1
|
Yunis J, Short KR, Yu D. Severe respiratory viral infections: T-cell functions diverging from immunity to inflammation. Trends Microbiol 2023; 31:644-656. [PMID: 36635162 PMCID: PMC9829516 DOI: 10.1016/j.tim.2022.12.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 12/20/2022] [Accepted: 12/21/2022] [Indexed: 01/11/2023]
Abstract
Respiratory viral infections such as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and influenza A virus (IAV) trigger distinct clinical outcomes defined by immunity-based viral clearance or disease associated with exaggerated and prolonged inflammation. The important role of T cells in shaping both antiviral immunity and inflammation has revived interest in understanding the host-pathogen interactions that lead to the diverse functions of T cells in respiratory viral infections. Inborn deficiencies and acquired insufficiency in immunity can prolong infection and shift the immune response towards exacerbated inflammation, which results from persistent innate immune activation and bystander T-cell activation that is nonspecific to the pathogen but is often driven by cytokines. This review discusses how virus variants, exposure doses, routes of infection, host genetics, and immune history can modulate the activation and function of T cells, thus influencing clinical outcomes. Knowledge of virus-host interaction can inform strategies to prevent immune dysfunction in respiratory viral infection and help in the treatment of associated diseases.
Collapse
Affiliation(s)
- Joseph Yunis
- Frazer Institute, Faculty of Medicine, The University of Queensland, Woolloongabba, QLD 4102, Australia.
| | - Kirsty R Short
- School of Chemistry and Molecular Biosciences, Faculty of Science, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Di Yu
- Frazer Institute, Faculty of Medicine, The University of Queensland, Woolloongabba, QLD 4102, Australia; Ian Frazer Centre for Children's Immunotherapy Research, Child Health Research Centre, Faculty of Medicine, The University of Queensland, Brisbane, Australia.
| |
Collapse
|
2
|
Inhaled particulate accumulation with age impairs immune function and architecture in human lung lymph nodes. Nat Med 2022; 28:2622-2632. [PMID: 36411343 PMCID: PMC9835154 DOI: 10.1038/s41591-022-02073-x] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Accepted: 10/03/2022] [Indexed: 11/22/2022]
Abstract
Older people are particularly susceptible to infectious and neoplastic diseases of the lung and it is unclear how lifelong exposure to environmental pollutants affects respiratory immune function. In an analysis of human lymph nodes (LNs) from 84 organ donors aged 11-93 years, we found a specific age-related decline in lung-associated, but not gut-associated, LN immune function linked to the accumulation of inhaled atmospheric particulate matter. Increasing densities of particulates were found in lung-associated LNs with age, but not in the corresponding gut-associated LNs. Particulates were specifically contained within CD68+CD169- macrophages, which exhibited decreased activation, phagocytic capacity, and altered cytokine production compared with non-particulate-containing macrophages. The structures of B cell follicles and lymphatic drainage were also disrupted in lung-associated LNs with particulates. Our results reveal that the cumulative effects of environmental exposure and age may compromise immune surveillance of the lung via direct effects on immune cell function and lymphoid architecture.
Collapse
|
3
|
Hartwig SM, Miller AM, Varga SM. Respiratory Syncytial Virus Provides Protection against a Subsequent Influenza A Virus Infection. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:720-731. [PMID: 35022269 PMCID: PMC8851717 DOI: 10.4049/jimmunol.2000751] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 11/23/2021] [Indexed: 02/03/2023]
Abstract
Respiratory infections are a leading cause of morbidity and mortality. The presence of multiple heterologous virus infections is routinely observed in a subset of individuals screened for the presence of respiratory viruses. However, the impact overlapping infections has on disease severity and the host immune response is not well understood. Respiratory syncytial virus (RSV) and influenza A virus (IAV) are two of the most common respiratory infections observed in hospitalized patients, particularly in the very young and aged populations. In this study, we examined how the order in which BALB/c mice were infected with both RSV and IAV impacts disease severity. RSV infection prior to an IAV infection was associated with decreased weight loss and increased survival as compared with IAV infection alone. In contrast, IAV infection prior to an RSV infection was associated with similar morbidity and mortality as compared with an IAV infection alone. Our results suggest that the order in which viral infections are acquired plays a critical role in the outcome of disease severity and the host immune response.
