101
|
Nguyen THO, Koutsakos M, van de Sandt CE, Crawford JC, Loh L, Sant S, Grzelak L, Allen EK, Brahm T, Clemens EB, Auladell M, Hensen L, Wang Z, Nüssing S, Jia X, Günther P, Wheatley AK, Kent SJ, Aban M, Deng YM, Laurie KL, Hurt AC, Gras S, Rossjohn J, Crowe J, Xu J, Jackson D, Brown LE, La Gruta N, Chen W, Doherty PC, Turner SJ, Kotsimbos TC, Thomas PG, Cheng AC, Kedzierska K. Immune cellular networks underlying recovery from influenza virus infection in acute hospitalized patients. Nat Commun 2021; 12:2691. [PMID: 33976217 PMCID: PMC8113517 DOI: 10.1038/s41467-021-23018-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 04/08/2021] [Indexed: 02/07/2023] Open
Abstract
How innate and adaptive immune responses work in concert to resolve influenza disease is yet to be fully investigated in one single study. Here, we utilize longitudinal samples from patients hospitalized with acute influenza to understand these immune responses. We report the dynamics of 18 important immune parameters, related to clinical, genetic and virological factors, in influenza patients across different severity levels. Influenza disease correlates with increases in IL-6/IL-8/MIP-1α/β cytokines and lower antibody responses. Robust activation of circulating T follicular helper cells correlates with peak antibody-secreting cells and influenza heamaglutinin-specific memory B-cell numbers, which phenotypically differs from vaccination-induced B-cell responses. Numbers of influenza-specific CD8+ or CD4+ T cells increase early in disease and retain an activated phenotype during patient recovery. We report the characterisation of immune cellular networks underlying recovery from influenza infection which are highly relevant to other infectious diseases.
Collapse
Affiliation(s)
- Thi H O Nguyen
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Parkville, VIC, Australia
| | - Marios Koutsakos
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Parkville, VIC, Australia
| | - Carolien E van de Sandt
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Parkville, VIC, Australia
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | | | - Liyen Loh
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Parkville, VIC, Australia
| | - Sneha Sant
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Parkville, VIC, Australia
| | - Ludivine Grzelak
- Biology Department, École Normale Supérieure Paris-Saclay, Université Paris-Saclay Cachan, Cachan, France
| | - Emma K Allen
- Department of Immunology, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Tim Brahm
- Department of Immunology, St Jude Children's Research Hospital, Memphis, TN, USA
| | - E Bridie Clemens
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Parkville, VIC, Australia
| | - Maria Auladell
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Parkville, VIC, Australia
| | - Luca Hensen
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Parkville, VIC, Australia
| | - Zhongfang Wang
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Parkville, VIC, Australia
| | - Simone Nüssing
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Parkville, VIC, Australia
| | - Xiaoxiao Jia
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Parkville, VIC, Australia
| | - Patrick Günther
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Parkville, VIC, Australia
| | - Adam K Wheatley
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Parkville, VIC, Australia
| | - Stephen J Kent
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Parkville, VIC, Australia
- Melbourne Sexual Health Centre and Department of Infectious Diseases, Alfred Hospital and Central Clinical School, Monash University, Melbourne, VIC, Australia
- ARC Centre for Excellence in Convergent Bio-Nano Science and Technology, University of Melbourne, Parkville, VIC, Australia
| | - Malet Aban
- World Health Organisation (WHO) Collaborating Centre for Reference and Research on Influenza, at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Yi-Mo Deng
- World Health Organisation (WHO) Collaborating Centre for Reference and Research on Influenza, at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Karen L Laurie
- World Health Organisation (WHO) Collaborating Centre for Reference and Research on Influenza, at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Aeron C Hurt
- World Health Organisation (WHO) Collaborating Centre for Reference and Research on Influenza, at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Stephanie Gras
- Infection and Immunity Program & Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
- Australian Research Council Centre of Excellence for Advanced Molecular Imaging, Monash University, Clayton, VIC, Australia
- Department of Biochemistry and Genetics, La Trobe Institute For Molecular Science, La Trobe University, Bundoora, VIC, Australia
| | - Jamie Rossjohn
- Infection and Immunity Program & Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
- Australian Research Council Centre of Excellence for Advanced Molecular Imaging, Monash University, Clayton, VIC, Australia
- Institute of Infection and Immunity, Cardiff University School of Medicine, Heath Park, Cardiff, UK
| | - Jane Crowe
- Deepdene Surgery, Deepdene, VIC, Australia
| | - Jianqing Xu
- Shanghai Public Health Clinical Centre and Institutes of Biomedical Sciences, Key Laboratory of Medical Molecular Virology of Ministry of Education/Health, Shanghai Medical College, Fudan University, Shanghai, China
| | - David Jackson
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Parkville, VIC, Australia
| | - Lorena E Brown
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Parkville, VIC, Australia
| | - Nicole La Gruta
- Infection and Immunity Program & Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Weisan Chen
- Department of Biochemistry and Genetics, La Trobe Institute For Molecular Science, La Trobe University, Bundoora, VIC, Australia
| | - Peter C Doherty
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Parkville, VIC, Australia
| | - Stephen J Turner
- Department of Microbiology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Tom C Kotsimbos
- Department of Allergy, Immunology and Respiratory Medicine, The Alfred Hospital, Melbourne, VIC, Australia
- Department of Medicine, Monash University, Central Clinical School, The Alfred Hospital, Melbourne, VIC, Australia
| | - Paul G Thomas
- Department of Immunology, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Allen C Cheng
- School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, Australia.
- Infection Prevention and Healthcare Epidemiology Unit, Alfred Health, Melbourne, VIC, Australia.
| | - Katherine Kedzierska
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Parkville, VIC, Australia.
| |
Collapse
|
102
|
Felt SA, Sun Y, Jozwik A, Paras A, Habibi MS, Nickle D, Anderson L, Achouri E, Feemster KA, Cárdenas AM, Turi KN, Chang M, Hartert TV, Sengupta S, Chiu C, López CB. Detection of respiratory syncytial virus defective genomes in nasal secretions is associated with distinct clinical outcomes. Nat Microbiol 2021; 6:672-681. [PMID: 33795879 PMCID: PMC9098209 DOI: 10.1038/s41564-021-00882-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 02/23/2021] [Indexed: 02/01/2023]
Abstract
Respiratory syncytial virus (RSV) causes respiratory illness in children, immunosuppressed individuals and the elderly. However, the viral factors influencing the clinical outcome of RSV infections remain poorly defined. Defective viral genomes (DVGs) can suppress virus replication by competing for viral proteins and by stimulating antiviral immunity. We studied the association between detection of DVGs of the copy-back type and disease severity in three RSV A-confirmed cohorts. In hospitalized children, detection of DVGs in respiratory samples at or around the time of admission associated strongly with more severe disease, higher viral load and a stronger pro-inflammatory response. Interestingly, in experimentally infected adults, the presence of DVGs in respiratory secretions differentially associated with RSV disease severity depending on when DVGs were detected. Detection of DVGs early after infection associated with low viral loads and mild disease, whereas detection of DVGs late after infection, especially if DVGs were present for prolonged periods, associated with high viral loads and severe disease. Taken together, we demonstrate that the kinetics of DVG accumulation and duration could predict clinical outcome of RSV A infection in humans, and thus could be used as a prognostic tool to identify patients at risk of worse clinical disease.
Collapse
Affiliation(s)
- Sébastien A. Felt
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Present address: Department of Molecular Microbiology and Center for Women Infectious Disease Research, Washington University School of Medicine, St Louis, MO, USA.,These authors contributed equally: Sébastien A. Felt, Yan Sun
| | - Yan Sun
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Present address: Department of Microbiology and Immunology, University of Rochester, Rochester, NY, USA.,These authors contributed equally: Sébastien A. Felt, Yan Sun
| | - Agnieszka Jozwik
- Department of Infectious Disease, Imperial College London, London, UK
| | - Allan Paras
- Department of Infectious Disease, Imperial College London, London, UK
| | | | | | - Larry Anderson
- Pediatric Infectious Disease, Emory University, Atlanta, GA, USA
| | - Emna Achouri
- Department of Molecular Microbiology and Center for Women Infectious Disease Research, Washington University School of Medicine, St Louis, MO, USA
| | - Kristen A. Feemster
- Division of Infectious Diseases, Children’s Hospital of Philadelphia, Philadelphia, PA, USA.,Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ana María Cárdenas
- Infectious Disease Diagnostics Laboratory, Children’s Hospital of Philadelphia, Philadelphia, PA, USA.,Division of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Present address: Becton, Dickinson and Company, Sparks, MD, USA
| | - Kedir N. Turi
- Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | - Tina V. Hartert
- Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Shaon Sengupta
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Division of Neonatology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Christopher Chiu
- Department of Infectious Disease, Imperial College London, London, UK
| | - Carolina B. López
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Present address: Department of Molecular Microbiology and Center for Women Infectious Disease Research, Washington University School of Medicine, St Louis, MO, USA
| |
Collapse
|
103
|
Camiolo MJ, Zhou X, Oriss TB, Yan Q, Gorry M, Horne W, Trudeau JB, Scholl K, Chen W, Kolls JK, Ray P, Weisel FJ, Weisel NM, Aghaeepour N, Nadeau K, Wenzel SE, Ray A. High-dimensional profiling clusters asthma severity by lymphoid and non-lymphoid status. Cell Rep 2021; 35:108974. [PMID: 33852838 PMCID: PMC8133874 DOI: 10.1016/j.celrep.2021.108974] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 02/26/2021] [Accepted: 03/19/2021] [Indexed: 12/12/2022] Open
Abstract
Clinical definitions of asthma fail to capture the heterogeneity of immune dysfunction in severe, treatment-refractory disease. Applying mass cytometry and machine learning to bronchoalveolar lavage (BAL) cells, we find that corticosteroid-resistant asthma patients cluster largely into two groups: one enriched in interleukin (IL)-4+ innate immune cells and another dominated by interferon (IFN)-γ+ T cells, including tissue-resident memory cells. In contrast, BAL cells of a healthier population are enriched in IL-10+ macrophages. To better understand cellular mediators of severe asthma, we developed the Immune Cell Linkage through Exploratory Matrices (ICLite) algorithm to perform deconvolution of bulk RNA sequencing of mixed-cell populations. Signatures of mitosis and IL-7 signaling in CD206-FcεRI+CD127+IL-4+ innate cells in one patient group, contrasting with adaptive immune response in T cells in the other, are preserved across technologies. Transcriptional signatures uncovered by ICLite identify T-cell-high and T-cell-poor severe asthma patients in an independent cohort, suggesting broad applicability of our findings.
Collapse
Affiliation(s)
- Matthew J Camiolo
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Center for Systems Immunology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Xiaoying Zhou
- Sean N Parker Center for Allergy Research and Division of Pulmonary, Allergy, and Critical Care Medicine, Stanford University, Stanford, CA, USA
| | - Timothy B Oriss
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Qi Yan
- Division of Pulmonary Medicine, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Michael Gorry
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - William Horne
- Richard King Mellon Foundation Institute for Pediatric Research, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - John B Trudeau
- Department of Environmental Medicine and Occupational Health, Graduate School of Public Health, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Kathryn Scholl
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Wei Chen
- Division of Pulmonary Medicine, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Jay K Kolls
- Department of Medicine and Center for Translational Research in Infection and Inflammation Tulane School of Medicine, New Orleans, LA, USA
| | - Prabir Ray
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Florian J Weisel
- Departments of Anesthesiology, Pain, and Peri-operative Medicine and Biomedical Data Sciences, Stanford University, Stanford, CA, USA
| | - Nadine M Weisel
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Nima Aghaeepour
- Departments of Anesthesiology, Pain, and Peri-operative Medicine and Biomedical Data Sciences, Stanford University, Stanford, CA, USA
| | - Kari Nadeau
- Sean N Parker Center for Allergy Research and Division of Pulmonary, Allergy, and Critical Care Medicine, Stanford University, Stanford, CA, USA
| | - Sally E Wenzel
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Environmental Medicine and Occupational Health, Graduate School of Public Health, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Anuradha Ray
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
104
|
Stephens LM, Ross KA, Waldstein KA, Legge KL, McLellan JS, Narasimhan B, Varga SM. Prefusion F-Based Polyanhydride Nanovaccine Induces Both Humoral and Cell-Mediated Immunity Resulting in Long-Lasting Protection against Respiratory Syncytial Virus. THE JOURNAL OF IMMUNOLOGY 2021; 206:2122-2134. [PMID: 33827894 DOI: 10.4049/jimmunol.2100018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 02/19/2021] [Indexed: 11/19/2022]
Abstract
Respiratory syncytial virus (RSV) is a leading cause of lower respiratory tract infection in both young children and in older adults. Despite the morbidity, mortality, and high economic burden caused by RSV worldwide, no licensed vaccine is currently available. We have developed a novel RSV vaccine composed of a prefusion-stabilized variant of the fusion (F) protein (DS-Cav1) and a CpG oligodeoxynucleotide adjuvant encapsulated within polyanhydride nanoparticles, termed RSVNanoVax. A prime-boost intranasal administration of RSVNanoVax in BALB/c mice significantly alleviated weight loss and pulmonary dysfunction in response to an RSV challenge, with protection maintained up to at least 6 mo postvaccination. In addition, vaccinated mice exhibited rapid viral clearance in the lungs as early as 2 d after RSV infection in both inbred and outbred populations. Vaccination induced tissue-resident memory CD4 and CD8 T cells in the lungs, as well as RSV F-directed neutralizing Abs. Based on the robust immune response elicited and the high level of durable protection observed, our prefusion RSV F nanovaccine is a promising new RSV vaccine candidate.
Collapse
Affiliation(s)
- Laura M Stephens
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA
| | - Kathleen A Ross
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA.,Nanovaccine Institute, Ames, IA
| | - Kody A Waldstein
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA
| | - Kevin L Legge
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA.,Nanovaccine Institute, Ames, IA.,Department of Microbiology and Immunology, University of Iowa, Iowa City, IA.,Department of Pathology, University of Iowa, Iowa City, IA; and
| | - Jason S McLellan
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX
| | - Balaji Narasimhan
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA.,Nanovaccine Institute, Ames, IA
| | - Steven M Varga
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA; .,Nanovaccine Institute, Ames, IA.,Department of Microbiology and Immunology, University of Iowa, Iowa City, IA.,Department of Pathology, University of Iowa, Iowa City, IA; and
| |
Collapse
|
105
|
Knight FC, Wilson JT. Engineering Vaccines for Tissue-Resident Memory T Cells. ADVANCED THERAPEUTICS 2021; 4:2000230. [PMID: 33997268 PMCID: PMC8114897 DOI: 10.1002/adtp.202000230] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Indexed: 01/01/2023]
Abstract
In recent years, tissue-resident memory T cells (TRM) have attracted significant attention in the field of vaccine development. Distinct from central and effector memory T cells, TRM cells take up residence in home tissues such as the lung or urogenital tract and are ideally positioned to respond quickly to pathogen encounter. TRM have been found to play a role in the immune response against many globally important infectious diseases for which new or improved vaccines are needed, including influenza and tuberculosis. It is also increasingly clear that TRM play a pivotal role in cancer immunity. Thus, vaccines that can generate this memory T cell population are highly desirable. The field of immunoengineering-that is, the application of engineering principles to study the immune system and design new and improved therapies that harness or modulate immune responses-is ideally poised to provide solutions to this need for next-generation TRM vaccines. This review covers recent developments in vaccine technologies for generating TRM and protecting against infection and cancer, including viral vectors, virus-like particles, and synthetic and natural biomaterials. In addition, it offers critical insights on the future of engineering vaccines for tissue-resident memory T cells.
