive Pneumology Center (CPC), Institute of Lung Biology and Disease, Helmholtz Zentrum München, Munich, Germany
- Member of the German Center for Lung Research (DZL), Munich, Germany
| |
Collapse
|
74
|
Ascough S, Paterson S, Chiu C. Induction and Subversion of Human Protective Immunity: Contrasting Influenza and Respiratory Syncytial Virus. Front Immunol 2018; 9:323. [PMID: 29552008 PMCID: PMC5840263 DOI: 10.3389/fimmu.2018.00323] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Accepted: 02/06/2018] [Indexed: 12/15/2022] Open
Abstract
Respiratory syncytial virus (RSV) and influenza are among the most important causes of severe respiratory disease worldwide. Despite the clinical need, barriers to developing reliably effective vaccines against these viruses have remained firmly in place for decades. Overcoming these hurdles requires better understanding of human immunity and the strategies by which these pathogens evade it. Although superficially similar, the virology and host response to RSV and influenza are strikingly distinct. Influenza induces robust strain-specific immunity following natural infection, although protection by current vaccines is short-lived. In contrast, even strain-specific protection is incomplete after RSV and there are currently no licensed RSV vaccines. Although animal models have been critical for developing a fundamental understanding of antiviral immunity, extrapolating to human disease has been problematic. It is only with recent translational advances (such as controlled human infection models and high-dimensional technologies) that the mechanisms responsible for differences in protection against RSV compared to influenza have begun to be elucidated in the human context. Influenza infection elicits high-affinity IgA in the respiratory tract and virus-specific IgG, which correlates with protection. Long-lived influenza-specific T cells have also been shown to ameliorate disease. This robust immunity promotes rapid emergence of antigenic variants leading to immune escape. RSV differs markedly, as reinfection with similar strains occurs despite natural infection inducing high levels of antibody against conserved antigens. The immunomodulatory mechanisms of RSV are thus highly effective in inhibiting long-term protection, with disturbance of type I interferon signaling, antigen presentation and chemokine-induced inflammation possibly all contributing. These lead to widespread effects on adaptive immunity with impaired B cell memory and reduced T cell generation and functionality. Here, we discuss the differences in clinical outcome and immune response following influenza and RSV. Specifically, we focus on differences in their recognition by innate immunity; the strategies used by each virus to evade these early immune responses; and effects across the innate-adaptive interface that may prevent long-lived memory generation. Thus, by comparing these globally important pathogens, we highlight mechanisms by which optimal antiviral immunity may be better induced and discuss the potential for these insights to inform novel vaccines.
Collapse
Affiliation(s)
- Stephanie Ascough
- Section of Infectious Diseases and Immunity, Imperial College London, London, United Kingdom
| | - Suzanna Paterson
- Section of Infectious Diseases and Immunity, Imperial College London, London, United Kingdom
| | - Christopher Chiu
- Section of Infectious Diseases and Immunity, Imperial College London, London, United Kingdom
| |
Collapse
|
75
|
Wong SS, Oshansky CM, Guo XZJ, Ralston J, Wood T, Seeds R, Newbern C, Waite B, Reynolds G, Widdowson MA, Huang QS, Webby RJ, Thomas PG. Severe Influenza Is Characterized by Prolonged Immune Activation: Results From the SHIVERS Cohort Study. J Infect Dis 2018; 217:245-256. [PMID: 29112724 PMCID: PMC7335675 DOI: 10.1093/infdis/jix571] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 10/30/2017] [Indexed: 12/21/2022] Open
Abstract
Background The immunologic factors underlying severe influenza are poorly understood. To address this, we compared the immune responses of influenza-confirmed hospitalized individuals with severe acute respiratory illness (SARI) to those of nonhospitalized individuals with influenza-like illness (ILI). Methods Peripheral blood lymphocytes were collected from 27 patients with ILI and 27 with SARI, at time of enrollment and then 2 weeks later. Innate and adaptive cellular immune responses were assessed by flow cytometry, and serum cytokine levels were assessed by a bead-based assay. Results During the acute phase, SARI was associated with significantly reduced numbers of circulating myeloid dendritic cells, CD192+ monocytes, and influenza virus-specific CD8+ and CD4+ T cells as compared to ILI. By the convalescent phase, however, most SARI cases displayed continued immune activation characterized by increased numbers of CD16+ monocytes and proliferating, and influenza virus-specific, CD8+ T cells as compared to ILI cases. SARI was also associated with reduced amounts of cytokines that regulate T-cell responses (ie, interleukin 4, interleukin 13, interleukin 12, interleukin 10, and tumor necrosis factor β) and hematopoiesis (interleukin 3 and granulocyte-macrophage colony-stimulating factor) but increased amounts of a proinflammatory cytokine (tumor necrosis factor α), chemotactic cytokines (MDC, MCP-1, GRO, and fractalkine), and growth-promoting cytokines (PDGFBB/AA, VEGF, and EGF) as compared to ILI. Conclusions Severe influenza cases showed a delay in the peripheral immune activation that likely led prolonged inflammation, compared with mild influenza cases.