Collapse
Affiliation(s)
- Stacey M. Hartwig
- Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa, 52242
| | - Ann M. Miller
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, Iowa, 52242
| | - Steven M. Varga
- Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa, 52242,Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, Iowa, 52242,Department of Pathology, University of Iowa, Iowa City, Iowa, 52242
| |
Collapse
|
4
|
Guo Z, Zhang Z, Prajapati M, Li Y. Lymphopenia Caused by Virus Infections and the Mechanisms Beyond. Viruses 2021; 13:v13091876. [PMID: 34578457 PMCID: PMC8473169 DOI: 10.3390/v13091876] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 09/14/2021] [Accepted: 09/18/2021] [Indexed: 02/07/2023] Open
Abstract
Viral infections can give rise to a systemic decrease in the total number of lymphocytes in the blood, referred to as lymphopenia. Lymphopenia may affect the host adaptive immune responses and impact the clinical course of acute viral infections. Detailed knowledge on how viruses induce lymphopenia would provide valuable information into the pathogenesis of viral infections and potential therapeutic targeting. In this review, the current progress of viruses-induced lymphopenia is summarized and the potential mechanisms and factors involved are discussed.
Collapse
Affiliation(s)
- Zijing Guo
- State Key Laboratory on Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730030, China;
- College of Animal Husbandry and Veterinary Medicine, Southwest Minzu University, Chengdu 610041, China; (Z.Z.); (M.P.)
| | - Zhidong Zhang
- College of Animal Husbandry and Veterinary Medicine, Southwest Minzu University, Chengdu 610041, China; (Z.Z.); (M.P.)
| | - Meera Prajapati
- College of Animal Husbandry and Veterinary Medicine, Southwest Minzu University, Chengdu 610041, China; (Z.Z.); (M.P.)
- National Animal Health Research Centre, Nepal Agricultural Research Council, Lalitpur 44700, Nepal
| | - Yanmin Li
- College of Animal Husbandry and Veterinary Medicine, Southwest Minzu University, Chengdu 610041, China; (Z.Z.); (M.P.)
- Correspondence: ; Tel.: +28-85528276
| |
Collapse
|
5
|
Lega S, Naviglio S, Volpi S, Tommasini A. Recent Insight into SARS-CoV2 Immunopathology and Rationale for Potential Treatment and Preventive Strategies in COVID-19. Vaccines (Basel) 2020; 8:224. [PMID: 32423059 PMCID: PMC7349555 DOI: 10.3390/vaccines8020224] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 05/10/2020] [Accepted: 05/11/2020] [Indexed: 01/08/2023] Open
Abstract
As the outbreak of the new coronavirus (SARS-CoV-2) infection is spreading globally, great effort is being made to understand the disease pathogenesis and host factors that predispose to disease progression in an attempt to find a window of opportunity for intervention. In addition to the direct cytopathic effect of the virus, the host hyper-inflammatory response has emerged as a key factor in determining disease severity and mortality. Accumulating clinical observations raised hypotheses to explain why some patients develop more severe disease while others only manifest mild or no symptoms. So far, Covid-19 management remains mainly supportive. However, many researches are underway to clarify the role of antiviral and immunomodulating drugs in changing morbidity and mortality in patients who become severely ill. This review summarizes the current state of knowledge on the interaction between SARS-CoV-2 and the host immune system and discusses recent findings on proposed pharmacologic treatments.