Collapse
Affiliation(s)
- Frances C. Knight
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - John T. Wilson
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
| |
Collapse
|
106
|
Detsika MG, Ampelakiotou K, Grigoriou E, Psarra K, Jahaj E, Roussos C, Dimopoulou I, Orfanos SE, Tsirogianni A, Kotanidou A. A novel ratio of CD8 +:B-cells as a prognostic marker of coronavirus disease 2019 patient progression and outcome. Virology 2021; 556:79-86. [PMID: 33550117 PMCID: PMC7831474 DOI: 10.1016/j.virol.2021.01.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 12/11/2020] [Accepted: 01/05/2021] [Indexed: 12/13/2022]
Abstract
Infection with SARS-COV-2 may result in severe pneumonia potentially leading to mechanical ventilation and intensive care treatment. The aim of the present study was to analyze the immune responses in critically ill coronavirus 2019 (COVID-19) patients requiring mechanical ventilation and assess their potential use as markers of clinical progression and outcome. Confirmed COVID-19 patients were grouped into those requiring mechanical ventilation (intubated) and non-intubated. Immune phenotyping was performed and cytokine levels were determined. A novel ratio of CD8+:B cells was significantly lower in intubated versus non-intubated (p = 0.015) and intubated non-survivors (NSV) versus survivors (SV) (p = 0.015). The same ratio correlated with outcome, CRP, IL-6 levels and neutrophil count. Receiving operating curve (ROC) analysis for prediction of requirement of mechanical ventilation by the CD8+:B cells ratio revealed an AUC of 0.747 and a p = 0.007. The ratio of CD8+:B cells may serve as a useful prognostic marker for disease severity and outcome.
Collapse
Affiliation(s)
- Maria G. Detsika
- 1st Department of Critical Care Medicine & Pulmonary Services, GP Livanos and M Simou Laboratories, Evangelismos Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Kleio Ampelakiotou
- Department of Immunology and Histocompatibility, ‘Evangelismos' General Hospital, Athens, Greece
| | - Eirini Grigoriou
- Department of Immunology and Histocompatibility, ‘Evangelismos' General Hospital, Athens, Greece
| | - Katherina Psarra
- Department of Immunology and Histocompatibility, ‘Evangelismos' General Hospital, Athens, Greece
| | - Edison Jahaj
- 1st Department of Critical Care Medicine & Pulmonary Services, GP Livanos and M Simou Laboratories, Evangelismos Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Charis Roussos
- 1st Department of Critical Care Medicine & Pulmonary Services, GP Livanos and M Simou Laboratories, Evangelismos Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Ioanna Dimopoulou
- 1st Department of Critical Care Medicine & Pulmonary Services, GP Livanos and M Simou Laboratories, Evangelismos Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Stylianos E. Orfanos
- 1st Department of Critical Care Medicine & Pulmonary Services, GP Livanos and M Simou Laboratories, Evangelismos Hospital, National and Kapodistrian University of Athens, Athens, Greece,2nd Department of Critical Care Medicine, Attikon Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Alexandra Tsirogianni
- Department of Immunology and Histocompatibility, ‘Evangelismos' General Hospital, Athens, Greece
| | - Anastasia Kotanidou
- 1st Department of Critical Care Medicine & Pulmonary Services, GP Livanos and M Simou Laboratories, Evangelismos Hospital, National and Kapodistrian University of Athens, Athens, Greece.
| |
Collapse
|
107
|
Harker JA, Lloyd CM. Overlapping and distinct features of viral and allergen immunity in the human lung. Immunity 2021; 54:617-631. [PMID: 33852829 DOI: 10.1016/j.immuni.2021.03.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 02/28/2021] [Accepted: 03/15/2021] [Indexed: 12/30/2022]
Abstract
Immunity in the human respiratory tract is provided by a diverse range of tissue-resident cells, including specialized epithelial and macrophage populations and a network of innate and innate-like lymphocytes, such as natural killer cells, innate lymphoid cells, and invariant T cells. Lung-resident memory T and B cells contribute to this network following initial exposure to antigenic stimuli. This review explores how advances in the study of human immunology have shaped our understanding of this resident immune network and its response to two of the most commonly encountered inflammatory stimuli in the airways: viruses and allergens. It discusses the many ways in which pathogenic infection and allergic inflammation mirror each other, highlighting the key checkpoints at which they diverge and how this can result in a lifetime of allergic exacerbation versus protective anti-viral immunity.
Collapse
Affiliation(s)
- James A Harker
- National Heart and Lung Institute, Imperial College London, London SW7 2AZ, UK
| | - Clare M Lloyd
- National Heart and Lung Institute, Imperial College London, London SW7 2AZ, UK.
| |
Collapse
|
108
|
Kume M, Kiyohara E, Matsumura Y, Koguchi-Yoshioka H, Tanemura A, Hanaoka Y, Taminato M, Tashima H, Tomita K, Kubo T, Watanabe R, Fujimoto M. Ganglioside GD3 May Suppress the Functional Activities of Benign Skin T Cells in Cutaneous T-Cell Lymphoma. Front Immunol 2021; 12:651048. [PMID: 33859643 PMCID: PMC8042233 DOI: 10.3389/fimmu.2021.651048] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 03/15/2021] [Indexed: 01/11/2023] Open
Abstract
In cutaneous T-cell lymphoma (CTCL), which arises from skin-tropic memory T cells, malignant T cells and benign T cells are confined in the same skin lesions. It is thus difficult to evaluate the phenotypic characteristics and functional activities of benign T cells in CTCL. Disialoganglioside with three glycosyl groups (GD3) is increasingly expressed on the surface of solid malignant tumor cells and takes part in tumor progression and suppression of tumor immunity. However, the role of GD3 in CTCL is not well-understood. In this study, the malignant and benign T cells in CTCL skin lesions were distinguished by flow cytometry and their phenotypic characteristics were compared with those of T cells from control skin specimens. In CTCL skin lesions, the benign T cells included limited resident memory T cells (TRM), which are sessile in skin and known to exert strong antitumor function. The benign T cells showed diminished Th17 property, and the expression of GD3 was high in the malignant T cells. The expression of GD3 in the malignant T cells inversely correlated with IL-17A production from the benign CD4 T cells. GD3 from the malignant T cells was implied to be involved in suppressing the Th17 activity of the benign T cells independent of the regulation of TRM differentiation in CTCL. Revealing the role of GD3 in inhibiting the production of IL-17A in CTCL would aid the understanding of the suppressive mechanism of the antitumor activity by malignant tumor cells.
Collapse
Affiliation(s)
- Miki Kume
- Department of Dermatology, Course of Integrated Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Eiji Kiyohara
- Department of Dermatology, Course of Integrated Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Yutaka Matsumura
- Department of Dermatology, Course of Integrated Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Hanako Koguchi-Yoshioka
- Department of Dermatology, Course of Integrated Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan.,Department of Integrative Medicine for Allergic and Immunological Diseases, Course of Integrated Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Atsushi Tanemura
- Department of Dermatology, Course of Integrated Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Yuma Hanaoka
- Department of Dermatology, Course of Integrated Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Mifue Taminato
- Department of Plastic Surgery, Course of Organ Regulation Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Hiroki Tashima
- Department of Plastic Surgery, Course of Organ Regulation Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Koichi Tomita
- Department of Plastic Surgery, Course of Organ Regulation Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Tateki Kubo
- Department of Plastic Surgery, Course of Organ Regulation Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Rei Watanabe
- Department of Dermatology, Course of Integrated Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan.,Department of Integrative Medicine for Allergic and Immunological Diseases, Course of Integrated Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Manabu Fujimoto
- Department of Dermatology, Course of Integrated Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| |
Collapse
|
109
|
Stolley JM, Johnston TS, Soerens AG, Beura LK, Rosato PC, Joag V, Wijeyesinghe SP, Langlois RA, Osum KC, Mitchell JS, Masopust D. Retrograde migration supplies resident memory T cells to lung-draining LN after influenza infection. J Exp Med 2021; 217:151876. [PMID: 32568362 PMCID: PMC7398169 DOI: 10.1084/jem.20192197] [Citation(s) in RCA: 80] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 03/20/2020] [Accepted: 05/12/2020] [Indexed: 01/06/2023] Open
Abstract
Numerous observations indicate that resident memory T cells (TRM) undergo unusually rapid attrition within the lung. Here we demonstrate that contraction of lung CD8+ T cell responses after influenza infection is contemporized with egress of CD69+/CD103+ CD8+ T cells to the draining mediastinal LN via the lymphatic vessels, which we term retrograde migration. Cells within the draining LN retained canonical markers of lung TRM, including CD103 and CD69, lacked Ly6C expression (also a feature of lung TRM), maintained granzyme B expression, and did not equilibrate among immunized parabiotic mice. Investigations of bystander infection or removal of the TCR from established memory cells revealed that the induction of the TRM phenotype was dependent on antigen recognition; however, maintenance was independent. Thus, local lung infection induces CD8+ T cells with a TRM phenotype that nevertheless undergo retrograde migration, yet remain durably committed to the residency program within the draining LN, where they provide longer-lived regional memory while chronicling previous upstream antigen experiences.
Collapse
Affiliation(s)
- J Michael Stolley
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN.,Center for Immunology, University of Minnesota, Minneapolis, MN
| | - Timothy S Johnston
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN.,Center for Immunology, University of Minnesota, Minneapolis, MN
| | - Andrew G Soerens
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN.,Center for Immunology, University of Minnesota, Minneapolis, MN
| | - Lalit K Beura
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN.,Center for Immunology, University of Minnesota, Minneapolis, MN.,Department of Microbiology and Immunology, Brown University, Providence, RI
| | - Pamela C Rosato
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN.,Center for Immunology, University of Minnesota, Minneapolis, MN
| | - Vineet Joag
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN.,Center for Immunology, University of Minnesota, Minneapolis, MN
| | - Sathi P Wijeyesinghe
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN.,Center for Immunology, University of Minnesota, Minneapolis, MN
| | - Ryan A Langlois
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN.,Center for Immunology, University of Minnesota, Minneapolis, MN
| | - Kevin C Osum
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN.,Center for Immunology, University of Minnesota, Minneapolis, MN
| | - Jason S Mitchell
- Center for Immunology, University of Minnesota, Minneapolis, MN.,Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN
| | - David Masopust
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN.,Center for Immunology, University of Minnesota, Minneapolis, MN
| |
Collapse
|
110
|
Shilovskiy IP, Yumashev KV, Nikolsky AA, Vishnyakova LI, Khaitov MR. Molecular and Cellular Mechanisms of Respiratory Syncytial Viral Infection: Using Murine Models to Understand Human Pathology. BIOCHEMISTRY. BIOKHIMIIA 2021; 86:290-306. [PMID: 33838630 PMCID: PMC7957450 DOI: 10.1134/s0006297921030068] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 10/18/2020] [Accepted: 10/18/2020] [Indexed: 12/28/2022]
Abstract
Respiratory syncytial virus (RSV) causes severe pathology of the lower respiratory tract in infants, immunocompromised people, and elderly. Despite decades of research, there is no licensed vaccine against RSV, and many therapeutic drugs are still under development. Detailed understanding of molecular and cellular mechanisms of the RSV infection pathology can accelerate the development of efficacious treatment. Current studies on the RSV pathogenesis are based on the analysis of biopsies from the infected patients; however deeper understanding of molecular and cellular mechanisms of the RSV pathology could be achieved using animal models. Mice are the most often used model for RSV infection because they exhibit manifestations similar to those observed in humans (bronchial obstruction, mucous hypersecretion, and pulmonary inflammation mediated by lymphocytes, macrophages, and neutrophils). Additionally, the use of mice is economically feasible, and many molecular tools are available for studying RSV infection pathogenesis at the molecular and cellular levels. This review summarizes new data on the pathogenesis of RSV infection obtained in mouse models, which demonstrated the role of T cells in both the antiviral defense and the development of lung immunopathology. T cells not only eliminate the infected cells, but also produce significant amounts of the proinflammatory cytokines TNFα and IFNγ. Recently, a new subset of tissue-resident memory T cells (TRM) was identified that provide a strong antiviral defense without induction of lung immunopathology. These cells accumulate in the lungs after local rather than systemic administration of RSV antigens, which suggests new approaches to vaccination. The studies in mouse models have revealed a minor role of interferons in the anti-RSV protection, as RSV possesses mechanisms to escape the antiviral action of type I and III interferons, which may explain the low efficacy of interferon-containing drugs. Using knockout mice, a significant breakthrough has been achieved in understanding the role of many pro-inflammatory cytokines in lung immunopathology. It was found that in addition to TNFα and IFNγ, the cytokines IL-4, IL-5, IL-13, IL-17A, IL-33, and TSLP mediate the major manifestations of the RSV pathogenesis, such as bronchial obstruction, mucus hyperproduction, and lung infiltration by pro-inflammatory cells, while IL-6, IL-10, and IL-27 exhibit the anti-inflammatory effect. Despite significant differences between the mouse and human immune systems, mouse models have made a significant contribution to the understanding of molecular and cellular mechanisms of the pathology of human RSV infection.
Collapse
Affiliation(s)
- Igor P Shilovskiy
- National Research Center, Institute of Immunology, Federal Medico-Biological Agency, Moscow, 115522, Russia.
| | - Kirill V Yumashev
- National Research Center, Institute of Immunology, Federal Medico-Biological Agency, Moscow, 115522, Russia
| | - Alexandr A Nikolsky
- National Research Center, Institute of Immunology, Federal Medico-Biological Agency, Moscow, 115522, Russia
| | - Liudmila I Vishnyakova
- National Research Center, Institute of Immunology, Federal Medico-Biological Agency, Moscow, 115522, Russia
| | - Musa R Khaitov
- National Research Center, Institute of Immunology, Federal Medico-Biological Agency, Moscow, 115522, Russia
| |
Collapse
|
111
|
Xu X, Qiao D, Dong C, Mann M, Garofalo RP, Keles S, Brasier AR. The SWI/SNF-Related, Matrix Associated, Actin-Dependent Regulator of Chromatin A4 Core Complex Represses Respiratory Syncytial Virus-Induced Syncytia Formation and Subepithelial Myofibroblast Transition. Front Immunol 2021; 12:633654. [PMID: 33732255 PMCID: PMC7957062 DOI: 10.3389/fimmu.2021.633654] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 01/18/2021] [Indexed: 12/15/2022] Open
Abstract
Epigenetics plays an important role in the priming the dynamic response of airway epithelial cells to infectious and environmental stressors. Here, we examine the epigenetic role of the SWI/SNF Related, Matrix Associated, Actin Dependent Regulator of Chromatin A4 (SMARCA4) in the epithelial response to RSV infection. Depletion of SMARCA4 destabilized the abundance of the SMARCE1/ARID1A SWI/SNF subunits, disrupting the innate response and triggering a hybrid epithelial/mesenchymal (E/M) state. Assaying SMARCA4 complex-regulated open chromatin domains by transposase cleavage -next generation sequencing (ATAC-Seq), we observed that the majority of cleavage sites in uninfected cells have reduced chromatin accessibility. Paradoxically, SMARCA4 complex-depleted cells showed enhanced RSV-inducible chromatin opening and gene expression in the EMT pathway genes, MMP9, SNAI1/2, VIM, and CDH2. Focusing on the key MMP9, we observed that SMARCA4 complex depletion reduced basal BRD4 and RNA Polymerase II binding, but enhanced BRD4/Pol II binding in response to RSV infection. In addition, we observed that MMP9 secretion in SMARCA4 complex deficient cells contributes to mesenchymal transition, cellular fusion (syncytia) and subepithelial myofibroblast transition. We conclude the SMARCA4 complex is a transcriptional repressor of epithelial plasticity, whose depletion triggers a hybrid E/M state that affects the dynamic response of the small airway epithelial cell in mucosal remodeling via paracrine MMP9 activity.