Collapse
Affiliation(s)
- Sook-San Wong
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis
| | | | - Xi-Zhi J Guo
- Department of Immunology, St. Jude Children's Research Hospital, Memphis
- Integrated Biomedical Sciences Program, University of Tennessee Health Science Center, Memphis
| | - Jacqui Ralston
- Institute for Environmental Science and Research, National Centre for Biosecurity and Infectious Disease-Wallaceville, Upper Hutt
| | - Timothy Wood
- Institute for Environmental Science and Research, National Centre for Biosecurity and Infectious Disease-Wallaceville, Upper Hutt
| | - Ruth Seeds
- Institute for Environmental Science and Research, National Centre for Biosecurity and Infectious Disease-Wallaceville, Upper Hutt
| | - Claire Newbern
- Institute for Environmental Science and Research, National Centre for Biosecurity and Infectious Disease-Wallaceville, Upper Hutt
| | - Ben Waite
- Institute for Environmental Science and Research, National Centre for Biosecurity and Infectious Disease-Wallaceville, Upper Hutt
| | - Gary Reynolds
- Immunisation Advisory Service, Department of Population Health, University of Auckland, New Zealand
| | - Marc-Alain Widdowson
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Q Sue Huang
- Institute for Environmental Science and Research, National Centre for Biosecurity and Infectious Disease-Wallaceville, Upper Hutt
| | - Richard J Webby
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis
| | - Paul G Thomas
- Department of Immunology, St. Jude Children's Research Hospital, Memphis
- Integrated Biomedical Sciences Program, University of Tennessee Health Science Center, Memphis
| |
Collapse
|
76
|
Sykes A, Gerhardt E, Tang L, Adderson EE. The Effectiveness of Trivalent Inactivated Influenza Vaccine in Children with Acute Leukemia. J Pediatr 2017; 191:218-224.e1. [PMID: 29173310 PMCID: PMC5726795 DOI: 10.1016/j.jpeds.2017.08.071] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 07/17/2017] [Accepted: 08/25/2017] [Indexed: 01/02/2023]
Abstract
OBJECTIVE The objective of this study was to determine the effectiveness of trivalent inactivated influenza vaccine (TIV) for the prevention of laboratory-confirmed influenza and influenza-like illnesses (ILI) among children and adolescents receiving therapy for acute leukemia. STUDY DESIGN A retrospective review of the demographic and clinical characteristics of 498 patients at a pediatric cancer center who received therapy for acute leukemia during 3 successive influenza seasons (2010-2011 through 2012-2013). RESULTS In 498 patient seasons with a known immunization history (median age, 6 years; range, 1-21), 354 patients (71.1%) were immunized with TIV and 98 (19.7%) received a booster dose of vaccine. Vaccinated and unvaccinated patients had generally similar demographic characteristics. There were no differences in the overall rates of influenza or ILI between vaccinated and unvaccinated patients overall, or in any individual season. There was no difference in the rates of influenza or ILI between patients who received 1 dose of vaccine and those who received 2 doses. Time to first influenza infection and time to first ILI in vaccinated and unvaccinated patients were not different. CONCLUSION TIV did not protect children and adolescents with acute leukemia against laboratory-confirmed influenza or ILI. Future prospective studies should assess TIV effectiveness in high-risk subpopulations and alternative strategies to prevent influenza should be considered in this population.
Collapse
Affiliation(s)
- April Sykes
- Department of Biostatistics, St. Jude Children's Research Hospital, Memphis, TN
| | - Elsie Gerhardt
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN
| | - Li Tang
- Department of Biostatistics, St. Jude Children's Research Hospital, Memphis, TN
| | - Elisabeth E Adderson
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Carmel, IN; Department of Pediatrics, University of Tennessee Health Sciences Center, Memphis, TN.
| |
Collapse
|
77
|
Saluzzo S, Gorki AD, Rana BMJ, Martins R, Scanlon S, Starkl P, Lakovits K, Hladik A, Korosec A, Sharif O, Warszawska JM, Jolin H, Mesteri I, McKenzie ANJ, Knapp S. First-Breath-Induced Type 2 Pathways Shape the Lung Immune Environment. Cell Rep 2017; 18:1893-1905. [PMID: 28228256 PMCID: PMC5329122 DOI: 10.1016/j.celrep.2017.01.071] [Citation(s) in RCA: 184] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 12/27/2016] [Accepted: 01/26/2017] [Indexed: 01/07/2023] Open
Abstract
From birth onward, the lungs are exposed to the external environment and therefore harbor a complex immunological milieu to protect this organ from damage and infection. We investigated the homeostatic role of the epithelium-derived alarmin interleukin-33 (IL-33) in newborn mice and discovered the immediate upregulation of IL-33 from the first day of life, closely followed by a wave of IL-13-producing type 2 innate lymphoid cells (ILC2s), which coincided with the appearance of alveolar macrophages (AMs) and their early polarization to an IL-13-dependent anti-inflammatory M2 phenotype. ILC2s contributed to lung quiescence in homeostasis by polarizing tissue resident AMs and induced an M2 phenotype in transplanted macrophage progenitors. ILC2s continued to maintain the M2 AM phenotype during adult life at the cost of a delayed response to Streptococcus pneumoniae infection in mice. These data highlight the homeostatic role of ILC2s in setting the activation threshold in the lung and underline their implications in anti-bacterial defenses. The first breath triggers IL-33 induction by AEC2 in lungs of newborn mice IL-33 promotes the perinatal expansion and activation of ST2-expressing ILC2s ILC2-derived IL-13 polarizes newborn’s AMs into an M2 phenotype This homeostatic type 2 pathway delays antibacterial effector responses
Collapse
Affiliation(s)
- Simona Saluzzo
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna 1090, Austria; Department of Medicine I, Laboratory of Infection Biology, Medical University of Vienna, Vienna 1090, Austria; MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | - Anna-Dorothea Gorki
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna 1090, Austria; Department of Medicine I, Laboratory of Infection Biology, Medical University of Vienna, Vienna 1090, Austria
| | - Batika M J Rana
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | - Rui Martins
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna 1090, Austria; Department of Medicine I, Laboratory of Infection Biology, Medical University of Vienna, Vienna 1090, Austria
| | - Seth Scanlon
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | - Philipp Starkl
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna 1090, Austria; Department of Medicine I, Laboratory of Infection Biology, Medical University of Vienna, Vienna 1090, Austria
| | - Karin Lakovits
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna 1090, Austria; Department of Medicine I, Laboratory of Infection Biology, Medical University of Vienna, Vienna 1090, Austria
| | - Anastasiya Hladik
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna 1090, Austria; Department of Medicine I, Laboratory of Infection Biology, Medical University of Vienna, Vienna 1090, Austria
| | - Ana Korosec
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna 1090, Austria; Department of Medicine I, Laboratory of Infection Biology, Medical University of Vienna, Vienna 1090, Austria
| | - Omar Sharif
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna 1090, Austria; Department of Medicine I, Laboratory of Infection Biology, Medical University of Vienna, Vienna 1090, Austria
| | - Joanna M Warszawska
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna 1090, Austria; Department of Medicine I, Laboratory of Infection Biology, Medical University of Vienna, Vienna 1090, Austria
| | - Helen Jolin
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | - Ildiko Mesteri
- Institute of Pathology Überlingen, Überlingen 88662, Germany
| | - Andrew N J McKenzie
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK.
| | - Sylvia Knapp
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna 1090, Austria; Department of Medicine I, Laboratory of Infection Biology, Medical University of Vienna, Vienna 1090, Austria.