Collapse
Affiliation(s)
- Sara Lega
- Institute for Maternal and Child Health IRCCS Burlo Garofolo, 34137 Trieste, Italy; (S.L.); (A.T.)
| | - Samuele Naviglio
- Institute for Maternal and Child Health IRCCS Burlo Garofolo, 34137 Trieste, Italy; (S.L.); (A.T.)
| | - Stefano Volpi
- Center for Autoinflammatory Diseases and Immunodeficiency, IRCCS Istituto Giannina Gaslini and Università degli Studi di Genova, 16147 Genova, Italy;
| | - Alberto Tommasini
- Institute for Maternal and Child Health IRCCS Burlo Garofolo, 34137 Trieste, Italy; (S.L.); (A.T.)
- Department of Medicine, Surgery and Health Sciences, University of Trieste, 34137 Trieste, Italy
| |
Collapse
|
6
|
Sumbria D, Berber E, Rouse BT. Factors Affecting the Tissue Damaging Consequences of Viral Infections. Front Microbiol 2019; 10:2314. [PMID: 31636623 PMCID: PMC6787772 DOI: 10.3389/fmicb.2019.02314] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 09/23/2019] [Indexed: 12/15/2022] Open
Abstract
Humans and animals are infected by multiple endogenous and exogenous viruses but few agents cause overt tissue damage. We review the circumstances which favor overt disease expression. These can include intrinsic virulence of the agent, new agents acquired from heterologous species, the circumstances of infection such as dose and route, current infection with other agents which includes the composition of the microbiome at mucosal and other sites, past history of exposure to other infections as well as the immune status of the host. We also briefly discuss promising therapeutic strategies that can expand immune response patterns that minimize tissue damaging responses to viral infections.
Collapse
Affiliation(s)
| | | | - Barry T. Rouse
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, The University of Tennessee, Knoxville, Knoxville, TN, United States
| |
Collapse
|
7
|
Host Single Nucleotide Polymorphisms Modulating Influenza A Virus Disease in Humans. Pathogens 2019; 8:pathogens8040168. [PMID: 31574965 PMCID: PMC6963926 DOI: 10.3390/pathogens8040168] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 09/27/2019] [Accepted: 09/28/2019] [Indexed: 12/14/2022] Open
Abstract
A large number of human genes associated with viral infections contain single nucleotide polymorphisms (SNPs), which represent a genetic variation caused by the change of a single nucleotide in the DNA sequence. SNPs are located in coding or non-coding genomic regions and can affect gene expression or protein function by different mechanisms. Furthermore, they have been linked to multiple human diseases, highlighting their medical relevance. Therefore, the identification and analysis of this kind of polymorphisms in the human genome has gained high importance in the research community, and an increasing number of studies have been published during the last years. As a consequence of this exhaustive exploration, an association between the presence of some specific SNPs and the susceptibility or severity of many infectious diseases in some risk population groups has been found. In this review, we discuss the relevance of SNPs that are important to understand the pathology derived from influenza A virus (IAV) infections in humans and the susceptibility of some individuals to suffer more severe symptoms. We also discuss the importance of SNPs for IAV vaccine effectiveness.