Collapse
Affiliation(s)
- Xiaofang Xu
- Department of Internal Medicine, University of Wisconsin-Madison School of Medicine and Public Health (SMPH), Madison, WI, United States
| | - Dianhua Qiao
- Department of Internal Medicine, University of Wisconsin-Madison School of Medicine and Public Health (SMPH), Madison, WI, United States
| | - Chenyang Dong
- Department of Statistics, University of Wisconsin-Madison, Madison, WI, United States
| | - Morgan Mann
- Department of Internal Medicine, University of Wisconsin-Madison School of Medicine and Public Health (SMPH), Madison, WI, United States
| | - Roberto P. Garofalo
- Department of Pediatrics, University of Texas Medical Branch, Galveston, TX, United States
| | - Sunduz Keles
- Department of Statistics, University of Wisconsin-Madison, Madison, WI, United States
- Department of Biostatistics & Medical Informatics, University of Wisconsin-Madison, Madison, WI, United States
| | - Allan R. Brasier
- Department of Internal Medicine, University of Wisconsin-Madison School of Medicine and Public Health (SMPH), Madison, WI, United States
- Institute for Clinical and Translational Research (ICTR), University of Wisconsin-Madison, Madison, WI, United States
| |
Collapse
|
112
|
Huang S, He Q, Zhou L. T cell responses in respiratory viral infections and chronic obstructive pulmonary disease. Chin Med J (Engl) 2021; 134:1522-1534. [PMID: 33655898 PMCID: PMC8280062 DOI: 10.1097/cm9.0000000000001388] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Indexed: 12/21/2022] Open
Abstract
ABSTRACT Respiratory viruses are major human pathogens that cause approximately 200 million pneumonia cases annually and induce various comorbidities with chronic obstructive pulmonary disease (COPD), resulting in significant health concerns and economic burdens. Clinical manifestations in respiratory viral infections and inflammations vary from asymptomatic, mild, to severe, depending on host immune cell responses to pathogens and interactions with airway epithelia. We critically review the activation, effector, and regulation of T cells in respiratory virus infections and chronic inflammations associated with COPD. Crosstalk among T cells, innate immune cells, and airway epithelial cells is discussed as essential parts of pathogenesis and protection in viral infections and COPD. We emphasize the specificity of peptide antigens and the functional heterogeneity of conventional CD4+ and CD8+ T cells to shed some light on potential cellular and molecular candidates for the future development of therapeutics and intervention against respiratory viral infections and inflammations.
Collapse
Affiliation(s)
- Shouxiong Huang
- Department of Environmental and Public Health Sciences, University of Cincinnati College of Medicine, Immunology Graduate Program, Cincinnati Children's Hospital, Cincinnati, OH 45249, USA
| | - Quan He
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Linfu Zhou
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| |
Collapse
|
113
|
Gelbard A, Wanjalla C, Wootten CT, Drake WP, Lowery AS, Wheeler DA, Cardenas MF, Sikora AG, Pathak RR, McDonnell W, Mallal S, Pilkinton M. The Proximal Airway Is a Reservoir for Adaptive Immunologic Memory in Idiopathic Subglottic Stenosis. Laryngoscope 2021; 131:610-617. [PMID: 32603507 PMCID: PMC7951501 DOI: 10.1002/lary.28840] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 05/18/2020] [Accepted: 05/21/2020] [Indexed: 01/20/2023]
Abstract
OBJECTIVES/HYPOTHESIS Characterization of the localized adaptive immune response in the airway scar of patients with idiopathic subglottic stenosis (iSGS). STUDY DESIGN Basic Science. METHODS Utilizing 36 patients with subglottic stenosis (25 idiopathic subglottic stenosis [iSGS], 10 iatrogenic post-intubation stenosis [iLTS], and one granulomatosis with polyangiitis [GPA]) we applied immunohistochemical and immunologic techniques coupled with RNA sequencing. RESULTS iSGS, iLTS, and GPA demonstrate a significant immune infiltrate in the subglottic scar consisting of adaptive cell subsets (T cells along with dendritic cells). Interrogation of T cell subtypes showed significantly more CD69+ CD103+ CD8+ tissue resident memory T cells (TRM ) in the iSGS airway scar than iLTS specimens (iSGS vs. iLTS; 50% vs. 28%, P = .0065). Additionally, subglottic CD8+ clones possessed T-cell receptor (TCR) sequences with known antigen specificity for viral and intracellular pathogens. CONCLUSIONS The human subglottis is significantly enriched for CD8+ tissue resident memory T cells in iSGS, which possess TCR sequences proven to recognize viral and intracellular pathogens. These results inform our understanding of iSGS, provide a direction for future discovery, and demonstrate immunologic function in the human proximal airway. Laryngoscope, 131:610-617, 2021.
Collapse
Affiliation(s)
- Alexander Gelbard
- Dept. of Otolaryngology-Head & Neck Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Celestine Wanjalla
- Dept. of Medicine, Division of Infectious Disease, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Christopher T. Wootten
- Dept. of Otolaryngology-Head & Neck Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Wonder P. Drake
- Dept. of Molecular and Human Genetics, Baylor College of Medicine, Houston Texas
| | - Anne S Lowery
- Dept. of Otolaryngology-Head & Neck Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | - David A. Wheeler
- Dept. of Molecular and Human Genetics, Baylor College of Medicine, Houston Texas
| | - Maria F. Cardenas
- Dept. of Molecular and Human Genetics, Baylor College of Medicine, Houston Texas
| | - Andrew G. Sikora
- Bobby R. Alford Dept. of Otolaryngology-Head & Neck Surgery, Baylor College of Medicine, Houston Texas
| | - Ravi R. Pathak
- Bobby R. Alford Dept. of Otolaryngology-Head & Neck Surgery, Baylor College of Medicine, Houston Texas
| | | | - Simon Mallal
- Dept. of Molecular and Human Genetics, Baylor College of Medicine, Houston Texas
| | - Mark Pilkinton
- Dept. of Medicine, Division of Infectious Disease, Vanderbilt University Medical Center, Nashville, Tennessee
| |
Collapse
|
114
|
Kirchner FR, LeibundGut-Landmann S. Tissue-resident memory Th17 cells maintain stable fungal commensalism in the oral mucosa. Mucosal Immunol 2021; 14:455-467. [PMID: 32719409 PMCID: PMC7946631 DOI: 10.1038/s41385-020-0327-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 07/06/2020] [Indexed: 02/04/2023]
Abstract
Keeping a stable equilibrium between the host and commensal microbes to which we are constantly exposed, poses a major challenge for the immune system. The host mechanisms that regulate homeostasis of the microbiota to prevent infection and inflammatory disorders are not fully understood. Here, we provide evidence that CD4+ tissue-resident memory T (TRM) cells act as central players in this process. Using a murine model of C. albicans commensalism we show that IL-17 producing CD69+CD103+CD4+ memory T cells persist in the colonized tissue long-term and independently of circulatory supplies. Consistent with the requirement of Th17 cells for limiting fungal growth, IL-17-producing TRM cells in the mucosa were sufficient to maintain prolonged colonization, while circulatory T cells were dispensable. Although TRM cells were first proposed to protect from pathogens causing recurrent acute infections, our results support a central function of TRM cells in the maintenance of commensalism.
Collapse
Affiliation(s)
- Florian R Kirchner
- Section of Immunology, Vetsuisse Faculty, University of Zürich, Winterthurerstrasse 266a, CH-8057, Zürich, Switzerland
- Institute of Experimental Immunology, University of Zürich, Winterthurerstrasse 190, CH-8057, Zürich, Switzerland
| | - Salomé LeibundGut-Landmann
- Section of Immunology, Vetsuisse Faculty, University of Zürich, Winterthurerstrasse 266a, CH-8057, Zürich, Switzerland.
- Institute of Experimental Immunology, University of Zürich, Winterthurerstrasse 190, CH-8057, Zürich, Switzerland.
| |
Collapse
|
115
|
Koutsakos M, Rowntree LC, Hensen L, Chua BY, van de Sandt CE, Habel JR, Zhang W, Jia X, Kedzierski L, Ashhurst TM, Putri GH, Marsh-Wakefield F, Read MN, Edwards DN, Clemens EB, Wong CY, Mordant FL, Juno JA, Amanat F, Audsley J, Holmes NE, Gordon CL, Smibert OC, Trubiano JA, Hughes CM, Catton M, Denholm JT, Tong SY, Doolan DL, Kotsimbos TC, Jackson DC, Krammer F, Godfrey DI, Chung AW, King NJ, Lewin SR, Wheatley AK, Kent SJ, Subbarao K, McMahon J, Thevarajan I, Nguyen TH, Cheng AC, Kedzierska K. Integrated immune dynamics define correlates of COVID-19 severity and antibody responses. CELL REPORTS MEDICINE 2021; 2:100208. [PMID: 33564749 PMCID: PMC7862905 DOI: 10.1016/j.xcrm.2021.100208] [Citation(s) in RCA: 87] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 11/05/2020] [Accepted: 02/01/2021] [Indexed: 12/15/2022]
Abstract
SARS-CoV-2 causes a spectrum of COVID-19 disease, the immunological basis of which remains ill defined. We analyzed 85 SARS-CoV-2-infected individuals at acute and/or convalescent time points, up to 102 days after symptom onset, quantifying 184 immunological parameters. Acute COVID-19 presented with high levels of IL-6, IL-18, and IL-10 and broad activation marked by the upregulation of CD38 on innate and adaptive lymphocytes and myeloid cells. Importantly, activated CXCR3+cTFH1 cells in acute COVID-19 significantly correlate with and predict antibody levels and their avidity at convalescence as well as acute neutralization activity. Strikingly, intensive care unit (ICU) patients with severe COVID-19 display higher levels of soluble IL-6, IL-6R, and IL-18, and hyperactivation of innate, adaptive, and myeloid compartments than patients with moderate disease. Our analyses provide a comprehensive map of longitudinal immunological responses in COVID-19 patients and integrate key cellular pathways of complex immune networks underpinning severe COVID-19, providing important insights into potential biomarkers and immunotherapies. Analyses of 184 immune features define kinetics of immune responses to SARS-CoV-2 Circulating TFH1 cells in acute COVID-19 correlate with antibodies sIL-6R levels are elevated in severe COVID-19 but do not correlate with IL-6 Elevated IL-6 and IL-18 correlate with immune cell hyperactivation
Collapse
Affiliation(s)
- Marios Koutsakos
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Louise C. Rowntree
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Luca Hensen
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Brendon Y. Chua
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
- Global Station for Zoonosis Control, Global Institution for Collaborative Research and Education (GI-CoRE), Hokkaido University, Sapporo, Japan
| | - Carolien E. van de Sandt
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Jennifer R. Habel
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Wuji Zhang
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Xiaoxiao Jia
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Lukasz Kedzierski
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
- Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Melbourne, VIC, Australia
| | - Thomas M. Ashhurst
- Sydney Cytometry Core Research Facility, Charles Perkins Centre, Centenary Institute and University of Sydney, Sydney, NSW, Australia
- Marie Bashir Institute for Infectious Diseases and Biosecurity, University of Sydney, Sydney, NSW, Australia
| | - Givanna H. Putri
- Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
- School of Computer Science, University of Sydney, Sydney, NSW, Australia
| | - Felix Marsh-Wakefield
- Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
- Vascular Immunology Unit, Discipline of Pathology, School of Medical Sciences, University of Sydney, Sydney, NSW, Australia
| | - Mark N. Read
- Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
- School of Computer Science, University of Sydney, Sydney, NSW, Australia
- The Westmead Initiative, University of Sydney, Sydney, NSW, Australia
| | - Davis N. Edwards
- School of Computer Science, University of Sydney, Sydney, NSW, Australia
- The Westmead Initiative, University of Sydney, Sydney, NSW, Australia
| | - E. Bridie Clemens
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Chinn Yi Wong
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Francesca L. Mordant
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Jennifer A. Juno
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Fatima Amanat
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029-6574, USA
| | - Jennifer Audsley
- Department of Infectious Diseases, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Natasha E. Holmes
- Department of Infectious Diseases, Austin Hospital, Heidelberg, VIC, Australia
- Department of Medicine and Radiology, University of Melbourne, Parkville, VIC, Australia
- Data Analytics Research and Evaluation (DARE) Centre, Austin Health and University of Melbourne, Heidelberg, VIC, Australia
- Centre for Antibiotic Allergy and Research, Department of Infectious Diseases, Austin Health, Heidelberg, VIC, Australia
| | - Claire L. Gordon
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
- Department of Infectious Diseases, Austin Hospital, Heidelberg, VIC, Australia
| | - Olivia C. Smibert
- Department of Infectious Diseases, Austin Hospital, Heidelberg, VIC, Australia
- Department of Infectious Diseases, Peter McCallum Cancer Centre, Melbourne, VIC, Australia
- National Centre for Infections in Cancer, Peter McCallum Cancer Centre, Melbourne, VIC, Australia
| | - Jason A. Trubiano
- Centre for Antibiotic Allergy and Research, Department of Infectious Diseases, Austin Health, Heidelberg, VIC, Australia
- Department of Infectious Diseases, Peter McCallum Cancer Centre, Melbourne, VIC, Australia
- National Centre for Infections in Cancer, Peter McCallum Cancer Centre, Melbourne, VIC, Australia
- Department of Medicine (Austin Health), University of Melbourne, Heidelberg, VIC, Australia
| | - Carly M. Hughes
- Monash Infectious Diseases, Monash Medical Centre, Monash Health, Melbourne, VIC, Australia
| | - Mike Catton
- Victorian Infectious Diseases Reference Laboratory, Royal Melbourne Hospital at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Justin T. Denholm
- Department of Infectious Diseases, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
- Victorian Infectious Diseases Services, Royal Melbourne Hospital and Doherty Department University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne VIC, Australia
| | - Steven Y.C. Tong
- Department of Infectious Diseases, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
- Victorian Infectious Diseases Services, Royal Melbourne Hospital and Doherty Department University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne VIC, Australia
- Menzies School of Health Research, Charles Darwin University, Darwin, NT, Australia
| | - Denise L. Doolan
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health & Medicine, James Cook University, Cairns, QLD, Australia
| | - Tom C. Kotsimbos
- Department of Medicine, Central Clinical School, Monash University, Melbourne, VIC, Australia
- Department of Allergy, Immunology, and Respiratory Medicine, Alfred Hospital, Melbourne, VIC, Australia
| | - David C. Jackson
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
- Global Station for Zoonosis Control, Global Institution for Collaborative Research and Education (GI-CoRE), Hokkaido University, Sapporo, Japan
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Dale I. Godfrey
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
- Australian Research Council Centre of Excellence for Advanced Molecular Imaging at the University of Melbourne, Melbourne, VIC, Australia
| | - Amy W. Chung
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Nicholas J.C. King
- Sydney Cytometry Core Research Facility, Charles Perkins Centre, Centenary Institute and University of Sydney, Sydney, NSW, Australia
- Marie Bashir Institute for Infectious Diseases and Biosecurity, University of Sydney, Sydney, NSW, Australia
- Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
- Viral Immunopathology Laboratory, Discipline of Pathology, School of Medical Sciences, University of Sydney, Sydney, NSW, Australia
- Sydney Nano, University of Sydney, Sydney, NSW 2006, Australia
| | - Sharon R. Lewin
- Department of Infectious Diseases, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
- Victorian Infectious Diseases Services, Royal Melbourne Hospital and Doherty Department University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne VIC, Australia
- Department of Infectious Diseases, Monash University and Alfred Hospital, Melbourne, VIC, Australia
| | - Adam K. Wheatley
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, University of Melbourne, Melbourne, VIC, Australia
| | - Stephen J. Kent
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, University of Melbourne, Melbourne, VIC, Australia
- Melbourne Sexual Health Centre, Infectious Diseases Department, Alfred Health, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Kanta Subbarao
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
- World Health Organization Collaborating Centre for Reference and Research on Influenza at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - James McMahon
- Monash Infectious Diseases, Monash Medical Centre, Monash Health, Melbourne, VIC, Australia
- Department of Infectious Diseases, Monash University and Alfred Hospital, Melbourne, VIC, Australia
| | - Irani Thevarajan
- Department of Infectious Diseases, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
- Victorian Infectious Diseases Services, Royal Melbourne Hospital and Doherty Department University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne VIC, Australia
| | - Thi H.O. Nguyen
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Allen C. Cheng
- School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, Australia
- Infection Prevention and Healthcare Epidemiology Unit, Alfred Health, Melbourne, VIC, Australia
| | - Katherine Kedzierska
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
- Global Station for Zoonosis Control, Global Institution for Collaborative Research and Education (GI-CoRE), Hokkaido University, Sapporo, Japan
- Corresponding author
| |
Collapse
|
116
|
Luangrath MA, Schmidt ME, Hartwig SM, Varga SM. Tissue-Resident Memory T Cells in the Lungs Protect against Acute Respiratory Syncytial Virus Infection. Immunohorizons 2021; 5:59-69. [PMID: 33536235 PMCID: PMC8299542 DOI: 10.4049/immunohorizons.2000067] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 01/12/2021] [Indexed: 01/01/2023] Open
Abstract
Respiratory syncytial virus (RSV) is the leading cause of lower respiratory tract infection in young children. The T cell response plays a critical role in facilitating clearance of an acute RSV infection, and memory T cell responses are vital for protection against secondary RSV exposures. Tissue-resident memory (TRM) T cells have been identified as a subset of memory T cells that reside in nonlymphoid tissues and are critical for providing long-term immunity. There is currently limited information regarding the establishment and longevity of TRM T cell responses elicited following an acute RSV infection as well as their role in protection against repeated RSV infections. In this study, we examined the magnitude, phenotype, and protective capacity of TRM CD4 and CD8 T cells in the lungs of BALB/c mice following an acute RSV infection. TRM CD4 and CD8 T cells were established within the lungs and waned by 149 d following RSV infection. To determine the protective capacity of TRMs, FTY720 administration was used to prevent trafficking of peripheral memory T cells into the lungs prior to challenge of RSV-immune mice, with a recombinant influenza virus expressing either an RSV-derived CD4 or CD8 T cell epitope. We observed enhanced viral clearance in RSV-immune mice, suggesting that TRM CD8 T cells can contribute to protection against a secondary RSV infection. Given the protective capacity of TRMs, future RSV vaccine candidates should focus on the generation of these cell populations within the lung to induce effective immunity against RSV infection.