| |
Collapse
|
78
|
Zheng K, Fan Y, Ji R, Ma S. Distinctive effects of pilose antler on mouse peripheral blood immune cell populations. FOOD AGR IMMUNOL 2017. [DOI: 10.1080/09540105.2017.1332011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Affiliation(s)
- Kexin Zheng
- College of Food Science and Technology, Shenyang Agricultural University, Shenyang, People’s Republic of China
| | - Yudan Fan
- College of Bioscience and Biotechnology, Shenyang Agricultural University, Shenyang, People’s Republic of China
| | - Ruiqin Ji
- College of Horticulture, Shenyang Agricultural University, Shenyang, People’s Republic of China
| | - Shiliang Ma
- College of Food Science and Technology, Shenyang Agricultural University, Shenyang, People’s Republic of China
- College of Bioscience and Biotechnology, Shenyang Agricultural University, Shenyang, People’s Republic of China
| |
Collapse
|
79
|
Humoral immune responses during asthma and influenza co-morbidity in mice. Immunobiology 2017; 222:1064-1073. [PMID: 28889999 DOI: 10.1016/j.imbio.2017.08.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Revised: 06/29/2017] [Accepted: 08/31/2017] [Indexed: 12/27/2022]
Abstract
Humoral immunity serve dual functions of direct pathogen neutralization and enhancement of leukocyte function. Antibody classes are determined by antigen triggers, and the resulting antibodies can contribute to disease pathogenesis and host defense. Although asthma and influenza are immunologically distinct diseases, since we have found that allergic asthma exacerbation promotes antiviral host responses to influenza A virus, we hypothesized that humoral immunity may contribute to allergic host protection during influenza. C57BL/6J mice sensitized and challenged with Aspergillus fumigatus (or not) were infected with pandemic influenza A/CA/04/2009 virus. Negative control groups included naïve mice, and mice with only 'asthma' or influenza. Concentrations of antibodies were quantified by ELISA, and in situ localization of IgA- and IgE-positive cells in the lungs was determined by immunohistochemistry. The number and phenotype of B cells in spleens and mediastinal lymph nodes were determined by flow cytometry at predetermined timepoints after virus infection until viral clearance. Mucosal and systemic antibodies remained elevated in mice with asthma and influenza with prominent production of IgE and IgA compared to influenza-only controls. B cell expansion was prominent in the mediastinal lymph nodes of allergic mice during influenza where most cells produced IgG1 and IgA. Although allergy-skewed B cell responses dominated in mice with allergic airways inflammation during influenza virus infection, virus-specific antibodies were also induced. Future studies are required to identify the mechanisms involved with B cell activation and function in allergic hosts facing respiratory viral infections.
Collapse
|
80
|
Pathogenicity and peramivir efficacy in immunocompromised murine models of influenza B virus infection. Sci Rep 2017; 7:7345. [PMID: 28779075 PMCID: PMC5544712 DOI: 10.1038/s41598-017-07433-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 06/28/2017] [Indexed: 02/06/2023] Open
Abstract
Influenza B viruses are important human pathogens that remain inadequately studied, largely because available animal models are poorly defined. Here, we developed an immunocompromised murine models for influenza B virus infection, which we subsequently used to study pathogenicity and to examine antiviral efficacy of the neuraminidase inhibitor peramivir. We studied three influenza B viruses that represent both the Yamagata (B/Massachusetts/2/2012 and B/Phuket/3073/2013) and Victoria (B/Brisbane/60/2008, BR/08) lineages. BR/08 was the most pathogenic in genetically modified immunocompromised mice [BALB scid and non-obese diabetic (NOD) scid strains] causing lethal infection without prior adaptation. The immunocompromised mice demonstrated prolonged virus shedding with modest induction of immune responses compared to BALB/c. Rather than severe virus burden, BR/08 virus-associated disease severity correlated with extensive virus spread and severe pulmonary pathology, stronger and persistent natural killer cell responses, and the extended induction of pro-inflammatory cytokines and chemokines. In contrast to a single-dose treatment (75 mg/kg/day), repeated doses of peramivir rescued BALB scid mice from lethal challenge with BR/08, but did not result in complete virus clearance. In summary, we have established immunocompromised murine models for influenza B virus infection that will facilitate evaluations of the efficacy of currently available and investigational anti-influenza drugs.
Collapse
|
81
|
Antiviral Lipopeptide-Cell Membrane Interaction Is Influenced by PEG Linker Length. Molecules 2017; 22:molecules22071190. [PMID: 28714870 PMCID: PMC5776016 DOI: 10.3390/molecules22071190] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 07/08/2017] [Accepted: 07/13/2017] [Indexed: 12/13/2022] Open
Abstract
A set of lipopeptides was recently reported for their broad-spectrum antiviral activity against viruses belonging to the Paramyxoviridae family, including human parainfluenza virus type 3 and Nipah virus. Among them, the peptide with a 24-unit PEG linker connecting it to a cholesterol moiety (VG-PEG24-Chol) was found to be the best membrane fusion inhibitory peptide. Here, we evaluated the interaction of the same set of peptides with biomembrane model systems and isolated human peripheral blood mononuclear cells (PBMC). VG-PEG24-Chol showed the highest insertion rate and it was among the peptides that induced a larger change on the surface pressure of cholesterol rich membranes. This peptide also displayed a high affinity towards PBMC membranes. These data provide new information about the dynamics of peptide-membrane interactions of a specific group of antiviral peptides, known for their potential as multipotent paramyxovirus antivirals.
Collapse
|
82
|
Pulmonary immunity to viruses. Clin Sci (Lond) 2017; 131:1737-1762. [PMID: 28667071 DOI: 10.1042/cs20160259] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 03/31/2017] [Accepted: 04/06/2017] [Indexed: 12/28/2022]
Abstract
Mucosal surfaces, such as the respiratory epithelium, are directly exposed to the external environment and therefore, are highly susceptible to viral infection. As a result, the respiratory tract has evolved a variety of innate and adaptive immune defenses in order to prevent viral infection or promote the rapid destruction of infected cells and facilitate the clearance of the infecting virus. Successful adaptive immune responses often lead to a functional state of immune memory, in which memory lymphocytes and circulating antibodies entirely prevent or lessen the severity of subsequent infections with the same virus. This is also the goal of vaccination, although it is difficult to vaccinate in a way that mimics respiratory infection. Consequently, some vaccines lead to robust systemic immune responses, but relatively poor mucosal immune responses that protect the respiratory tract. In addition, adaptive immunity is not without its drawbacks, as overly robust inflammatory responses may lead to lung damage and impair gas exchange or exacerbate other conditions, such as asthma or chronic obstructive pulmonary disease (COPD). Thus, immune responses to respiratory viral infections must be strong enough to eliminate infection, but also have mechanisms to limit damage and promote tissue repair in order to maintain pulmonary homeostasis. Here, we will discuss the components of the adaptive immune system that defend the host against respiratory viral infections.