Collapse
|
8
|
Franchini AM, Myers JR, Jin GB, Shepherd DM, Lawrence BP. Genome-Wide Transcriptional Analysis Reveals Novel AhR Targets That Regulate Dendritic Cell Function during Influenza A Virus Infection. Immunohorizons 2019; 3:219-235. [PMID: 31356168 DOI: 10.4049/immunohorizons.1900004] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 05/25/2019] [Indexed: 12/16/2022] Open
Abstract
Activation of the ligand inducible aryl hydrocarbon receptor (AhR) during primary influenza A virus infection diminishes host responses by negatively regulating the ability of dendritic cells (DC) to prime naive CD8+ T cells, which reduces the generation of CTL. However, AhR-regulated genes and signaling pathways in DCs are not fully known. In this study, we used unbiased gene expression profiling to identify differentially expressed genes and signaling pathways in DCs that are modulated by AhR activation in vivo. Using the prototype AhR agonist TCDD, we identified the lectin receptor Cd209a (DC-SIGN) and chemokine Ccl17 as novel AhR target genes. We further show the percentage of DCs expressing CD209a on their surface was significantly decreased by AhR activation during infection. Whereas influenza A virus infection increased CCL17 protein levels in the lung and lung-draining lymph nodes, this was significantly reduced following AhR activation. Targeted excision of AhR in the hematopoietic compartment confirmed AhR is required for downregulation of CCL17 and CD209a. Loss of AhR's functional DNA-binding domain demonstrates that AhR activation alone is necessary but not sufficient to drive downregulation. AhR activation induced similar changes in gene expression in human monocyte-derived DCs. Analysis of the murine and human upstream regulatory regions of Cd209a and Ccl17 revealed a suite of potential transcription factor partners for AhR, which may coregulate these genes in vivo. This study highlights the breadth of AhR-regulated pathways within DCs, and that AhR likely interacts with other transcription factors to modulate DC functions during infection.
Collapse
Affiliation(s)
- Anthony M Franchini
- Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642
| | - Jason R Myers
- Genomics Research Center, James P. Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY 14642
| | - Guang-Bi Jin
- Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642
| | - David M Shepherd
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, MT 59812; and.,Center for Translational Medicine, University of Montana, Missoula, MT 59812
| | - B Paige Lawrence
- Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642;
| |
Collapse
|
9
|
Hornick EE, Dagvadorj J, Zacharias ZR, Miller AM, Langlois RA, Chen P, Legge KL, Bishop GA, Sutterwala FS, Cassel SL. Dendritic cell NLRC4 regulates influenza A virus-specific CD4 T cell responses through FasL expression. J Clin Invest 2019; 129:2888-2897. [PMID: 31038471 DOI: 10.1172/jci124937] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Influenza A virus (IAV)-specific T cell responses are important correlates of protection during primary and subsequent infections. Generation and maintenance of robust IAV-specific T cell responses relies on T cell interactions with dendritic cells (DCs). In this study, we explore the role of nucleotide-binding domain leucine-rich repeat containing receptor family member NLRC4 in modulating the DC phenotype during IAV infection. Nlrc4-/- mice had worsened survival and increased viral titers during infection, normal innate immune cell recruitment and IAV-specific CD8 T cell responses, but severely blunted IAV-specific CD4 T cell responses compared to wild-type mice. The defect in the pulmonary IAV-specific CD4 T cell response was not a result of defective priming or migration of these cells in Nlrc4-/- mice but was instead due to an increase in FasL+ DCs, resulting in IAV-specific CD4 T cell death. Together, our data support a novel role for NLRC4 in regulating the phenotype of lung DCs during a respiratory viral infection, and thereby influencing the magnitude of protective T cell responses.