Collapse
Affiliation(s)
- Mitchell A Luangrath
- Division of Critical Care, Stead Family Department of Pediatrics, University of Iowa, Iowa City, IA 52242
| | - Megan E Schmidt
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA 52242
| | - Stacey M Hartwig
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA 52242; and
| | - Steven M Varga
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA 52242;
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA 52242; and
- Department of Pathology, University of Iowa, Iowa City, IA 52242
| |
Collapse
|
117
|
Qian Y, Zhu Y, Li Y, Li B. Legend of the Sentinels: Development of Lung Resident Memory T Cells and Their Roles in Diseases. Front Immunol 2021; 11:624411. [PMID: 33603755 PMCID: PMC7884312 DOI: 10.3389/fimmu.2020.624411] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 12/21/2020] [Indexed: 01/23/2023] Open
Abstract
SARS-CoV-2 is wreaking havoc around the world. To get the world back on track, hundreds of vaccines are under development. A deeper understanding of how the immune system responds to SARS-CoV-2 re-infection will certainly help. Studies have highlighted various aspects of T cell response in resolving acute infection and preventing re-infections. Lung resident memory T (TRM) cells are sentinels in the secondary immune response. They are mostly differentiated from effector T cells, construct specific niches and stay permanently in lung tissues. If the infection recurs, locally activated lung TRM cells can elicit rapid immune response against invading pathogens. In addition, they can significantly limit tumor growth or lead to pathologic immune responses. Vaccines targeting TRM cells are under development, with the hope to induce stable and highly reactive lung TRM cells through mucosal administration or "prime-and-pull" strategy. In this review, we will summarize recent advances in lung TRM cell generation and maintenance, explore their roles in different diseases and discuss how these cells may guide the development of future vaccines targeting infectious disease, cancer, and pathologic immune response.
Collapse
Affiliation(s)
| | | | - Yangyang Li
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bin Li
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
118
|
Alternative mRNA Processing of Innate Response Pathways in Respiratory Syncytial Virus (RSV) Infection. Viruses 2021; 13:v13020218. [PMID: 33572560 PMCID: PMC7912025 DOI: 10.3390/v13020218] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 01/27/2021] [Accepted: 01/28/2021] [Indexed: 12/14/2022] Open
Abstract
The innate immune response (IIR) involves rapid genomic expression of protective interferons (IFNs) and inflammatory cytokines triggered by intracellular viral replication. Although the transcriptional control of the innate pathway is known in substantial detail, little is understood about the complexity of alternative splicing (AS) and alternative polyadenylation (APA) of mRNAs underlying the cellular IIR. In this study, we applied single-molecule, real-time (SMRT) sequencing with mRNA quantitation using short-read mRNA sequencing to characterize changes in mRNA processing in the epithelial response to respiratory syncytial virus (RSV) replication. Mock or RSV-infected human small-airway epithelial cells (hSAECs) were profiled using SMRT sequencing and the curated transcriptome analyzed by structural and quality annotation of novel transcript isoforms (SQANTI). We identified 113,082 unique isoforms; 28,561 represented full splice matches, and 45% of genes expressed six or greater AS mRNA isoforms. Identification of differentially expressed AS isoforms was accomplished by mapping a short-read RNA sequencing expression matrix to the curated transcriptome, and 905 transcripts underwent differential polyadenylation site analysis enriched in protein secretion, translation, and mRNA degradation. We focused on 355 genes showing differential isoform utilization (DIU), indicating where a new AS isoform becomes a major fraction of mRNA isoforms expressed. In pathway and network enrichment analyses, we observed that DIU transcripts are substantially enriched in cell cycle control and IIR pathways. Interestingly, the RelA/IRF7 innate regulators showed substantial DIU where major transcripts included distinct isoforms with exon occlusion, intron inclusion, and alternative transcription start site utilization. We validated the presence of RelA and IRF7 AS isoforms as well as their induction by RSV using eight isoform-specific RT-PCR assays. These isoforms were identified in both immortalized and primary small-airway epithelial cells. We concluded that the cell cycle and IIR are differentially spliced in response to RSV. These data indicate that substantial post-transcriptional complexity regulates the antiviral response.
Collapse
|
119
|
T Cell Immunity against Influenza: The Long Way from Animal Models Towards a Real-Life Universal Flu Vaccine. Viruses 2021; 13:v13020199. [PMID: 33525620 PMCID: PMC7911237 DOI: 10.3390/v13020199] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 01/23/2021] [Accepted: 01/25/2021] [Indexed: 02/07/2023] Open
Abstract
Current flu vaccines rely on the induction of strain-specific neutralizing antibodies, which leaves the population vulnerable to drifted seasonal or newly emerged pandemic strains. Therefore, universal flu vaccine approaches that induce broad immunity against conserved parts of influenza have top priority in research. Cross-reactive T cell responses, especially tissue-resident memory T cells in the respiratory tract, provide efficient heterologous immunity, and must therefore be a key component of universal flu vaccines. Here, we review recent findings about T cell-based flu immunity, with an emphasis on tissue-resident memory T cells in the respiratory tract of humans and different animal models. Furthermore, we provide an update on preclinical and clinical studies evaluating T cell-evoking flu vaccines, and discuss the implementation of T cell immunity in real-life vaccine policies.
Collapse
|
120
|
Fisher E, Padula L, Podack K, O’Neill K, Seavey MM, Jayaraman P, Jasuja R, Strbo N. Induction of SARS-CoV-2 Protein S-Specific CD8+ T Cells in the Lungs of gp96-Ig-S Vaccinated Mice. Front Immunol 2021; 11:602254. [PMID: 33584668 PMCID: PMC7873992 DOI: 10.3389/fimmu.2020.602254] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 12/17/2020] [Indexed: 01/08/2023] Open
Abstract
Given the aggressive spread of COVID-19-related deaths, there is an urgent public health need to support the development of vaccine candidates to rapidly improve the available control measures against SARS-CoV-2. To meet this need, we are leveraging our existing vaccine platform to target SARS-CoV-2. Here, we generated cellular heat shock chaperone protein, glycoprotein 96 (gp96), to deliver SARS-CoV-2 protein S (spike) to the immune system and to induce cell-mediated immune responses. We showed that our vaccine platform effectively stimulates a robust cellular immune response against protein S. Moreover, we confirmed that gp96-Ig, secreted from allogeneic cells expressing full-length protein S, generates powerful, protein S polyepitope-specific CD4+ and CD8+ T cell responses in both lung interstitium and airways. These findings were further strengthened by the observation that protein-S -specific CD8+ T cells were induced in human leukocyte antigen HLA-A2.1 transgenic mice thus providing encouraging translational data that the vaccine is likely to work in humans, in the context of SARS-CoV-2 antigen presentation.
Collapse
Affiliation(s)
- Eva Fisher
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Laura Padula
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Kristin Podack
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Katelyn O’Neill
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL, United States
| | | | | | - Rahul Jasuja
- Heat Biologics, Inc., Morrisville, NC, United States
| | - Natasa Strbo
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL, United States
| |
Collapse
|
121
|
Mara K, Dai M, Brice AM, Alexander MR, Tribolet L, Layton DS, Bean AGD. Investigating the Interaction between Negative Strand RNA Viruses and Their Hosts for Enhanced Vaccine Development and Production. Vaccines (Basel) 2021; 9:vaccines9010059. [PMID: 33477334 PMCID: PMC7830660 DOI: 10.3390/vaccines9010059] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Accepted: 01/13/2021] [Indexed: 11/30/2022] Open
Abstract
The current pandemic has highlighted the ever-increasing risk of human to human spread of zoonotic pathogens. A number of medically-relevant zoonotic pathogens are negative-strand RNA viruses (NSVs). NSVs are derived from different virus families. Examples like Ebola are known for causing severe symptoms and high mortality rates. Some, like influenza, are known for their ease of person-to-person transmission and lack of pre-existing immunity, enabling rapid spread across many countries around the globe. Containment of outbreaks of NSVs can be difficult owing to their unpredictability and the absence of effective control measures, such as vaccines and antiviral therapeutics. In addition, there remains a lack of essential knowledge of the host–pathogen response that are induced by NSVs, particularly of the immune responses that provide protection. Vaccines are the most effective method for preventing infectious diseases. In fact, in the event of a pandemic, appropriate vaccine design and speed of vaccine supply is the most critical factor in protecting the population, as vaccination is the only sustainable defense. Vaccines need to be safe, efficient, and cost-effective, which is influenced by our understanding of the host–pathogen interface. Additionally, some of the major challenges of vaccines are the establishment of a long-lasting immunity offering cross protection to emerging strains. Although many NSVs are controlled through immunisations, for some, vaccine design has failed or efficacy has proven unreliable. The key behind designing a successful vaccine is understanding the host–pathogen interaction and the host immune response towards NSVs. In this paper, we review the recent research in vaccine design against NSVs and explore the immune responses induced by these viruses. The generation of a robust and integrated approach to development capability and vaccine manufacture can collaboratively support the management of outbreaking NSV disease health risks.
Collapse
|
122
|
Reizine F, Lesouhaitier M, Gregoire M, Pinceaux K, Gacouin A, Maamar A, Painvin B, Camus C, Le Tulzo Y, Tattevin P, Revest M, Le Bot A, Ballerie A, Cador-Rousseau B, Lederlin M, Lebouvier T, Launey Y, Latour M, Verdy C, Rossille D, Le Gallou S, Dulong J, Moreau C, Bendavid C, Roussel M, Cogne M, Tarte K, Tadié JM. SARS-CoV-2-Induced ARDS Associates with MDSC Expansion, Lymphocyte Dysfunction, and Arginine Shortage. J Clin Immunol 2021; 41:515-525. [PMID: 33387156 PMCID: PMC7775842 DOI: 10.1007/s10875-020-00920-5] [Citation(s) in RCA: 88] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Accepted: 11/12/2020] [Indexed: 11/20/2022]
Abstract
Purpose The SARS-CoV-2 infection can lead to a severe acute respiratory distress syndrome (ARDS) with prolonged mechanical ventilation and high mortality rate. Interestingly, COVID-19-associated ARDS share biological and clinical features with sepsis-associated immunosuppression since lymphopenia and acquired infections associated with late mortality are frequently encountered. Mechanisms responsible for COVID-19-associated lymphopenia need to be explored since they could be responsible for delayed virus clearance and increased mortality rate among intensive care unit (ICU) patients. Methods A series of 26 clinically annotated COVID-19 patients were analyzed by thorough phenotypic and functional investigations at days 0, 4, and 7 after ICU admission. Results We revealed that, in the absence of any difference in demographic parameters nor medical history between the two groups, ARDS patients presented with an increased number of myeloid-derived suppressor cells (MDSC) and a decreased number of CD8pos effector memory cell compared to patients hospitalized for COVID-19 moderate pneumonia. Interestingly, COVID-19-related MDSC expansion was directly correlated to lymphopenia and enhanced arginase activity. Lastly, T cell proliferative capacity in vitro was significantly reduced among COVID-19 patients and could be restored through arginine supplementation. Conclusions The present study reports a critical role for MDSC in COVID-19-associated ARDS. Our findings open the possibility of arginine supplementation as an adjuvant therapy for these ICU patients, aiming to reduce immunosuppression and help virus clearance, thereby decreasing the duration of mechanical ventilation, nosocomial infection acquisition, and mortality. Supplementary Information The online version contains supplementary material available at 10.1007/s10875-020-00920-5.