Collapse
|
83
|
Rossi GA, Colin AA. Respiratory syncytial virus-Host interaction in the pathogenesis of bronchiolitis and its impact on respiratory morbidity in later life. Pediatr Allergy Immunol 2017; 28:320-331. [PMID: 28339145 DOI: 10.1111/pai.12716] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/13/2017] [Indexed: 02/06/2023]
Abstract
Respiratory syncytial virus (RSV) is the most common agent of severe airway disease in infants and young children. Large epidemiologic studies have demonstrated a clear relationship between RSV infection and subsequent recurrent wheezing and asthma into childhood, thought to be predominantly related to long-term changes in neuroimmune control of airway tone rather than to allergic sensitization. These changes appear to be governed by the severity of the first RSV infection in infancy which in term depends on viral characteristics and load, but perhaps as importantly, on the genetic susceptibility and on the constitutional characteristic of the host. A variety of viral and host factors and their interplay modify the efficiency of the response to infection, including viral replication and the magnitude of structural and functional damage to the respiratory structures, and ultimately the extent, severity, and duration of subsequent wheezing.
Collapse
Affiliation(s)
- Giovanni A Rossi
- Pulmonary and Allergy Disease Pediatric Unit and Cystic Fibrosis Center, Istituto Giannina Gaslini, Genoa, Italy
| | - Andrew A Colin
- Division of Pediatric Pulmonology, Miller School of Medicine, University of Miami, Miami, FL, USA
| |
Collapse
|
84
|
Lee ACY, To KKW, Zhu H, Chu H, Li C, Mak WWN, Zhang AJX, Yuen KY. Avian influenza virus A H7N9 infects multiple mononuclear cell types in peripheral blood and induces dysregulated cytokine responses and apoptosis in infected monocytes. J Gen Virol 2017; 98:922-934. [PMID: 28555541 DOI: 10.1099/jgv.0.000751] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Most patients with avian influenza A H7N9 virus (H7N9) infection suffer from severe illness, accompanied by dysregulated cytokine/chemokine response, delayed viral clearance and impaired neutralizing antibody response. Here, we evaluated the role of peripheral blood mononuclear cells (PBMCs) in the pathogenesis of H7N9 infection using an ex vivo infection model. H7N9 infected a significantly higher percentage of PBMCs (23.9 %) than those of avian influenza A H5N1 virus (H5N1) (12.3 %) and pandemic H1N1 virus (pH1N1) (5.5 %) (P<0.01). H7N9 infected significantly more B and T lymphocytes than H5N1. When compared with pH1N1, H7N9-infected PBMCs had significantly higher mRNA levels of proinflammatory cytokines and type I interferons (IFNs) at 6 h post-infection (p.i.), but significantly lower levels of IFN-γ and IP-10 at 12 h p.i. Among the PBMCs, CD14+ monocytes were most permissive to H7N9 infection. The percentage of infected CD14+ monocytes was significantly higher for H7N9 than that of pH1N1, but not significantly different from that of H5N1. H7N9-infected monocytes showed higher expression of MIP-1α, MIP-1β and RANTES than that of pH1N1 at 6 h p.i. H7N9- but not pH1N1-infected monocytes died rapidly via apoptosis. Furthermore, pH1N1- but not H7N9-infected monocytes showed increased expression of the monocyte activation and differentiation markers. Unlike pH1N1, H7N9 showed similar PBMC/monocyte cytokine/chemokine expression profile, monocyte cell death and expression of activation/differentiation markers to H5N1. Besides proinflammatory cytokine activation leading to a cytokine storm, impaired IFN-γ production, rapid monocytic death and lack of monocyte differentiation may affect the ability of H7N9-infected innate immune cells to recruit protective adaptive immunity.
Collapse
Affiliation(s)
- Andrew C Y Lee
- Department of Microbiology, The University of Hong Kong, Hong Kong, PR China
| | - Kelvin K W To
- Research Centre of Infection and Immunology, The University of Hong Kong, Hong Kong, PR China.,Department of Microbiology, The University of Hong Kong, Hong Kong, PR China.,State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Hong Kong, PR China.,Carol Yu Centre for Infection, The University of Hong Kong, Hong Kong, PR China
| | - Houshun Zhu
- Department of Medicine, The University of Hong Kong, Hong Kong, PR China
| | - Hin Chu
- Department of Microbiology, The University of Hong Kong, Hong Kong, PR China
| | - Can Li
- Department of Microbiology, The University of Hong Kong, Hong Kong, PR China
| | - Winger W N Mak
- Department of Microbiology, The University of Hong Kong, Hong Kong, PR China
| | - Anna J X Zhang
- Department of Microbiology, The University of Hong Kong, Hong Kong, PR China.,Research Centre of Infection and Immunology, The University of Hong Kong, Hong Kong, PR China.,State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Hong Kong, PR China
| | - Kwok-Yung Yuen
- Department of Microbiology, The University of Hong Kong, Hong Kong, PR China.,State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Hong Kong, PR China.,Carol Yu Centre for Infection, The University of Hong Kong, Hong Kong, PR China.,Research Centre of Infection and Immunology, The University of Hong Kong, Hong Kong, PR China
| |
Collapse
|
85
|
Graham BS. Vaccine development for respiratory syncytial virus. Curr Opin Virol 2017; 23:107-112. [PMID: 28525878 DOI: 10.1016/j.coviro.2017.03.012] [Citation(s) in RCA: 118] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 03/22/2017] [Indexed: 10/24/2022]
Abstract
Respiratory syncytial virus (RSV) is an important and ubiquitous respiratory pathogen for which no vaccine is available notwithstanding more than 50 years of effort. It causes the most severe disease at the extremes of age and in settings of immunodeficiency. Although RSV is susceptible to neutralizing antibody, it has evolved multiple mechanisms of immune evasion allowing it to repeatedly infect people despite relatively little genetic diversity. Recent breakthroughs in determining the structure and antigenic content of the fusion (F) glycoprotein in its metastable untriggered prefusion form (pre-F) and the stable rearranged postfusion form (post-F) have yielded vaccine strategies that can induce potent neutralizing antibody responses and effectively boost pre-existing neutralizing activity. In parallel, novel live-attenuated and chimeric virus vaccine candidates and other novel approaches to deliver vaccine antigens have been developed. These events and activities have aroused optimism and a robust pipeline of potential vaccine products that promise to provide a means to reduce the public health burden of RSV infection.