Collapse
Affiliation(s)
- Emma E Hornick
- Interdisciplinary Program in Immunology, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Jargalsaikhan Dagvadorj
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Women's Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Zeb R Zacharias
- Interdisciplinary Program in Immunology, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA.,Department of Pathology, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Ann M Miller
- Department of Surgery, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Ryan A Langlois
- Center for Immunology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Peter Chen
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Women's Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Kevin L Legge
- Interdisciplinary Program in Immunology, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA.,Department of Pathology, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA.,Department of Microbiology and Immunology, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Gail A Bishop
- Interdisciplinary Program in Immunology, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA.,Department of Microbiology and Immunology, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA.,Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA.,Veterans Affairs Medical Center, Iowa City, Iowa, USA
| | - Fayyaz S Sutterwala
- Interdisciplinary Program in Immunology, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA.,Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Women's Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Suzanne L Cassel
- Interdisciplinary Program in Immunology, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA.,Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Women's Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| |
Collapse
|
10
|
Fujikura D, Miyazaki T. Programmed Cell Death in the Pathogenesis of Influenza. Int J Mol Sci 2018; 19:ijms19072065. [PMID: 30012970 PMCID: PMC6073994 DOI: 10.3390/ijms19072065] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 07/04/2018] [Accepted: 07/12/2018] [Indexed: 12/17/2022] Open
Abstract
Influenza is a respiratory disease induced by infection by the influenza virus, which is a member of Orthomyxoviridae family. This infectious disease has serious impacts on public health systems and results in considerable mortality and economic costs throughout the world. Based on several experimental studies, massive host immune reaction is associated with the disease severity of influenza. Programmed cell death is typically induced during virus infection as a consequence of host immune reaction to limit virus spread by eliminating niches for virus propagation without causing inflammation. However, in some viral infectious diseases, such as influenza, in the process of immune reaction, aberrant induction of programmed cell death disturbs the maintenance of organ function. Current reports show that there are different types of programmed cell death that vary in terms of molecular mechanisms and/or associations with inflammation. In addition, these novel types of programmed cell death are associated with pathogenesis rather than suppressing virus propagation in the disease course. Here, we review our current understanding of mechanisms of programmed cell death in the pathogenesis of influenza.
Collapse
Affiliation(s)
- Daisuke Fujikura
- Center for Advanced Research and Education (CARE), Asahikawa Medical University, 2-1-1-1 Midorigaoka-Higashi, Asahikawa 078-8510, Japan.
| | - Tadaaki Miyazaki
- Department of Probiotics Immunology, Institute for Genetic Medicine, Hokkaido University, North-15, West-7, Kita-ku, Sapporo 060-0815, Japan.
| |
Collapse
|
11
|
Hua X, Vijay R, Channappanavar R, Athmer J, Meyerholz DK, Pagedar N, Tilley S, Perlman S. Nasal priming by a murine coronavirus provides protective immunity against lethal heterologous virus pneumonia. JCI Insight 2018; 3:99025. [PMID: 29875310 DOI: 10.1172/jci.insight.99025] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 04/19/2018] [Indexed: 12/28/2022] Open
Abstract
The nasal mucosa is an important component of mucosal immunity. Immunogenic particles in inspired air are known to activate the local nasal mucosal immune system and can lead to sinonasal inflammation; however, little is known about the effect of this activation on the lung immune environment. Here, we showed that nasal inoculation of murine coronavirus (CoV) in the absence of direct lung infection primes the lung immune environment by recruiting activated monocytes (Ly6C+ inflammatory monocytes) and NK cells into the lungs. Unlike infiltration of these cells into directly infected lungs, a process that requires type I IFN signaling, nasally induced infiltration of Ly6C+ inflammatory monocytes into the lungs is IFN-I independent. These activated macrophages ingested antigen and migrated to pulmonary lymph nodes, and enhanced both innate and adaptive immunity after heterologous virus infection. Clinically, such nasal-only inoculation of MHV-1 failed to cause pneumonia but significantly reduced mortality and morbidity of lethal pneumonia caused by severe acute respiratory syndrome CoV (SARS-CoV) or influenza A virus. Together, the data indicate that the nose and upper airway remotely prime the lung immunity to protect the lungs from direct viral infections.