Collapse
Affiliation(s)
- Florian Reizine
- Infectious Diseases and Intensive Care Unit, Pontchaillou University Hospital, 2 rue Henri Le Guilloux, 35033, Rennes, France.,SITI, Pole de Biologie, Pontchaillou University Hospital, 2 rue Henri Le Guilloux, 35033, Rennes, France.,UMR 1236, University of Rennes, INSERM, Etablissement Français du Sang Bretagne, Rennes, France
| | - Mathieu Lesouhaitier
- Infectious Diseases and Intensive Care Unit, Pontchaillou University Hospital, 2 rue Henri Le Guilloux, 35033, Rennes, France.,SITI, Pole de Biologie, Pontchaillou University Hospital, 2 rue Henri Le Guilloux, 35033, Rennes, France.,UMR 1236, University of Rennes, INSERM, Etablissement Français du Sang Bretagne, Rennes, France
| | - Murielle Gregoire
- SITI, Pole de Biologie, Pontchaillou University Hospital, 2 rue Henri Le Guilloux, 35033, Rennes, France.,UMR 1236, University of Rennes, INSERM, Etablissement Français du Sang Bretagne, Rennes, France
| | - Kieran Pinceaux
- Infectious Diseases and Intensive Care Unit, Pontchaillou University Hospital, 2 rue Henri Le Guilloux, 35033, Rennes, France
| | - Arnaud Gacouin
- Infectious Diseases and Intensive Care Unit, Pontchaillou University Hospital, 2 rue Henri Le Guilloux, 35033, Rennes, France
| | - Adel Maamar
- Infectious Diseases and Intensive Care Unit, Pontchaillou University Hospital, 2 rue Henri Le Guilloux, 35033, Rennes, France
| | - Benoit Painvin
- Infectious Diseases and Intensive Care Unit, Pontchaillou University Hospital, 2 rue Henri Le Guilloux, 35033, Rennes, France
| | - Christophe Camus
- Infectious Diseases and Intensive Care Unit, Pontchaillou University Hospital, 2 rue Henri Le Guilloux, 35033, Rennes, France
| | - Yves Le Tulzo
- Infectious Diseases and Intensive Care Unit, Pontchaillou University Hospital, 2 rue Henri Le Guilloux, 35033, Rennes, France
| | - Pierre Tattevin
- Infectious Diseases and Intensive Care Unit, Pontchaillou University Hospital, 2 rue Henri Le Guilloux, 35033, Rennes, France
| | - Matthieu Revest
- Infectious Diseases and Intensive Care Unit, Pontchaillou University Hospital, 2 rue Henri Le Guilloux, 35033, Rennes, France
| | - Audrey Le Bot
- Infectious Diseases and Intensive Care Unit, Pontchaillou University Hospital, 2 rue Henri Le Guilloux, 35033, Rennes, France
| | - Alice Ballerie
- Department of Internal Medicine and Clinical Immunology, Pontchaillou University Hospital, 2 rue Henri Le Guilloux, 35033, Rennes, France
| | - Berengère Cador-Rousseau
- Department of Internal Medicine and Clinical Immunology, Pontchaillou University Hospital, 2 rue Henri Le Guilloux, 35033, Rennes, France
| | - Mathieu Lederlin
- Department of Radiology, Pontchaillou University Hospital, 2 rue Henri Le Guilloux, 35033, Rennes, France
| | - Thomas Lebouvier
- Department of Anesthesia and Critical Care, Pontchaillou University Hospital, 2 rue Henri Le Guilloux, 35033, Rennes, France
| | - Yoann Launey
- Department of Anesthesia and Critical Care, Pontchaillou University Hospital, 2 rue Henri Le Guilloux, 35033, Rennes, France
| | - Maelle Latour
- SITI, Pole de Biologie, Pontchaillou University Hospital, 2 rue Henri Le Guilloux, 35033, Rennes, France.,UMR 1236, University of Rennes, INSERM, Etablissement Français du Sang Bretagne, Rennes, France
| | - Clotilde Verdy
- SITI, Pole de Biologie, Pontchaillou University Hospital, 2 rue Henri Le Guilloux, 35033, Rennes, France.,UMR 1236, University of Rennes, INSERM, Etablissement Français du Sang Bretagne, Rennes, France
| | - Delphine Rossille
- SITI, Pole de Biologie, Pontchaillou University Hospital, 2 rue Henri Le Guilloux, 35033, Rennes, France.,UMR 1236, University of Rennes, INSERM, Etablissement Français du Sang Bretagne, Rennes, France
| | - Simon Le Gallou
- SITI, Pole de Biologie, Pontchaillou University Hospital, 2 rue Henri Le Guilloux, 35033, Rennes, France.,UMR 1236, University of Rennes, INSERM, Etablissement Français du Sang Bretagne, Rennes, France
| | - Joelle Dulong
- SITI, Pole de Biologie, Pontchaillou University Hospital, 2 rue Henri Le Guilloux, 35033, Rennes, France.,UMR 1236, University of Rennes, INSERM, Etablissement Français du Sang Bretagne, Rennes, France
| | - Caroline Moreau
- Department of Biochemistry, Pontchaillou University Hospital, 2 rue Henri Le Guilloux, 35033, Rennes, France
| | - Claude Bendavid
- Department of Biochemistry, Pontchaillou University Hospital, 2 rue Henri Le Guilloux, 35033, Rennes, France
| | - Mikael Roussel
- SITI, Pole de Biologie, Pontchaillou University Hospital, 2 rue Henri Le Guilloux, 35033, Rennes, France.,UMR 1236, University of Rennes, INSERM, Etablissement Français du Sang Bretagne, Rennes, France
| | - Michel Cogne
- SITI, Pole de Biologie, Pontchaillou University Hospital, 2 rue Henri Le Guilloux, 35033, Rennes, France.,UMR 1236, University of Rennes, INSERM, Etablissement Français du Sang Bretagne, Rennes, France
| | - Karin Tarte
- SITI, Pole de Biologie, Pontchaillou University Hospital, 2 rue Henri Le Guilloux, 35033, Rennes, France.,UMR 1236, University of Rennes, INSERM, Etablissement Français du Sang Bretagne, Rennes, France
| | - Jean-Marc Tadié
- Infectious Diseases and Intensive Care Unit, Pontchaillou University Hospital, 2 rue Henri Le Guilloux, 35033, Rennes, France. .,SITI, Pole de Biologie, Pontchaillou University Hospital, 2 rue Henri Le Guilloux, 35033, Rennes, France. .,UMR 1236, University of Rennes, INSERM, Etablissement Français du Sang Bretagne, Rennes, France. .,Service des Maladies Infectieuses et Réanimation Médicale, Hôpital Pontchaillou, 2 rue Henri Le Guilloux, 35033, Rennes Cedex 9, France.
| |
Collapse
|
123
|
Cosgrove PR, Redhu NS, Tang Y, Monuteaux MC, Horwitz BH. Characterizing T cell subsets in the nasal mucosa of children with acute respiratory symptoms. Pediatr Res 2021; 90:1023-1030. [PMID: 33504970 PMCID: PMC7838854 DOI: 10.1038/s41390-021-01364-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Revised: 11/21/2020] [Accepted: 01/05/2021] [Indexed: 01/30/2023]
Abstract
BACKGROUND In infants admitted to an ICU with respiratory failure, there is an association between the ratio of CD8+ to CD4+ T cells within the upper respiratory tract and disease severity. Whether this ratio is associated with respiratory disease severity within children presenting to a pediatric emergency department is not known. METHODS We studied a convenience sample of 63 children presenting to a pediatric emergency department with respiratory symptoms. T cell subsets in the nasal mucosa were analyzed by flow cytometry. We compared CD4+ and CD8+ T cells subsets in these samples and analyzed the proportion of these subsets that expressed markers associated with tissue residency. RESULTS We were able to identify major subsets of CD8 and CD4 T cells within the nasal mucosa using flocked swabs. We found no difference in the ratio CD8+ to CD4+ T cells in children with upper or lower respiratory illness. A positive association between tissue-resident memory T cell frequency and patient age was identified. CONCLUSIONS In our patient populations, the CD8+:CD4+ ratio was not associated with disease severity. The majority of T cells collected on nasal swabs are antigen experienced, and there is an association between the frequency of tissue-resident T cells and age. IMPACT Immune cell populations from the nasal mucosa can be captured using flocked nasal swabs and analyzed by flow cytometry. Nasal CD8+:CD4+ ratio does not predict respiratory illness severity in children presenting to the emergency department. The frequency of CD8+ and CD4+ resident memory T cells within the nasal mucosa increases with age.
Collapse
Affiliation(s)
- Peter R. Cosgrove
- grid.2515.30000 0004 0378 8438Division of Emergency Medicine, Boston Children’s Hospital, Boston, MA USA ,grid.38142.3c000000041936754XDepartment of Pediatrics, Harvard Medical School, Boston, MA USA
| | - Naresh S. Redhu
- grid.2515.30000 0004 0378 8438Division of Gastroenterology, Hepatology, and Nutrition, Boston Children’s Hospital, Boston, MA USA ,Present Address: Morphic Therapeutic, Waltham, MA USA
| | - Ying Tang
- grid.2515.30000 0004 0378 8438Division of Gastroenterology, Hepatology, and Nutrition, Boston Children’s Hospital, Boston, MA USA
| | - Michael C. Monuteaux
- grid.2515.30000 0004 0378 8438Division of Emergency Medicine, Boston Children’s Hospital, Boston, MA USA ,grid.38142.3c000000041936754XDepartment of Pediatrics, Harvard Medical School, Boston, MA USA
| | - Bruce H. Horwitz
- grid.2515.30000 0004 0378 8438Division of Emergency Medicine, Boston Children’s Hospital, Boston, MA USA ,grid.38142.3c000000041936754XDepartment of Pediatrics, Harvard Medical School, Boston, MA USA
| |
Collapse
|
124
|
Armentrout EI, Liu GY, Martins GA. T Cell Immunity and the Quest for Protective Vaccines against Staphylococcus aureus Infection. Microorganisms 2020; 8:microorganisms8121936. [PMID: 33291260 PMCID: PMC7762175 DOI: 10.3390/microorganisms8121936] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 11/27/2020] [Accepted: 11/30/2020] [Indexed: 12/14/2022] Open
Abstract
Staphylococcus aureus is a wide-spread human pathogen, and one of the top causative agents of nosocomial infections. The prevalence of antibiotic-resistant S. aureus strains, which are associated with higher mortality and morbidity rates than antibiotic-susceptible strains, is increasing around the world. Vaccination would be an effective preventive measure against S. aureus infection, but to date, every vaccine developed has failed in clinical trials, despite inducing robust antibody responses. These results suggest that induction of humoral immunity does not suffice to confer protection against the infection. Evidence from studies in murine models and in patients with immune defects support a role of T cell-mediated immunity in protective responses against S. aureus. Here, we review the current understanding of the mechanisms underlying adaptive immunity to S. aureus infections and discuss these findings in light of the recent S. aureus vaccine trial failures. We make the case for the need to develop anti-S. aureus vaccines that can specifically elicit robust and durable protective memory T cell subsets.
Collapse
Affiliation(s)
- Erin I. Armentrout
- Lung Institute, Cedars-Sinai Medical Center (CSMC), Los Angeles, CA 90048, USA;
- Division of Pulmonary and Critical Care Medicine, CSMC, Los Angeles, CA 90048, USA
| | - George Y. Liu
- Collaborative to Halt Antibiotic-Resistant Microbes, University of California, San Diego, La Jolla, CA 92161, USA;
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
| | - Gislâine A. Martins
- F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute (IBIRI), CSMC, Los Angeles, CA 90048, USA
- Department of Biomedical Sciences, Research Division of Immunology, CSMC, Los Angeles, CA 90048, USA
- Department of Medicine, Division of Gastroenterology, CSMC, Los Angeles, CA 90048, USA
- Correspondence:
| |
Collapse
|
125
|
Migueles SA, Rogan DC, Gavil NV, Kelly EP, Toulmin SA, Wang LT, Lack J, Ward AJ, Pryal PF, Ludwig AK, Medina RG, Apple BJ, Toumanios CN, Poole AL, Rehm CA, Jones SE, Liang CJ, Connors M. Antigenic Restimulation of Virus-Specific Memory CD8 + T Cells Requires Days of Lytic Protein Accumulation for Maximal Cytotoxic Capacity. J Virol 2020; 94:e01595-20. [PMID: 32907983 PMCID: PMC7654275 DOI: 10.1128/jvi.01595-20] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 09/05/2020] [Indexed: 02/07/2023] Open
Abstract
In various infections or vaccinations of mice or humans, reports of the persistence and the requirements for restimulation of the cytotoxic mediators granzyme B (GrB) and perforin (PRF) in CD8+ T cells have yielded disparate results. In this study, we examined the kinetics of PRF and GrB mRNA and protein expression after stimulation and associated changes in cytotoxic capacity in virus-specific memory cells in detail. In patients with controlled HIV or cleared respiratory syncytial virus (RSV) or influenza virus infections, all virus-specific CD8+ T cells expressed low PRF levels without restimulation. Following stimulation, they displayed similarly delayed kinetics for lytic protein expression, with significant increases occurring by days 1 to 3 before peaking on days 4 to 6. These increases were strongly correlated with, but were not dependent upon, proliferation. Incremental changes in PRF and GrB percent expression and mean fluorescence intensity (MFI) were highly correlated with increases in HIV-specific cytotoxicity. mRNA levels in HIV-specific CD8+ T-cells exhibited delayed kinetics after stimulation as with protein expression, peaking on day 5. In contrast to GrB, PRF mRNA transcripts were little changed over 5 days of stimulation (94-fold versus 2.8-fold, respectively), consistent with posttranscriptional regulation. Changes in expression of some microRNAs, including miR-17, miR-150, and miR-155, suggested that microRNAs might play a significant role in regulation of PRF expression. Therefore, under conditions of extremely low or absent antigen levels, memory virus-specific CD8+ T cells require prolonged stimulation over days to achieve maximal lytic protein expression and cytotoxic capacity.IMPORTANCE Antigen-specific CD8+ T cells play a major role in controlling most virus infections, primarily by perforin (PRF)- and granzyme B (GrB)-mediated apoptosis. There is considerable controversy regarding whether PRF is constitutively expressed, rapidly increased similarly to a cytokine, or delayed in its expression with more prolonged stimulation in virus-specific memory CD8+ T cells. In this study, the degree of cytotoxic capacity of virus-specific memory CD8+ T cells was directly proportional to the content of lytic molecules, which required antigenic stimulation over several days for maximal levels. This appeared to be modulated by increases in GrB transcription and microRNA-mediated posttranscriptional regulation of PRF expression. Clarifying the requirements for maximal cytotoxic capacity is critical to understanding how viral clearance might be mediated by memory cells and what functions should be induced by vaccines and immunotherapies.