Collapse
Affiliation(s)
- Barney S Graham
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, United States.
| |
Collapse
|
86
|
Abstract
PURPOSE OF REVIEW Pneumonia is a common disease that becomes severe in a subset of patients, dependent on host biology including mechanisms of immune resistance and tissue resilience. This review emphasizes discoveries in pneumonia biology from 2016, highlighting questions and directions that are especially pressing or newly emerging. RECENT FINDINGS Novel cell-cell interactions mediating innate immune responses against microbes in the lung have been elucidated, between distinct leukocyte subtypes as well as between leukocytes and the structural cells of the lung. Adaptive immunity has received growing attention for determining the outcome of pneumonia, particularly the lung resident memory cells that arise from repeated prior respiratory infections and direct heterotypic recall responses. New tissue resilience components have been identified that contribute to anti-inflammatory, proresolution, tissue-protective, and reparative regeneration pathways in the infected lung. SUMMARY Recent findings will direct research into fundamental mechanisms of lung protection. Over the longer term, manipulating these pathways has implications for clinical practice, as strategies to bolster resistance and resilience have potential to ameliorate severe pneumonia.
Collapse
Affiliation(s)
- Joseph P. Mizgerd
- Professor of Medicine, Microbiology, and Biochemistry, Director, Pulmonary Center, Boston University School of Medicine, 72 E. Concord Street, Boston, MA 02118, Phone 617-638-5201, Fax 617-638-5227,
| |
Collapse
|
87
|
Openshaw PJ, Chiu C, Culley FJ, Johansson C. Protective and Harmful Immunity to RSV Infection. Annu Rev Immunol 2017; 35:501-532. [DOI: 10.1146/annurev-immunol-051116-052206] [Citation(s) in RCA: 136] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Peter J.M. Openshaw
- Respiratory Infections, National Heart and Lung Institute, Imperial College London, London W2 1PG, United Kingdom
| | - Chris Chiu
- Respiratory Infections, National Heart and Lung Institute, Imperial College London, London W2 1PG, United Kingdom
| | - Fiona J. Culley
- Respiratory Infections, National Heart and Lung Institute, Imperial College London, London W2 1PG, United Kingdom
| | - Cecilia Johansson
- Respiratory Infections, National Heart and Lung Institute, Imperial College London, London W2 1PG, United Kingdom
| |
Collapse
|
88
|
Enhanced immunogenicity following miR-155 incorporation into the influenza A virus genome. Virus Res 2017; 235:115-120. [PMID: 28392443 DOI: 10.1016/j.virusres.2017.04.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 03/28/2017] [Accepted: 04/02/2017] [Indexed: 12/14/2022]
Abstract
Influenza A vaccine efficacy in the elderly is generally poor and so identification of novel molecular adjuvants to improve immunogenicity is important to reduce the overall burden of disease. Short non-coding RNAs, known as microRNAs (miRNAs) are known to regulate gene expression and have the potential to influence immune responses. One such miRNA, miR-155, has been shown to modulate T and B cell development and function. We incorporated miR-155 into the influenza A virus (IAV) genome creating a self-adjuvanting 'live vaccine' with the ability to modify immunogenicity. Infection of mice with a recombinant influenza virus encoding miR-155 in the NS gene segment altered epitope-specific expansion of influenza-specific CD8+ T cells and induced significantly higher levels of neutralising antibody.
Collapse
|
89
|
Sevimli S, Knight FC, Gilchuk P, Joyce S, Wilson JT. Fatty Acid-Mimetic Micelles for Dual Delivery of Antigens and Imidazoquinoline Adjuvants. ACS Biomater Sci Eng 2017; 3:179-194. [PMID: 29046894 PMCID: PMC5642296 DOI: 10.1021/acsbiomaterials.6b00408] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Vaccine design has undergone a shift towards the use of purified protein subunit vaccines, which offer increased safety and greater control over antigen specificity, but at the expense of immunogenicity. Here we report the development of a new polymer-based vaccine delivery platform engineered to enhance immunity through the co-delivery of protein antigens and the Toll-like receptor 7 (TLR7) agonist imiquimod (IMQ). Owing to the preferential solubility of IMQ in fatty acids, a series of block copolymer micelles with a fatty acid-mimetic core comprising lauryl methacrylate (LMA) and methacrylic acid (MAA), and a poly(ethylene glycol) methyl ether methacrylate (PEGMA) corona decorated with pyridyl disulfide ethyl methacrylate (PDSM) moieties for antigen conjugation were synthesized via reversible addition-fragmentation chain transfer (RAFT) polymerization. Carriers composed of 50 mole% LMA (LMA50) demonstrated the highest IMQ loading (2.2 w/w%) and significantly enhanced the immunostimulatory capacity of IMQ to induce dendritic cell maturation and proinflammatory cytokine production. Conjugation of a model antigen, ovalbumin (OVA), to the corona of IMQ-loaded LMA50 micelles enhanced in vitro antigen uptake and cross-presentation on MHC class I (MHC-I). A single intranasal (IN) immunization of mice with carriers co-loaded with IMQ and OVA elicited significantly higher pulmonary and systemic CD8+ T cell responses and increased serum IgG titer relative to a soluble formulation of antigen and adjuvant. Collectively, these data demonstrate that rationally designed fatty acid-mimetic micelles enhance intracellular antigen and IMQ delivery and have potential as synthetic vectors for enhancing the immunogenicity of subunit vaccines.