Collapse
Affiliation(s)
- Xiaoyang Hua
- Department of Otolaryngology-Head and Neck Surgery
| | - Rahul Vijay
- Interdisciplinary Program in Immunology.,Department of Microbiology and Immunology, and
| | | | | | | | | | - Stephen Tilley
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Stanley Perlman
- Interdisciplinary Program in Immunology.,Department of Microbiology and Immunology, and
| |
Collapse
|
12
|
Carlin LE, Hemann EA, Zacharias ZR, Heusel JW, Legge KL. Natural Killer Cell Recruitment to the Lung During Influenza A Virus Infection Is Dependent on CXCR3, CCR5, and Virus Exposure Dose. Front Immunol 2018; 9:781. [PMID: 29719539 PMCID: PMC5913326 DOI: 10.3389/fimmu.2018.00781] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 03/28/2018] [Indexed: 12/31/2022] Open
Abstract
Natural killer (NK) cells are vital components of the antiviral immune response, but their contributions in defense against influenza A virus (IAV) are not well understood. To better understand NK cell responses during IAV infections, we examined the magnitude, kinetics, and contribution of NK cells to immunity and protection during high- and low-dose IAV infections. Herein, we demonstrate an increased accumulation of NK cells in the lung in high-dose vs. low-dose infections. In part, this increase is due to the local proliferation of pulmonary NK cells. However, the majority of NK cell accumulation within the lungs and airways during an IAV infection is due to recruitment that is partially dependent upon CXCR3 and CCR5, respectively. Therefore, altogether, our results demonstrate that NK cells are actively recruited to the lungs and airways during IAV infection and that the magnitude of the recruitment may relate to the inflammatory environment found within the tissues during high- and low-dose IAV infections.
Collapse
Affiliation(s)
- Lindsey E Carlin
- Interdisciplinary Graduate Program in Immunology, Department of Pathology, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Emily A Hemann
- Interdisciplinary Graduate Program in Immunology, Department of Pathology, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Zeb R Zacharias
- Interdisciplinary Graduate Program in Immunology, Department of Pathology, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Jonathan W Heusel
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO, United States
| | - Kevin L Legge
- Interdisciplinary Graduate Program in Immunology, Department of Pathology, University of Iowa Carver College of Medicine, Iowa City, IA, United States.,Department of Microbiology and Immunology, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| |
Collapse
|
13
|
Richards KA, DiPiazza AT, Rattan A, Knowlden ZAG, Yang H, Sant AJ. Diverse Epitope Specificity, Immunodominance Hierarchy, and Functional Avidity of Effector CD4 T Cells Established During Priming Is Maintained in Lung After Influenza A Virus Infection. Front Immunol 2018; 9:655. [PMID: 29681900 PMCID: PMC5897437 DOI: 10.3389/fimmu.2018.00655] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 03/16/2018] [Indexed: 11/13/2022] Open
Abstract
One of the major contributions to protective immunity to influenza viruses that is provided by virus-specific CD4 T cells is delivery of effector function to the infected lung. However, there is little known about the selection and breadth of viral epitope-specific CD4 T cells that home to the lung after their initial priming. In this study, using a mouse model of influenza A infection and an unbiased method of epitope identification, the viral epitope-specific CD4 T cells elicited after infection were identified and quantified. We found that a very diverse specificity of CD4 T cells is primed by infection, including epitopes from hemagglutinin, neuraminidase, matrix protein, nucleoprotein, and non-structural protein-1. Using peptide-specific cytokine EliSpots, the diversity and immunodominance hierarchies established in the lung-draining lymph node were compared with specificities of CD4 T cells that home to the lung. Our studies revealed that CD4 T cells of all epitope specificities identified in peripheral lymphoid tissue home back to the lung and that most of these lung-homing cells are localized within the tissue rather than the pulmonary vasculature. There is a striking shift of CD4 T cell functionality that enriches for IFN-γ production as cells are primed in the lymph node, enter the lung vasculature, and finally establish residency in the tissue, but with no apparent shifts in their functional avidity. We conclude that CD4 T cells of broad viral epitope specificity are recruited into the lung after influenza infection, where they then have the opportunity to encounter infected or antigen-bearing antigen-presenting cells.