Collapse
Affiliation(s)
- Stephen A Migueles
- HIV-Specific Immunity Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Daniel C Rogan
- HIV-Specific Immunity Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Noah V Gavil
- HIV-Specific Immunity Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Elizabeth P Kelly
- HIV-Specific Immunity Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Sushila A Toulmin
- HIV-Specific Immunity Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Lawrence T Wang
- HIV-Specific Immunity Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Justin Lack
- NIAID Collaborative Bioinformatics Resource (NCBR), Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, Maryland, USA
| | - Addison J Ward
- HIV-Specific Immunity Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Patrick F Pryal
- HIV-Specific Immunity Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Amanda K Ludwig
- HIV-Specific Immunity Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Renata G Medina
- HIV-Specific Immunity Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Benjamin J Apple
- HIV-Specific Immunity Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Christina N Toumanios
- HIV-Specific Immunity Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - April L Poole
- HIV-Specific Immunity Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Catherine A Rehm
- HIV-Specific Immunity Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Sara E Jones
- Clinical Research Program Directorate, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, Maryland, USA
| | - C Jason Liang
- Biostatistics Research Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Mark Connors
- HIV-Specific Immunity Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
126
|
Stephens LM, Varga SM. Nanoparticle vaccines against respiratory syncytial virus. Future Virol 2020; 15:763-778. [PMID: 33343684 PMCID: PMC7737143 DOI: 10.2217/fvl-2020-0174] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 11/11/2020] [Indexed: 12/13/2022]
Abstract
Respiratory syncytial virus (RSV) is a leading cause of respiratory disease in infants, the elderly and immunocompromised individuals. Despite the global burden, there is no licensed vaccine for RSV. Recent advances in the use of nanoparticle technology have provided new opportunities to address some of the limitations of conventional vaccines. Precise control over particle size and surface properties enhance antigen stability and prolong antigen release. Particle size can also be modified to target specific antigen-presenting cells in order to induce specific types of effector T-cell responses. Numerous nanoparticle-based vaccines are currently being evaluated for RSV including inorganic, polymeric and virus-like particle-based formulations. Here, we review the potential advantages of using different nanoparticle formulations in a vaccine for RSV, and discuss many examples of safe, and effective vaccines currently in both preclinical and clinical stages of testing.
Collapse
Affiliation(s)
- Laura M Stephens
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA 52242, USA
| | - Steven M Varga
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA 52242, USA
- Department of Microbiology & Immunology, University of Iowa, Iowa City, IA 52242, USA
- Department of Pathology, University of Iowa, Iowa City, IA 52242, USA
| |
Collapse
|
127
|
Paulsen M, Varese A, Pinpathomrat N, Kirsebom FCM, Paulsen M, Johansson C. MAVS Deficiency Is Associated With a Reduced T Cell Response Upon Secondary RSV Infection in Mice. Front Immunol 2020; 11:572747. [PMID: 33123150 PMCID: PMC7573121 DOI: 10.3389/fimmu.2020.572747] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 09/16/2020] [Indexed: 11/17/2022] Open
Abstract
Infections with respiratory syncytial virus (RSV) occurs repeatedly throughout life because sustained, protective memory responses fail to develop. Why this occurs is not known. During RSV infection the recognition of the virus via the cytosolic RIG-I like receptors and signaling via the adaptor protein MAVS is crucial for mounting an innate immune response. However, if this signaling pathway is important for T cell responses during primary infection and during re-infection is not fully elucidated. We describe a second peak of pro-inflammatory mediators during the primary immune response to RSV that coincides with the arrival of T cells into the lung. This second peak of cytokines/chemokines is regulated differently than the early peak and is largely independent of signaling via MAVS. This was concurrent with Mavs−/− mice mounting a strong T cell response to primary RSV infection, with robust IFN-γ; and Granzyme B production. However, after RSV re-infection, Mavs−/− mice showed fewer CD4+ and CD8+ short term memory T cells and their capacity to produce IFN-γ; and Granzyme B, was decreased. In sum, cytosolic recognition of RSV is important not only for initiating innate anti-viral responses but also for generating or maintaining efficient, short term T cell memory responses.
Collapse
Affiliation(s)
- Michelle Paulsen
- Respiratory Infections Section, St Mary's Campus, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Augusto Varese
- Respiratory Infections Section, St Mary's Campus, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Nawamin Pinpathomrat
- Respiratory Infections Section, St Mary's Campus, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Freja C M Kirsebom
- Respiratory Infections Section, St Mary's Campus, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Malte Paulsen
- Respiratory Infections Section, St Mary's Campus, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Cecilia Johansson
- Respiratory Infections Section, St Mary's Campus, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| |
Collapse
|
128
|
Habibi MS, Thwaites RS, Chang M, Jozwik A, Paras A, Kirsebom F, Varese A, Owen A, Cuthbertson L, James P, Tunstall T, Nickle D, Hansel TT, Moffatt MF, Johansson C, Chiu C, Openshaw PJM. Neutrophilic inflammation in the respiratory mucosa predisposes to RSV infection. Science 2020; 370:eaba9301. [PMID: 33033192 PMCID: PMC7613218 DOI: 10.1126/science.aba9301] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 06/30/2020] [Accepted: 08/19/2020] [Indexed: 12/12/2022]
Abstract
The variable outcome of viral exposure is only partially explained by known factors. We administered respiratory syncytial virus (RSV) to 58 volunteers, of whom 57% became infected. Mucosal neutrophil activation before exposure was highly predictive of symptomatic RSV disease. This was associated with a rapid, presymptomatic decline in mucosal interleukin-17A (IL-17A) and other mediators. Conversely, those who resisted infection showed presymptomatic activation of IL-17- and tumor necrosis factor-related pathways. Vulnerability to infection was not associated with baseline microbiome but was reproduced in mice by preinfection chemokine-driven airway recruitment of neutrophils, which caused enhanced disease mediated by pulmonary CD8+ T cell infiltration. Thus, mucosal neutrophilic inflammation at the time of RSV exposure enhances susceptibility, revealing dynamic, time-dependent local immune responses before symptom onset and explaining the as-yet unpredictable outcomes of pathogen exposure.
Collapse
Affiliation(s)
| | - Ryan S Thwaites
- National Heart and Lung Institute, Imperial College London, London, UK
| | | | - Agnieszka Jozwik
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Allan Paras
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Freja Kirsebom
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Augusto Varese
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Amber Owen
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Leah Cuthbertson
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Phillip James
- National Heart and Lung Institute, Imperial College London, London, UK
| | | | - David Nickle
- Genetics & Pharmacogenomics, Department of Translational Medicine, Merck & Co., Inc., Boston, MA, USA
| | - Trevor T Hansel
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Miriam F Moffatt
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Cecilia Johansson
- National Heart and Lung Institute, Imperial College London, London, UK.
| | - Christopher Chiu
- Department of Infectious Disease, Imperial College London, London, UK.
| | | |
Collapse
|
129
|
Wiseman DJ, Thwaites RS, Drysdale SB, Janet S, Donaldson GC, Wedzicha JA, Openshaw PJ. Immunological and Inflammatory Biomarkers of Susceptibility and Severity in Adult Respiratory Syncytial Virus Infections. J Infect Dis 2020; 222:S584-S591. [PMID: 32227102 DOI: 10.1093/infdis/jiaa063] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Respiratory syncytial virus (RSV) is the most common cause of bronchiolitis in young infants. However, it is also a significant pathogen in older adults. Validated biomarkers of RSV disease severity would benefit diagnostics, treatment decisions, and prophylactic interventions. This review summarizes knowledge of biomarkers for RSV disease in adults. METHODS A literature review was performed using Ovid Medline, Embase, Global health, Scopus, and Web of Science for articles published 1946-October 2016. Nine articles were identified plus 9 from other sources. RESULTS From observational studies of natural infection and challenge studies in volunteers, biomarkers of RSV susceptibility or disease severity in adults were: (1) lower anti-RSV neutralizing antibodies, where neutralizing antibody (and local IgA) may be a correlate of susceptibility/severity; (2) RSV-specific CD8+ T cells in bronchoalveolar lavage fluid preinfection (subjects with higher levels had less severe illness); and (3) elevated interleukin-6 (IL-6), IL-8, and myeloperoxidase levels in the airway are indicative of severe infection. CONCLUSIONS Factors determining susceptibility to and severity of RSV disease in adults have not been well defined. Respiratory mucosal antibodies and CD8+ T cells appear to contribute to preventing infection and modulation of disease severity. Studies of RSV pathogenesis in at-risk populations are needed.
Collapse
Affiliation(s)
| | - Ryan S Thwaites
- National Heart and Lung Institute, Imperial College, London, UK
| | - Simon B Drysdale
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK.,National Institute for Health Research Oxford Biomedical Research Centre, Children's Hospital, Oxford, UK.,Institute for Infection and Immunity, St George's, University of London, London, UK
| | - Sophie Janet
- National Institute for Health Research Oxford Biomedical Research Centre, Children's Hospital, Oxford, UK
| | | | | | | | | |
Collapse
|
130
|
Stambas J, Lu C, Tripp RA. Innate and adaptive immune responses in respiratory virus infection: implications for the clinic. Expert Rev Respir Med 2020; 14:1141-1147. [PMID: 32762572 DOI: 10.1080/17476348.2020.1807945] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
INTRODUCTION The innate immune response is the first line of defense and consists of physical, chemical and cellular defenses. The adaptive immune response is the second line of defense and is pathogen-specific. Innate immunity occurs immediately while adaptive immunity develops upon pathogen exposure, and is long-lasting, highly specific, and sustained by memory T cells. Respiratory virus infection typically induces effective immunity but over-exuberant responses are associated with pathophysiology. Cytokines expressed in response to viral infection can enhance biological responses, activate, and trigger signaling pathways leading to adaptive immunity Vaccines induce immunity, specifically B and T cell responses. Vaccination is generally efficacious, but for many viruses, our understanding of vaccination strategies and immunity is incomplete or in its infancy. Studies that examine innate and adaptive immune responses to respiratory virus infection will aid vaccine development and may reduce the burden of respiratory viral disease. AREAS COVERED A literature search was performed using PubMed. The search covered: innate, adaptive, respiratory virus, vaccine development, B cell, and T cell. EXPERT OPINION Immunity rests on two pillars, i.e. the innate and adaptive immune system, which function together on different tasks to maintain homeostasis. a better understanding of immunity is necessary for disease prevention and intervention.
Collapse
Affiliation(s)
- John Stambas
- School of Medicine, Deakin University , Melbourne, Australia
| | - Chunni Lu
- School of Medicine, Deakin University , Melbourne, Australia
| | - Ralph A Tripp
- Department of Infectious Diseases, University of Georgia , Athens, GA, USA
| |
Collapse
|
131
|
Jordan E, Lawrence SJ, Meyer TPH, Schmidt D, Schultz S, Mueller J, Stroukova D, Koenen B, Gruenert R, Silbernagl G, Vidojkovic S, Chen LM, Weidenthaler H, Samy N, Chaplin P. Broad Antibody and Cellular Immune Response From a Phase 2 Clinical Trial With a Novel Multivalent Poxvirus-Based Respiratory Syncytial Virus Vaccine. J Infect Dis 2020; 223:1062-1072. [PMID: 32726422 DOI: 10.1093/infdis/jiaa460] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 07/24/2020] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Respiratory syncytial virus (RSV) is a major cause of severe respiratory disease in young children and the elderly. Protective immunity is not generated after repeated infections, but vaccination may hopefully prove effective. METHODS This phase 2 clinical study investigated a multivalent RSV vaccine (MVA-BN-RSV) designed to induce broad antibody and cellular immune responses by encoding RSV surface proteins F, G (for both A and B subtypes), and internal antigens (M2, N). This study evaluated the immune response in adults aged ≥55 years to identify the optimal MVA-BN-RSV dose and vaccination schedule. RESULTS A single dose increased the levels of neutralizing (plaque reduction neutralization test to RSV A and B) and total (IgG and IgA ELISA) antibodies (1.6 to 3.4-fold increase from baseline) and induced a broad Th1-biased cellular immune response (interferon-γ ELISPOT) to all 5 vaccine inserts (5.4 to 9.7-fold increases). Antibody responses remained above baseline for 6 months. A 12-month booster dose elicited a booster effect in antibody and T-cell responses (up to 2.8-fold from preboost levels). No drug-related serious adverse events were reported. CONCLUSIONS MVA-BN-RSV induces a broad immune response that persists at least 6 months and can be boosted at 12 months, without significant safety findings. CLINICAL TRIALS REGISTRATION NCT02873286.
Collapse
Affiliation(s)
| | - Steven J Lawrence
- Division of Infectious Diseases, Washington University School of Medicine, St Louis, Missouri, USA
| | | | | | | | | | | | | | | | | | | | - Liddy M Chen
- Bavarian Nordic Inc., Morrisville, North Carolina, USA
| | | | | | | |
Collapse
|
132
|
Respiratory Syncytial Virus Infection Induces Chromatin Remodeling to Activate Growth Factor and Extracellular Matrix Secretion Pathways. Viruses 2020; 12:v12080804. [PMID: 32722537 PMCID: PMC7472097 DOI: 10.3390/v12080804] [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: 06/23/2020] [Revised: 07/17/2020] [Accepted: 07/23/2020] [Indexed: 12/12/2022] Open
Abstract
Lower respiratory tract infection (LRTI) with respiratory syncytial virus (RSV) is associated with reduced lung function through unclear mechanisms. In this study, we test the hypothesis that RSV infection induces genomic reprogramming of extracellular matrix remodeling pathways. For this purpose, we sought to identify transcriptionally active open chromatin domains using assay for transposase-accessible-next generation sequencing (ATAC-Seq) in highly differentiated lower airway epithelial cells. High confidence nucleosome-free regions were those predicted independently using two peak-calling algorithms. In uninfected cells, ~12,650 high-confidence open chromatin regions were identified. These mapped to ~8700 gene bodies, whose genes functionally controlled organelle synthesis and Th2 pathways (IL6, TSLP). These latter cytokines are preferentially secreted by RSV-infected bronchiolar cells and linked to mucous production, obstruction, and atopy. By contrast, in RSV infection, we identify ~1700 high confidence open chromatin domains formed in 1120 genes, primarily in introns. These induced chromatin modifications are associated with complex gene expression profiles controlling tyrosine kinase growth factor signaling and extracellular matrix (ECM) secretory pathways. Of these, RSV induces formation of nucleosome-free regions on TGFB1/JUNB//FN1/MMP9 genes and the rate limiting enzyme in the hexosamine biosynthetic pathway (HBP), Glutamine-Fructose-6-Phosphate Transaminase 2 (GFPT2). RSV-induced open chromatin domains are highly enriched in AP1 binding motifs and overlap experimentally determined JUN peaks in GEO ChIP-Seq data sets. Our results provide a topographical map of chromatin accessibility and suggest a growth factor and AP1-dependent mechanism for upregulation of the HBP and ECM remodeling in lower epithelial cells that may be linked to long-term airway remodeling.