Collapse
Affiliation(s)
- Sema Sevimli
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, 2400 Highland Avenue
| | - Frances C. Knight
- Department of Biomedical Engineering, Vanderbilt University, 2301 Vanderbilt Place
| | - Pavlo Gilchuk
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Medical Center North
- Department of Veterans Administration Tennessee Valley Healthcare System, 1310 24th Avenue South
| | - Sebastian Joyce
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Medical Center North
- Department of Veterans Administration Tennessee Valley Healthcare System, 1310 24th Avenue South
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, 2301 Vanderbilt Place, Nashville, TN 37235, USA
| | - John T. Wilson
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, 2400 Highland Avenue
- Department of Biomedical Engineering, Vanderbilt University, 2301 Vanderbilt Place
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, 2301 Vanderbilt Place, Nashville, TN 37235, USA
| |
Collapse
|
90
|
Dos Santos Pereira Andrade AC, Lima MT, Oliveira GP, Calixto RS, de Sales E Souza ÉL, da Glória de Souza D, de Almeida Leite CM, Ferreira JMS, Kroon EG, de Oliveira DB, Dos Santos Martins F, Abrahão JS. Daily ingestion of the probiotic Lactobacillus paracasei ST11 decreases Vaccinia virus dissemination and lethality in a mouse model. Benef Microbes 2017; 8:73-80. [PMID: 27873547 DOI: 10.3920/bm2016.0074] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Vaccinia virus (VACV) is an important pathogen. Although studies have shown relationships between probiotics and viruses, the effect of probiotics on VACV infection is unknown. Therefore, this work aims to investigate the probiotics effects on VACV infection. Mice were divided into four groups, two non-infected groups, one receiving the probiotic, the other one not receiving it, and two groups infected intranasally with VACV Western Reserve (VACV-WR) receiving or not receiving the probiotic. Viral titres in organs and cytokine production in the lungs were analysed. Lung samples were also subjected to histological analysis. The intake of probiotic results in reduction in viral spread with a significant decrease of VACV titer on lung, liver and brain of treated group. In addition,treatment with the probiotic results in attenuated mice lung inflammation showing fewer lesions on histological findings and decreased lethality in mice infected with VACV. The ingestion of Lactobacillus paracasei ST11 (LPST11) after VACV infection resulted in 2/9 animal lethality compared with 4/9 in the VACV group. This is the first study on probiotics and VACV interactions, providing not only information about this interaction, but also proposing a model for future studies involving probiotics and other poxvirus.
Collapse
Affiliation(s)
- A C Dos Santos Pereira Andrade
- 1 Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos 6627, 31270-901 Belo Horizonte, MG, Brazil
| | - M Teixeira Lima
- 1 Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos 6627, 31270-901 Belo Horizonte, MG, Brazil
| | - G Pereira Oliveira
- 1 Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos 6627, 31270-901 Belo Horizonte, MG, Brazil
| | - R Silva Calixto
- 1 Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos 6627, 31270-901 Belo Horizonte, MG, Brazil
| | - É Lorenna de Sales E Souza
- 1 Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos 6627, 31270-901 Belo Horizonte, MG, Brazil
| | - D da Glória de Souza
- 1 Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos 6627, 31270-901 Belo Horizonte, MG, Brazil
| | - C M de Almeida Leite
- 2 Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos 6627, 31270-901 Belo Horizonte, MG, Brazil
| | - J M Siqueira Ferreira
- 3 Laboratório de Microbiologia, Universidade Federal de São João del-Rei, Av. Sebastião Gonçalves Coelho 400, 35501-296 Divinópolis, MG, Brazil
| | - E G Kroon
- 1 Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos 6627, 31270-901 Belo Horizonte, MG, Brazil
| | - D Bretas de Oliveira
- 1 Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos 6627, 31270-901 Belo Horizonte, MG, Brazil
| | - F Dos Santos Martins
- 1 Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos 6627, 31270-901 Belo Horizonte, MG, Brazil
| | - J S Abrahão
- 1 Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos 6627, 31270-901 Belo Horizonte, MG, Brazil
| |
Collapse
|
91
|
Abstract
Respiratory syncytial virus (RSV) is a common cause of upper respiratory tract infection in children and adults. However, infection with this virus sometimes leads to severe lower respiratory disease and is the major cause of infant hospitalisations in the developed world. Several risk factors such as baby prematurity and congenital heart disease are known to predispose towards severe disease but previously healthy, full-term infants can also develop bronchiolitis and viral pneumonia during RSV infection. The causes of severe disease are not fully understood but may include dysregulation of the immune response to the virus, resulting in excessive recruitment and activation of innate and adaptive immune cells that can cause damage. This review highlights recent discoveries on the balancing act of immune-mediated virus clearance versus immunopathology during RSV infection.
Collapse
Affiliation(s)
- Cecilia Johansson
- Respiratory Infections Section, St Mary's campus, National Heart and Lung Institute, Imperial College London, London, W2 1PG, UK
| |
Collapse
|
92
|
Human newborn B cells mount an interferon-α/β receptor-dependent humoral response to respiratory syncytial virus. J Allergy Clin Immunol 2016; 139:1997-2000.e4. [PMID: 27931976 DOI: 10.1016/j.jaci.2016.10.032] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Revised: 09/13/2016] [Accepted: 10/06/2016] [Indexed: 01/23/2023]
|
93
|
Bellinghausen C, Rohde GGU, Savelkoul PHM, Wouters EFM, Stassen FRM. Viral-bacterial interactions in the respiratory tract. J Gen Virol 2016; 97:3089-3102. [PMID: 27902340 DOI: 10.1099/jgv.0.000627] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
In the respiratory tract, viruses and bacteria can interact on multiple levels. It is well known that respiratory viruses, particularly influenza viruses, increase the susceptibility to secondary bacterial infections. Numerous mechanisms, including compromised physical and immunological barriers, and changes in the microenvironment have hereby been shown to contribute to the development of secondary bacterial infections. In contrast, our understanding of how bacteria shape a response to subsequent viral infection is still limited. There is emerging evidence that persistent infection (or colonization) of the lower respiratory tract (LRT) with potential pathogenic bacteria, as observed in diseases like chronic obstructive pulmonary disease or cystic fibrosis, modulates subsequent viral infections by increasing viral entry receptors and modulating the inflammatory response. Moreover, recent studies suggest that even healthy lungs are not, as had long been assumed, sterile. The composition of the lung microbiome may thus modulate responses to viral infections. Here we summarize the current knowledge on the co-pathogenesis between viruses and bacteria in LRT infections.
Collapse
Affiliation(s)
- Carla Bellinghausen
- Department of Respiratory Medicine, NUTRIM - School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands.,Department of Medical Microbiology, NUTRIM - School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Gernot G U Rohde
- Department of Respiratory Medicine, NUTRIM - School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Paul H M Savelkoul
- Department of Medical Microbiology, NUTRIM - School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands.,Department of Medical Microbiology & Infection Control, VU University Medical Center, Amsterdam, The Netherlands
| | - Emiel F M Wouters
- Department of Respiratory Medicine, NUTRIM - School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Frank R M Stassen
- Department of Medical Microbiology, NUTRIM - School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands
| |
Collapse
|
94
|
Seasonal influenza vaccines and hurdles to mutual protection. Clin Microbiol Infect 2016; 22 Suppl 5:S113-S119. [PMID: 27568914 DOI: 10.1016/j.cmi.2016.03.021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 03/19/2016] [Indexed: 11/22/2022]
Abstract
While vaccines against seasonal influenza are available, major hurdles still exist that prevent their use having any impact on epidemic spread. Recent epidemiologic data question the appropriateness of traditional vaccination timing (prior to the winter season) in many parts of the world. Furthermore, vaccine uptake in most countries even in high-risk populations does not reach the 75% target recommended by the World Health Organization. Influenza viruses continually undergo antigenic variation, and both inactivated and live attenuated influenza vaccines confer only short-lived strain-specific immunity, so annual revaccination is required. Improving vaccine-induced immunity is therefore an important goal. A vaccine that could confer durable protection against emerging influenza strains could significantly reduce onward transmission. Therefore, further understanding of protective immunity against influenza (including broadly cross-protective immune mechanisms such as haemagglutinin stem-binding antibodies and T cells) offers the hope of vaccines that can confer the long-lived heterosubtypic immune responses required for mutual protection.
Collapse
|
95
|
Polverino F, Seys LJM, Bracke KR, Owen CA. B cells in chronic obstructive pulmonary disease: moving to center stage. Am J Physiol Lung Cell Mol Physiol 2016; 311:L687-L695. [PMID: 27542809 DOI: 10.1152/ajplung.00304.2016] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Accepted: 08/15/2016] [Indexed: 12/22/2022] Open
Abstract
Chronic inflammatory responses in the lungs contribute to the development and progression of chronic obstructive pulmonary disease (COPD). Although research studies focused initially on the contributions of the innate immune system to the pathogenesis of COPD, more recent studies have implicated adaptive immune responses in COPD. In particular, studies have demonstrated increases in B cell counts and increases in the number and size of B cell-rich lymphoid follicles in COPD lungs that correlate directly with COPD severity. There are also increases in lung levels of mediators that promote B cell maturation, activation, and survival in COPD patients. B cell products such as autoantibodies directed against lung cells, components of cells, and extracellular matrix proteins are also present in COPD lungs. These autoantibodies may contribute to lung inflammation and injury in COPD patients, in part, by forming immune complexes that activate complement components. Studies of B cell-deficient mice and human COPD patients have linked B cells most strongly to the emphysema phenotype. However, B cells have protective activities during acute exacerbations of COPD by promoting adaptive immune responses that contribute to host defense against pathogens. This review outlines the evidence that links B cells and B cell-rich lymphoid follicles to the pathogenesis of COPD and the mechanisms involved. It also reviews the potential and limitations of B cells as therapeutic targets to slow the progression of human COPD.
Collapse
Affiliation(s)
- Francesca Polverino
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts; COPD Program, Lovelace Respiratory Research Institute, Albuquerque, New Mexico; University of Parma, Parma, Italy; and
| | - Leen J M Seys
- Laboratory for Translational Research in Obstructive Pulmonary Diseases, Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Ken R Bracke
- Laboratory for Translational Research in Obstructive Pulmonary Diseases, Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Caroline A Owen
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts; COPD Program, Lovelace Respiratory Research Institute, Albuquerque, New Mexico;
| |
Collapse
|
96
|
Innate cell communication kick-starts pathogen-specific immunity. Nat Immunol 2016; 17:356-63. [PMID: 27002843 DOI: 10.1038/ni.3375] [Citation(s) in RCA: 175] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 12/10/2015] [Indexed: 12/13/2022]
Abstract
Innate cells are responsible for the rapid recognition of infection and mediate essential mechanisms of pathogen elimination, and also facilitate adaptive immune responses. We review here the numerous intricate interactions among innate cells that initiate protective immunity. The efficient eradication of pathogens depends on the coordinated actions of multiple cells, including innate cells and epithelial cells. Rather than acting as isolated effector cells, innate cells are in constant communication with other responding cells of the immune system, locally and distally. These interactions are critically important for the efficient control of primary infections as well for the development of 'trained' innate cells that facilitate the rapid elimination of homologous or heterologous infections.
Collapse
|
97
|
Shaikh SR, Fessler MB, Gowdy KM. Role for phospholipid acyl chains and cholesterol in pulmonary infections and inflammation. J Leukoc Biol 2016; 100:985-997. [PMID: 27286794 PMCID: PMC5069085 DOI: 10.1189/jlb.4vmr0316-103r] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 05/19/2016] [Indexed: 12/15/2022] Open
Abstract
Review on how complex mixtures of bioactive lipids and cholesterol may influence the pulmonary immune response during infection. Bacterial and viral respiratory tract infections result in millions of deaths worldwide and are currently the leading cause of death from infection. Acute inflammation is an essential element of host defense against infection, but can be damaging to the host when left unchecked. Effective host defense requires multiple lipid mediators, which collectively have proinflammatory and/or proresolving effects on the lung. During pulmonary infections, phospholipid acyl chains and cholesterol can be chemically and enzymatically oxidized, as well as truncated and modified, producing complex mixtures of bioactive lipids. We review recent evidence that phospholipids and cholesterol and their derivatives regulate pulmonary innate and adaptive immunity during infection. We first highlight data that oxidized phospholipids generated in the lung during infection stimulate pattern recognition receptors, such as TLRs and scavenger receptors, thereby amplifying the pulmonary inflammatory response. Next, we discuss evidence that oxidation of endogenous pools of cholesterol during pulmonary infections produces oxysterols that also modify the function of both innate and adaptive immune cells. Last, we conclude with data that n‐3 polyunsaturated fatty acids, both in the form of phospholipid acyl chains and through enzymatic processing into endogenous proresolving lipid mediators, aid in the resolution of lung inflammation through distinct mechanisms. Unraveling the complex mechanisms of induction and function of distinct classes of bioactive lipids, both native and modified, may hold promise for developing new therapeutic strategies for improving pulmonary outcomes in response to infection.