Collapse
|
133
|
Matyushenko V, Kotomina T, Kudryavtsev I, Mezhenskaya D, Prokopenko P, Matushkina A, Sivak K, Muzhikyan A, Rudenko L, Isakova-Sivak I. Conserved T-cell epitopes of respiratory syncytial virus (RSV) delivered by recombinant live attenuated influenza vaccine viruses efficiently induce RSV-specific lung-localized memory T cells and augment influenza-specific resident memory T-cell responses. Antiviral Res 2020; 182:104864. [PMID: 32585323 PMCID: PMC7313889 DOI: 10.1016/j.antiviral.2020.104864] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 05/30/2020] [Accepted: 06/16/2020] [Indexed: 12/14/2022]
Abstract
Respiratory syncytial virus (RSV) can cause recurrent infection in people because it does not stimulate a long-lived immunological memory. There is an urgent need to develop a safe and efficacious vaccine against RSV that would induce immunological memory without causing immunopathology following natural RSV infection. We have previously generated two recombinant live attenuated influenza vaccine (LAIV) viruses that encode immunodominant T-cell epitopes of RSV M2 protein in the neuraminidase or NS1 genes. These chimeric vaccines afforded protection against influenza and RSV infection in mice, without causing pulmonary eosinophilia or inflammatory RSV disease. The current study assessed the formation of influenza-specific and RSV-specific CD4 and CD8 T-cell responses in the lungs of mice, with special attention to the lung tissue-resident memory T cell subsets (TRM). The RSV epitopes did not affect influenza-specific CD4 effector memory T cell (Tem) levels in the lungs. The majority of these cells formed by LAIV or LAIV-RSV viruses had CD69+CD103- phenotype. Both LAIV+NA/RSV and LAIV+NS/RSV recombinant viruses induced significant levels of RSV M282 epitope-specific lung-localized CD8 Tem cells expressing both CD69 and CD103 TRM markers. Surprisingly, the CD69+CD103+ influenza-specific CD8 Tem responses were augmented by the addition of RSV epitopes, possibly as a result of the local microenvironment formed by the RSV-specific memory T cells differentiating to TRM in the lungs of mice immunized with LAIV-RSV chimeric viruses. This study provides evidence that LAIV vector-based vaccination can induce robust lung-localized T-cell immunity to the inserted T-cell epitope of a foreign pathogen, without altering the immunogenicity of the viral vector itself. Two LAIV-RSV vaccine viruses induced RSV M282-specific effector memory CD8 T cells producing both IFNγ and TNFα cytokines. The inserted RSV epitopes did not affect influenza-specific CD4 Tem levels in the lungs of immunized mice. LAIV-RSV viruses induced RSV M282-specific lung-localized CD8 Tem cells expressing both CD69 and CD103 TRM markers. The magnitude of RSV M282-specific CD8 Tem responses correlates with protection against RSV-induced lung pathology. The addition of RSV epitopes into the LAIV strain augmented CD69+CD103+ influenza-specific CD8 Tem responses in the lungs.
Collapse
Affiliation(s)
- Victoria Matyushenko
- Department of Virology, Institute of Experimental Medicine, Saint Petersburg, Russia
| | - Tatiana Kotomina
- Department of Virology, Institute of Experimental Medicine, Saint Petersburg, Russia
| | - Igor Kudryavtsev
- Department of Immunology, Institute of Experimental Medicine, Saint Petersburg, Russia
| | - Daria Mezhenskaya
- Department of Virology, Institute of Experimental Medicine, Saint Petersburg, Russia
| | - Polina Prokopenko
- Department of Virology, Institute of Experimental Medicine, Saint Petersburg, Russia
| | - Anastasia Matushkina
- Department of Virology, Institute of Experimental Medicine, Saint Petersburg, Russia
| | - Konstantin Sivak
- Smorodintsev Research Institute of Influenza, Saint Petersburg, Russia
| | - Arman Muzhikyan
- Smorodintsev Research Institute of Influenza, Saint Petersburg, Russia
| | - Larisa Rudenko
- Department of Virology, Institute of Experimental Medicine, Saint Petersburg, Russia
| | - Irina Isakova-Sivak
- Department of Virology, Institute of Experimental Medicine, Saint Petersburg, Russia.
| |
Collapse
|
134
|
Mammas IN, Drysdale SB, Rath B, Theodoridou M, Papaioannou G, Papatheodoropoulou A, Koutsounaki E, Koutsaftiki C, Kozanidou E, Achtsidis V, Korovessi P, Chrousos GP, Spandidos DA. Update on current views and advances on RSV infection (Review). Int J Mol Med 2020; 46:509-520. [PMID: 32626981 PMCID: PMC7307844 DOI: 10.3892/ijmm.2020.4641] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 06/15/2020] [Indexed: 12/11/2022] Open
Abstract
Respiratory syncytial virus (RSV) infection represents an excellent paradigm of precision medicine in modern paediatrics and several clinical trials are currently performed in the prevention and management of RSV infection. A new taxonomic terminology for RSV was recently adopted, while the diagnostic and omics techniques have revealed new modalities in the early identification of RSV infections and for better understanding of the disease pathogenesis. Coordinated clinical and research efforts constitute an important step in limiting RSV global predominance, improving epidemiological surveillance, and advancing neonatal and paediatric care. This review article presents the key messages of the plenary lectures, oral presentations and posters of the '5th workshop on paediatric virology' (Sparta, Greece, 12th October 2019) organized by the Paediatric Virology Study Group, focusing on recent advances in the epidemiology, pathogenesis, diagnosis, prognosis, clinical management and prevention of RSV infection in childhood.
Collapse
Affiliation(s)
- Ioannis N Mammas
- Laboratory of Clinical Virology, School of Medicine, University of Crete, 71003 Heraklion, Greece
| | | | - Barbara Rath
- Vienna Vaccine Safety Initiative, D‑10437 Berlin, Germany
| | - Maria Theodoridou
- First Department of Paediatrics, University of Athens School of Medicine, 11527 Athens, Greece
| | - Georgia Papaioannou
- Department of Paediatric Radiology, 'Mitera' Children's Hospital, 15123 Athens, Greece
| | | | - Eirini Koutsounaki
- Neonatal Department, 'Alexandra' Maternity Hospital, 15123 Athens, Greece
| | - Chryssie Koutsaftiki
- Paediatric Intensive Care Unit (PICU), 'Penteli' Children's Hospital, 15236 Penteli, Greece
| | - Eleftheria Kozanidou
- 2nd Department of Internal Medicine, 'St Panteleimon' General Hospital of Nikaia, 18454 Piraeus, Greece
| | - Vassilis Achtsidis
- Department of Ophthalmology, Royal Cornwall Hospitals, Cornwall TR1 3LQ, UK
| | - Paraskevi Korovessi
- Department of Paediatrics, 'Penteli' Children's Hospital, 15236 Penteli, Greece
| | - George P Chrousos
- First Department of Paediatrics, University of Athens School of Medicine, 11527 Athens, Greece
| | - Demetrios A Spandidos
- Laboratory of Clinical Virology, School of Medicine, University of Crete, 71003 Heraklion, Greece
| |
Collapse
|
135
|
Pollard AJ, Sauerwein R, Baay M, Neels P. Third human challenge trial conference, Oxford, United Kingdom, February 6-7, 2020, a meeting report. Biologicals 2020; 66:41-52. [PMID: 32505512 DOI: 10.1016/j.biologicals.2020.04.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 04/27/2020] [Indexed: 02/06/2023] Open
Abstract
The third Human Challenge Trial Meeting brought together a broad range of international stakeholders, including academia, regulators, funders and industry, with a considerable delegation from Low- and Middle-Income Countries. Controlled human infection models (CHIMs) can be helpful to study pathogenesis and for the development of vaccines. As challenge agents are used to infect healthy volunteers, ethical considerations include that the challenge studies need to be safe and results should be meaningful. The meeting provided a state-of-the-art overview on a wide range of CHIMs, including viral, bacterial and parasitic challenge agents. Recommendations included globally aligned guidance documents for CHIM studies; further definition of a CHIM, based on the challenge agent used; standardization of methodology and study endpoints; capacity building in Low- and Middle-Income Countries, in performance as well as regulation of CHIM studies; guidance on compensation for participation in CHIM studies; and preparation of CHIM studies, with strong engagement with stakeholders.
Collapse
Affiliation(s)
- Andrew J Pollard
- Department of Paediatrics, University of Oxford, United Kingdom.
| | | | - Marc Baay
- P95 Epidemiology & Pharmacovigilance, Leuven, Belgium.
| | - Pieter Neels
- International Alliance for Biological Standardization, Belgium.
| | | |
Collapse
|
136
|
Morris SE, Farber DL, Yates AJ. Tissue-Resident Memory T Cells in Mice and Humans: Towards a Quantitative Ecology. THE JOURNAL OF IMMUNOLOGY 2020; 203:2561-2569. [PMID: 31685700 DOI: 10.4049/jimmunol.1900767] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 09/16/2019] [Indexed: 12/13/2022]
Abstract
In recent years, tissue-resident memory T cells (TRM) have emerged as essential components of immunological memory. Following antigenic challenge, TRM remain in nonlymphoid tissues and defend against re-exposure. Although accumulating evidence suggests important roles for TRM in mediating protective immunity, fundamental aspects of the population biology of TRM remain poorly understood. In this article, we discuss how results from different systems shed light on the ecological dynamics of TRM in mice and humans. We highlight the importance of dissecting processes contributing to TRM maintenance, and how these might vary across phenotypically and spatially heterogeneous subsets. We also discuss how the diversity of TRM communities within specific tissues may evolve under competition and in response to antigenic perturbation. Throughout, we illustrate how mathematical models can clarify inferences obtained from experimental data and help elucidate the homeostatic mechanisms underpinning the ecology of TRM populations.
Collapse
Affiliation(s)
- Sinead E Morris
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY 10032
| | - Donna L Farber
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY 10032.,Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY 10032; and.,Department of Surgery, Columbia University Medical Center, New York, NY 10032
| | - Andrew J Yates
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY 10032;
| |
Collapse
|
137
|
Brasier AR. RSV Reprograms the CDK9•BRD4 Chromatin Remodeling Complex to Couple Innate Inflammation to Airway Remodeling. Viruses 2020; 12:v12040472. [PMID: 32331282 PMCID: PMC7232410 DOI: 10.3390/v12040472] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 04/17/2020] [Accepted: 04/19/2020] [Indexed: 02/06/2023] Open
Abstract
Respiratory syncytial virus infection is responsible for seasonal upper and lower respiratory tract infections worldwide, causing substantial morbidity. Self-inoculation of the virus into the nasopharynx results in epithelial replication and distal spread into the lower respiratory tract. Here, respiratory syncytial virus (RSV) activates sentinel cells important in the host inflammatory response, resulting in epithelial-derived cytokine and interferon (IFN) expression resulting in neutrophilia, whose intensity is associated with disease severity. I will synthesize key findings describing how RSV replication activates intracellular NFκB and IRF signaling cascades controlling the innate immune response (IIR). Recent studies have implicated a central role for Scg1a1+ expressing progenitor cells in IIR, a cell type uniquely primed to induce neutrophilic-, T helper 2 (Th2)-polarizing-, and fibrogenic cytokines that play distinct roles in disease pathogenesis. Molecular studies have linked the positive transcriptional elongation factor-b (P-TEFb), a pleiotrophic chromatin remodeling complex in immediate-early IIR gene expression. Through intrinsic kinase activity of cyclin dependent kinase (CDK) 9 and atypical histone acetyl transferase activity of bromodomain containing protein 4 (BRD4), P-TEFb mediates transcriptional elongation of IIR genes. Unbiased proteomic studies show that the CDK9•BRD4 complex is dynamically reconfigured by the innate response and targets TGFβ-dependent fibrogenic gene networks. Chronic activation of CDK9•BRD4 mediates chromatin remodeling fibrogenic gene networks that cause epithelial mesenchymal transition (EMT). Mesenchymal transitioned epithelial cells elaborate TGFβ and IL6 that function in a paracrine manner to expand the population of subepithelial myofibroblasts. These findings may account for the long-term reduction in pulmonary function in children with severe lower respiratory tract infection (LRTI). Modifying chromatin remodeling properties of the CDK9•BRD4 coactivators may provide a mechanism for reducing post-infectious airway remodeling that are a consequence of severe RSV LRTIs.
Collapse
Affiliation(s)
- Allan R Brasier
- Institute for Clinical and Translational Research; University of Wisconsin-Madison School of Medicine and Public Health; Madison, WI 53705, USA
| |
Collapse
|
138
|
Szabo PA, Miron M, Farber DL. Location, location, location: Tissue resident memory T cells in mice and humans. Sci Immunol 2020; 4:4/34/eaas9673. [PMID: 30952804 DOI: 10.1126/sciimmunol.aas9673] [Citation(s) in RCA: 383] [Impact Index Per Article: 95.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 03/04/2019] [Indexed: 12/13/2022]
Abstract
The discovery of T cells resident in diverse tissues has altered our understanding of adaptive immunity to encompass site-specific responses mediated by tissue-adapted memory T cells throughout the body. Here, we discuss the key phenotypic, transcriptional, and functional features of these tissue-resident memory T cells (TRM) as established in mouse models of infection and translated to humans by novel tissue sampling approaches. Integration of findings from mouse and human studies may hold the key to unlocking the potential of TRM for promoting tissue immunity and preventing infection.
Collapse
Affiliation(s)
- Peter A Szabo
- Columbia Center for Translational Immunology, Columbia University, New York, NY, USA
| | - Michelle Miron
- Columbia Center for Translational Immunology, Columbia University, New York, NY, USA.,Department of Microbiology and Immunology, Columbia University, New York, NY, USA
| | - Donna L Farber
- Columbia Center for Translational Immunology, Columbia University, New York, NY, USA. .,Department of Microbiology and Immunology, Columbia University, New York, NY, USA.,Department of Surgery, Columbia University, New York, NY, USA
| |
Collapse
|
139
|
Shacklett BL, Ferre AL, Kiniry BE. Defining T Cell Tissue Residency in Humans: Implications for HIV Pathogenesis and Vaccine Design. Curr HIV/AIDS Rep 2020; 17:109-117. [PMID: 32052270 PMCID: PMC7072053 DOI: 10.1007/s11904-020-00481-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
PURPOSE OF REVIEW This review summarizes recent literature defining tissue-resident memory T cells (TRM) and discusses implications for HIV pathogenesis, vaccines, and eradication efforts. RECENT FINDINGS Investigations using animal models and human tissues have identified a TRM transcriptional profile and elucidated signals within the tissue microenvironment leading to TRM development and maintenance. TRM are major contributors to host response in infectious diseases and cancer; in addition, TRM contribute to pathogenic inflammation in a variety of settings. Although TRM are daunting to study in HIV infection, recent work has helped define their molecular signatures and effector functions and tested strategies for their mobilization. Exclusive reliance on blood sampling to gain an understanding of host immunity overlooks the contribution of TRM, which differ in significant ways from their counterparts in circulation. It is hoped that greater understanding of these cells will lead to novel approaches to prevent and/or eradicate HIV infection.
Collapse
Affiliation(s)
- Barbara L Shacklett
- Department of Medical Microbiology and Immunology, School of Medicine, University of California, Davis, CA, 95616, USA.