Collapse
Affiliation(s)
- Saame Raza Shaikh
- Department of Biochemistry and Molecular Biology, East Carolina Diabetes and Obesity Institute, East Carolina Heart Institute, Brody School of Medicine, East Carolina University (ECU), Greenville, North Carolina, USA
| | - Michael B Fessler
- Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health (NIEHS/NIH), Research Triangle Park, North Carolina, USA
| | - Kymberly M Gowdy
- Department of Pharmacology and Toxicology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, USA;
| |
Collapse
|
98
|
Sridhar S. Heterosubtypic T-Cell Immunity to Influenza in Humans: Challenges for Universal T-Cell Influenza Vaccines. Front Immunol 2016; 7:195. [PMID: 27242800 PMCID: PMC4871858 DOI: 10.3389/fimmu.2016.00195] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 05/03/2016] [Indexed: 11/25/2022] Open
Abstract
Influenza A virus (IAV) remains a significant global health issue causing annual epidemics, pandemics, and sporadic human infections with highly pathogenic avian or swine influenza viruses. Current inactivated and live vaccines are the mainstay of the public health response to influenza, although vaccine efficacy is lower against antigenically distinct viral strains. The first pandemic of the twenty-first century underlined the urgent need to develop new vaccines capable of protecting against a broad range of influenza strains. Such “universal” influenza vaccines are based on the idea of heterosubtypic immunity, wherein immune responses to epitopes conserved across IAV strains can confer protection against subsequent infection and disease. T-cells recognizing conserved antigens are a key contributor in reducing viral load and limiting disease severity during heterosubtypic infection in animal models. Recent studies undertaken during the 2009 H1N1 pandemic provided key insights into the role of cross-reactive T-cells in mediating heterosubtypic protection in humans. This review focuses on human influenza to discuss the epidemiological observations that underpin cross-protective immunity, the role of T-cells as key players in mediating heterosubtypic immunity including recent data from natural history cohort studies and the ongoing clinical development of T-cell-inducing universal influenza vaccines. The challenges and knowledge gaps for developing vaccines to generate long-lived protective T-cell responses is discussed.
Collapse
|
99
|
Xu Z, Takizawa F, Parra D, Gómez D, von Gersdorff Jørgensen L, LaPatra SE, Sunyer JO. Mucosal immunoglobulins at respiratory surfaces mark an ancient association that predates the emergence of tetrapods. Nat Commun 2016; 7:10728. [PMID: 26869478 PMCID: PMC4754351 DOI: 10.1038/ncomms10728] [Citation(s) in RCA: 146] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Accepted: 01/14/2016] [Indexed: 12/24/2022] Open
Abstract
Gas-exchange structures are critical for acquiring oxygen, but they also represent portals for pathogen entry. Local mucosal immunoglobulin responses against pathogens in specialized respiratory organs have only been described in tetrapods. Since fish gills are considered a mucosal surface, we hypothesized that a dedicated mucosal immunoglobulin response would be generated within its mucosa on microbial exposure. Supporting this hypothesis, here we demonstrate that following pathogen exposure, IgT+ B cells proliferate and generate pathogen-specific IgT within the gills of fish, thus providing the first example of locally induced immunoglobulin in the mucosa of a cold-blooded species. Moreover, we demonstrate that gill microbiota is predominantly coated with IgT, thus providing previously unappreciated evidence that the microbiota present at a respiratory surface of a vertebrate is recognized by a mucosal immunoglobulin. Our findings indicate that respiratory surfaces and mucosal immunoglobulins are part of an ancient association that predates the emergence of tetrapods. In teleost fish the gills perform—in addition to respiration—functions such as immune defence. Here the authors show that IgT, a teleost specific Ig previously shown to be involved in gut and skin mucosal immunity, is locally induced in the gill, where it plays a key role in immunity in rainbow trout.
Collapse
Affiliation(s)
- Zhen Xu
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, 413 Rosenthal building, 3800 Spruce Street, Philadelphia, Pennsylvania 19104, USA.,Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Fumio Takizawa
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, 413 Rosenthal building, 3800 Spruce Street, Philadelphia, Pennsylvania 19104, USA
| | - David Parra
- Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Barcelona 08193, Spain
| | - Daniela Gómez
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, 413 Rosenthal building, 3800 Spruce Street, Philadelphia, Pennsylvania 19104, USA
| | - Louise von Gersdorff Jørgensen
- Laboratory of Aquatic Pathobiology, Department of Veterinary Disease Biology, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiskberg DK-1870, Denmark
| | - Scott E LaPatra
- Research Division, Clear Springs Foods Inc., P O Box 712, Buhl, Idaho 83316, USA
| | - J Oriol Sunyer
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, 413 Rosenthal building, 3800 Spruce Street, Philadelphia, Pennsylvania 19104, USA
| |
Collapse
|
100
|
Duan S, Thomas PG. Balancing Immune Protection and Immune Pathology by CD8(+) T-Cell Responses to Influenza Infection. Front Immunol 2016; 7:25. [PMID: 26904022 PMCID: PMC4742794 DOI: 10.3389/fimmu.2016.00025] [Citation(s) in RCA: 108] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 01/18/2016] [Indexed: 02/05/2023] Open
Abstract
Influenza A virus (IAV) is a significant human pathogen causing annual epidemics and periodic pandemics. CD8+ cytotoxic T lymphocyte (CTL)-mediated immunity contributes to the clearance of virus-infected cells, and CTL immunity targeting the conserved internal proteins of IAVs is a key protection mechanism when neutralizing antibodies are absent during heterosubtypic IAV infection. However, CTL infiltration into the airways, its cytotoxicity, and the effects of produced proinflammatory cytokines can cause severe lung tissue injury, thereby contributing to immunopathology. Studies have discovered complicated and exquisite stimulatory and inhibitory mechanisms that regulate CTL magnitude and effector activities during IAV infection. Here, we review the state of knowledge on the roles of IAV-specific CTLs in immune protection and immunopathology during IAV infection in animal models, highlighting the key findings of various requirements and constraints regulating the balance of immune protection and pathology involved in CTL immunity. We also discuss the evidence of cross-reactive CTL immunity as a positive correlate of cross-subtype protection during secondary IAV infection in both animal and human studies. We argue that the effects of CTL immunity on protection and immunopathology depend on multiple layers of host and viral factors, including complex host mechanisms to regulate CTL magnitude and effector activity, the pathogenic nature of the IAV, the innate response milieu, and the host historical immune context of influenza infection. Future efforts are needed to further understand these key host and viral factors, especially to differentiate those that constrain optimally effective CTL antiviral immunity from those necessary to restrain CTL-mediated non-specific immunopathology in the various contexts of IAV infection, in order to develop better vaccination and therapeutic strategies for modifying protective CTL immunity.
Collapse
Affiliation(s)
- Susu Duan
- Department of Immunology, St. Jude Children's Research Hospital , Memphis, TN , USA
| | - Paul G Thomas
- Department of Immunology, St. Jude Children's Research Hospital , Memphis, TN , USA
| |
Collapse
|