- Division of Infectious Disease, Department of Medicine, School of Medicine, University of California, Davis, CA, 95616, USA.
| | - April L Ferre
- Department of Medical Microbiology and Immunology, School of Medicine, University of California, Davis, CA, 95616, USA
| | - Brenna E Kiniry
- Department of Medical Microbiology and Immunology, School of Medicine, University of California, Davis, CA, 95616, USA
| |
Collapse
|
140
|
Booth JS, Goldberg E, Patil SA, Barnes RS, Greenwald BD, Sztein MB. Effect of the live oral attenuated typhoid vaccine, Ty21a, on systemic and terminal ileum mucosal CD4+ T memory responses in humans. Int Immunol 2020; 31:101-116. [PMID: 30346608 PMCID: PMC6376105 DOI: 10.1093/intimm/dxy070] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Accepted: 10/07/2018] [Indexed: 01/01/2023] Open
Abstract
Our current understanding of CD4+ T-cell-mediated immunity (CMI) elicited by the oral live attenuated typhoid vaccine Ty21a is primarily derived from studies using peripheral blood. Very limited data are available in humans regarding mucosal immunity (especially CD4+ T) at the site of infection (e.g. terminal ileum; TI). Here using multiparametric flow cytometry, we examined the effect of Ty21a immunization on TI-lamina propria mononuclear cells (LPMC) and peripheral blood CD4+ T memory (TM) subsets in volunteers undergoing routine colonoscopy. Interestingly, we observed significant increases in the frequencies of LPMC CD4+ T cells following Ty21a immunization, restricted to the T effector/memory (TEM)-CD45RA+ (TEMRA) subset. Importantly, Ty21a immunization elicited Salmonella Typhi-responsive LPMC CD4+ T cells in all major TM subsets [interferon (IFN)γ and interleukin (IL)-17A in TEM; IFNγ and macrophage inflammatory protein (MIP)1β in T central/memory (TCM); and IL-2 in TEMRA]. Subsequently, we analyzed LPMC S. Typhi-responsive CD4+ T cells in depth for multifunctional (MF) effectors. We found that LPMC CD4+ TEM responses were mostly MF, except for those cells exhibiting the characteristics associated with IL-17A responses. Finally, we compared mucosal to systemic responses and observed that LPMC CD4+S. Typhi-specific responses were unique and distinct from their systemic counterparts. This study provides the first demonstration of S. Typhi-specific CD4+ TM responses in the human TI mucosa and provides valuable information about the generation of mucosal immune responses following oral Ty21a immunization.
Collapse
Affiliation(s)
- Jayaum S Booth
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA.,Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Eric Goldberg
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA.,Division of Gastroenterology and Hepatology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Seema A Patil
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA.,Division of Gastroenterology and Hepatology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Robin S Barnes
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Bruce D Greenwald
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA.,Division of Gastroenterology and Hepatology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Marcelo B Sztein
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA.,Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, USA.,Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
141
|
Siefker DT, Vu L, You D, McBride A, Taylor R, Jones TL, DeVincenzo J, Cormier SA. Respiratory Syncytial Virus Disease Severity Is Associated with Distinct CD8 + T-Cell Profiles. Am J Respir Crit Care Med 2020; 201:325-334. [PMID: 31644878 PMCID: PMC6999109 DOI: 10.1164/rccm.201903-0588oc] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 10/21/2019] [Indexed: 01/03/2023] Open
Abstract
Rationale: Respiratory syncytial virus (RSV) causes significant morbidity and mortality in infants worldwide. Although T-helper type 2 (Th2) cell pathology is implicated in severe disease, the mechanisms underlying the development of immunopathology are incompletely understood.Objectives: We aimed to identify local immune responses associated with severe RSV in infants. Our hypothesis was that disease severity would correlate with enhanced Th2 cellular responses.Methods: Nasal aspirates were collected from infants hospitalized with severe (admitted to the pediatric ICU) or moderate (maintained in the general ward) RSV disease at 5 to 9 days after enrollment. The immune response was investigated by evaluating T-lymphocyte cellularity, cytokine concentration, and viral load.Measurements and Main Results: Patients with severe disease had increased proportions of CD8 (cluster of differentiation 8)-positive T cells expressing IL-4 (Tc2) and reduced proportions of CD8+ T cells expressing IFNγ (Tc1). Nasal aspirates from patients with severe disease had reduced concentrations of IL-17. Patients with greater frequencies of Tc1, CD8+ T cells expressing IL-17 (Tc17), and CD4+ T cells expressing IL-17 (Th17) had shorter durations of hospitalization.Conclusions: Severe RSV disease was associated with distinct T-cell profiles. Tc1, Tc17, and Th17 were associated with shorter hospital stay and may play a protective role, whereas Tc2 cells may play a previously underappreciated role in pathology.
Collapse
Affiliation(s)
- David T. Siefker
- Department of Biological Sciences, College of Science, and Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana
| | - Luan Vu
- Department of Biological Sciences, College of Science, and Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana
| | - Dahui You
- Department of Pediatrics, and
- Children’s Foundation Research Institute, Le Bonheur Children’s Hospital, Memphis, Tennessee
| | | | | | - Tamekia L. Jones
- Department of Pediatrics, and
- Department of Preventive Medicine, University of Tennessee Health Science Center, Memphis, Tennessee; and
- Children’s Foundation Research Institute, Le Bonheur Children’s Hospital, Memphis, Tennessee
| | - John DeVincenzo
- Department of Pediatrics, and
- Children’s Foundation Research Institute, Le Bonheur Children’s Hospital, Memphis, Tennessee
| | - Stephania A. Cormier
- Department of Biological Sciences, College of Science, and Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana
| |
Collapse
|
142
|
Kirsebom F, Michalaki C, Agueda-Oyarzabal M, Johansson C. Neutrophils do not impact viral load or the peak of disease severity during RSV infection. Sci Rep 2020; 10:1110. [PMID: 31980667 PMCID: PMC6981203 DOI: 10.1038/s41598-020-57969-w] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 01/07/2020] [Indexed: 11/30/2022] Open
Abstract
Lung and airway neutrophils are a hallmark of severe disease in infants with respiratory syncytial virus (RSV)-induced lower respiratory tract infections. Despite their abundance in the lungs during RSV infection of both mice and man, the role of neutrophils in viral control and in immune pathology is not clear. Here, antibody mediated neutrophil depletion was used to investigate the degree to which neutrophils impact the lung immune environment, the control of viral replication and the peak severity of disease after RSV infection of mice. Neutrophil depletion did not substantially affect the levels of inflammatory mediators such as type I interferons, IL-6, TNF-α or IL-1β in response to RSV. In addition, the lack of neutrophils did not change the viral load during RSV infection. Neither neutrophil depletion nor the enhancement of lung neutrophils by administration of the chemoattractant CXCL1 during RSV infection affected disease severity as measured by weight loss. Therefore, in this model of RSV infection, lung neutrophils do not offer obvious benefits to the host in terms of increasing anti-viral inflammatory responses or restricting viral replication and neutrophils do not contribute to disease severity.
Collapse
Affiliation(s)
- Freja Kirsebom
- National Heart and Lung Institute, Imperial College London, London, UK
| | | | | | - Cecilia Johansson
- National Heart and Lung Institute, Imperial College London, London, UK.
| |
Collapse
|
143
|
Local heroes or villains: tissue-resident memory T cells in human health and disease. Cell Mol Immunol 2020; 17:113-122. [PMID: 31969685 DOI: 10.1038/s41423-019-0359-1] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 12/17/2019] [Indexed: 12/14/2022] Open
Abstract
Tissue-resident memory T (TRM) cells are increasingly associated with the outcomes of health and disease. TRM cells can mediate local immune protection against infections and cancer, which has led to interest in TRM cells as targets for vaccination and immunotherapies. However, these cells have also been implicated in mediating detrimental pro-inflammatory responses in autoimmune skin diseases such as psoriasis, alopecia areata, and vitiligo. Here, we summarize the biology of TRM cells established in animal models and in translational human studies. We review the beneficial effects of TRM cells in mediating protective responses against infection and cancer and the adverse role of TRM cells in driving pathology in autoimmunity. A further understanding of the breadth and mechanisms of TRM cell activity is essential for the safe design of strategies that manipulate TRM cells, such that protective responses can be enhanced without unwanted tissue damage, and pathogenic TRM cells can be eliminated without losing local immunity.
Collapse
|
144
|
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
|
145
|
Orihuela CJ, Maus UA, Brown JS. Can animal models really teach us anything about pneumonia? Pro. Eur Respir J 2020; 55:55/1/1901539. [DOI: 10.1183/13993003.01539-2019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 10/03/2019] [Indexed: 01/03/2023]
|
146
|
Adenovectors encoding RSV-F protein induce durable and mucosal immunity in macaques after two intramuscular administrations. NPJ Vaccines 2019; 4:54. [PMID: 31885877 PMCID: PMC6925274 DOI: 10.1038/s41541-019-0150-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 11/20/2019] [Indexed: 12/20/2022] Open
Abstract
Respiratory Syncytial Virus (RSV) can cause severe respiratory disease, yet a licensed vaccine is not available. We determined the immunogenicity of two homologous and one heterologous intramuscular prime-boost vaccination regimens using replication-incompetent adenoviral vectors of human serotype 26 and 35 (Ad26 and Ad35), expressing a prototype antigen based on the wild-type fusion (F) protein of RSV strain A2 in adult, RSV-naive cynomolgus macaques. All regimens induced substantial, boostable antibody responses that recognized the F protein in pre- and postfusion conformation, neutralized multiple strains of RSV, and persisted for at least 80 weeks. Vaccination induced durable systemic RSV-F-specific T-cell responses characterized mainly by CD4+ T cells expressing Th1-type cytokines, as well as RSV-F-specific CD4+ and CD8+ T cells, IgG, and IgA in the respiratory tract. Intramuscular immunization with Ad26 and 35 vectors thus is a promising approach for the development of an optimized RSV vaccine expected to induce long-lasting humoral and cellular immune responses that distribute systemically and to mucosal sites.
Collapse
|
147
|
Thwaites RS, Openshaw PJ. The Respiratory Mucosa: Front and Center in Respiratory Syncytial Virus Disease. Am J Respir Crit Care Med 2019; 200:1340-1342. [PMID: 31412707 PMCID: PMC6884050 DOI: 10.1164/rccm.201907-1306ed] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Affiliation(s)
- Ryan S Thwaites
- National Heart and Lung InstituteImperial College LondonLondon, United Kingdom
| | - Peter J Openshaw
- National Heart and Lung InstituteImperial College LondonLondon, United Kingdom
| |
Collapse
|
148
|
Antunes KH, Becker A, Franceschina C, de Freitas DDN, Lape I, da Cunha MD, Leitão L, Rigo MM, Pinto LA, Stein RT, de Souza APD. Respiratory syncytial virus reduces STAT3 phosphorylation in human memory CD8 T cells stimulated with IL-21. Sci Rep 2019; 9:17766. [PMID: 31780735 PMCID: PMC6882881 DOI: 10.1038/s41598-019-54240-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 11/07/2019] [Indexed: 12/21/2022] Open
Abstract
Respiratory syncytial virus (RSV) is a common cause of childhood lower respiratory tract infections. The recent failure of a vaccine candidate based on recombinant F protein underlines the urgent need to better understand the protective human memory immune response against RSV. Signal transducer and activator of transcription 3 (STAT3) protein is a transcription factor that promotes the maturation of the memory CD8 T cell response in cooperation with IL-10 and IL-21. However, the role of STAT3 in the memory CD8 T cell response during RSV infection remains to be elucidated. We found that in infants with bronchiolitis infected with RSV, the expression of STAT3 detected in nasal washes is reduced when compared to that in infants infected by other viruses. In vitro, RSV impairs STAT3 phosphorylation induced by IL-21 in purified human memory CD8 T cells. In addition, RSV decreases granzyme B production by memory CD8 T cells, reducing its cytotoxic activity against RSV-infected epithelial pulmonary cell lines. Together, these data indicate that RSV modulates the IL-21/STAT3 pathway in human memory CD8 T cells, and this could be a mechanism to be further explored to improve the memory response against the infection.
Collapse
Affiliation(s)
- Krist Helen Antunes
- Laboratory of Clinical and experimental Immunology, Infant Center, School of Medicine, Pontificia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - André Becker
- Laboratory of Clinical and experimental Immunology, Infant Center, School of Medicine, Pontificia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Caroline Franceschina
- Laboratory of Clinical and experimental Immunology, Infant Center, School of Medicine, Pontificia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Deise do Nascimento de Freitas
- Laboratory of Clinical and experimental Immunology, Infant Center, School of Medicine, Pontificia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Isadora Lape
- Laboratory of Clinical and experimental Immunology, Infant Center, School of Medicine, Pontificia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Mariana D'Ávila da Cunha
- Laboratory of Clinical and experimental Immunology, Infant Center, School of Medicine, Pontificia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Lidiane Leitão
- Laboratory of Respiratory Physiology, Infant Center, School of Medicine, PUCRS, Porto Alegre, Brazil
| | - Mauricio M Rigo
- Laboratory of Clinical and experimental Immunology, Infant Center, School of Medicine, Pontificia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Leonardo Araújo Pinto
- Laboratory of Respiratory Physiology, Infant Center, School of Medicine, PUCRS, Porto Alegre, Brazil
| | - Renato T Stein
- Laboratory of Respiratory Physiology, Infant Center, School of Medicine, PUCRS, Porto Alegre, Brazil
| | - Ana Paula Duarte de Souza
- Laboratory of Clinical and experimental Immunology, Infant Center, School of Medicine, Pontificia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brazil. .,School of Health and Life Sciences, PUCRS, Porto Alegre, Brazil.
| |
Collapse
|
149
|
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
|
150
|
Knight FC, Gilchuk P, Kumar A, Becker KW, Sevimli S, Jacobson ME, Suryadevara N, Wang-Bishop L, Boyd KL, Crowe JE, Joyce S, Wilson JT. Mucosal Immunization with a pH-Responsive Nanoparticle Vaccine Induces Protective CD8 + Lung-Resident Memory T Cells. ACS NANO 2019; 13:10939-10960. [PMID: 31553872 PMCID: PMC6832804 DOI: 10.1021/acsnano.9b00326] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Tissue-resident memory T cells (TRM) patrol nonlymphoid organs and provide superior protection against pathogens that commonly infect mucosal and barrier tissues, such as the lungs, intestine, liver, and skin. Thus, there is a need for vaccine technologies that can induce a robust, protective TRM response in these tissues. Nanoparticle (NP) vaccines offer important advantages over conventional vaccines; however, there has been minimal investigation into the design of NP-based vaccines for eliciting TRM responses. Here, we describe a pH-responsive polymeric nanoparticle vaccine for generating antigen-specific CD8+ TRM cells in the lungs. With a single intranasal dose, the NP vaccine elicited airway- and lung-resident CD8+ TRM cells and protected against respiratory virus challenge in both sublethal (vaccinia) and lethal (influenza) infection models for up to 9 weeks after immunization. In elucidating the contribution of material properties to the resulting TRM response, we found that the pH-responsive activity of the carrier was important, as a structurally analogous non-pH-responsive control carrier elicited significantly fewer lung-resident CD8+ T cells. We also demonstrated that dual-delivery of protein antigen and nucleic acid adjuvant on the same NP substantially enhanced the magnitude, functionality, and longevity of the antigen-specific CD8+ TRM response in the lungs. Compared to administration of soluble antigen and adjuvant, the NP also mediated retention of vaccine cargo in pulmonary antigen-presenting cells (APCs), enhanced APC activation, and increased production of TRM-related cytokines. Overall, these data suggest a promising vaccine platform technology for rapid generation of protective CD8+ TRM cells in the lungs.
Collapse
Affiliation(s)
- Frances C. Knight
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Pavlo Gilchuk
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN 37212, USA
| | - Amrendra Kumar
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN 37212, USA
| | - Kyle W. Becker
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Sema Sevimli
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Max E. Jacobson
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Naveenchandra Suryadevara
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN 37212, USA
| | - Lihong Wang-Bishop
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Kelli L. Boyd
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - James E. Crowe
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Chemical and Physical Biology Program, Vanderbilt University, Nashville, TN 37235, USA
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37235, USA
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Sebastian Joyce
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN 37212, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - John T. Wilson
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN 37235, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Corresponding Author:
| |
Collapse
|