Collapse
Affiliation(s)
- Katherine A. Richards
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, United States
| | - Anthony T. DiPiazza
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, United States
- Viral Pathogenesis Laboratory, Vaccine Research Center NIAID, Bethesda, MD, United States
| | - Ajitanuj Rattan
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, United States
| | - Zackery A. G. Knowlden
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, United States
| | - Hongmei Yang
- Department of Biostatistics and Computational Biology, University of Rochester, Rochester, NY, United States
| | - Andrea J. Sant
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, United States
| |
Collapse
|
14
|
Cheng MA, Farmer E, Huang C, Lin J, Hung CF, Wu TC. Therapeutic DNA Vaccines for Human Papillomavirus and Associated Diseases. Hum Gene Ther 2018; 29:971-996. [PMID: 29316817 DOI: 10.1089/hum.2017.197] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Human papillomavirus (HPV) has long been recognized as the causative agent of cervical cancer. High-risk HPV types 16 and 18 alone are responsible for over 70% of all cases of cervical cancers. More recently, HPV has been identified as an etiological factor for several other forms of cancers, including oropharyngeal, anogenital, and skin. Thus, the association of HPV with these malignancies creates an opportunity to control these HPV lesions and HPV-associated malignancies through immunization. Strategies to prevent or to therapeutically treat HPV infections have been developed and are still pushing innovative boundaries. Currently, commercial prophylactic HPV vaccines are widely available, but they are not able to control established infections or lesions. As a result, there is an urgent need for the development of therapeutic HPV vaccines, to treat existing infections, and to prevent the development of HPV-associated cancers. In particular, DNA vaccination has emerged as a promising form of therapeutic HPV vaccine. DNA vaccines have great potential for the treatment of HPV infections and HPV-associated cancers due to their safety, stability, simplicity of manufacturability, and ability to induce antigen-specific immunity. This review focuses on the current state of therapeutic HPV DNA vaccines, including results from recent and ongoing clinical trials, and outlines different strategies that have been employed to improve their potencies. The continued progress and improvements made in therapeutic HPV DNA vaccine development holds great potential for innovative ways to effectively treat HPV infections and HPV-associated diseases.
Collapse
Affiliation(s)
- Max A Cheng
- 1 Department of Pathology, Johns Hopkins Medical Institutions , Baltimore, Maryland
| | - Emily Farmer
- 1 Department of Pathology, Johns Hopkins Medical Institutions , Baltimore, Maryland
| | - Claire Huang
- 1 Department of Pathology, Johns Hopkins Medical Institutions , Baltimore, Maryland
| | - John Lin
- 1 Department of Pathology, Johns Hopkins Medical Institutions , Baltimore, Maryland
| | - Chien-Fu Hung
- 1 Department of Pathology, Johns Hopkins Medical Institutions , Baltimore, Maryland.,2 Department of Oncology, Johns Hopkins Medical Institutions , Baltimore, Maryland
| | - T-C Wu
- 1 Department of Pathology, Johns Hopkins Medical Institutions , Baltimore, Maryland.,2 Department of Oncology, Johns Hopkins Medical Institutions , Baltimore, Maryland.,3 Department of Obstetrics and Gynecology, Johns Hopkins Medical Institutions , Baltimore, Maryland.,4 Department of Molecular Microbiology and Immunology, Johns Hopkins Medical Institutions , Baltimore, Maryland
| |
Collapse
|
15
|
Hornick EE, Banoth B, Miller AM, Zacharias ZR, Jain N, Wilson ME, Gibson-Corley KN, Legge KL, Bishop GA, Sutterwala FS, Cassel SL. Nlrp12 Mediates Adverse Neutrophil Recruitment during Influenza Virus Infection. THE JOURNAL OF IMMUNOLOGY 2017; 200:1188-1197. [PMID: 29282312 DOI: 10.4049/jimmunol.1700999] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 11/28/2017] [Indexed: 12/23/2022]
Abstract
Exaggerated inflammatory responses during influenza A virus (IAV) infection are typically associated with severe disease. Neutrophils are among the immune cells that can drive this excessive and detrimental inflammation. In moderation, however, neutrophils are necessary for optimal viral control. In this study, we explore the role of the nucleotide-binding domain leucine-rich repeat containing receptor family member Nlrp12 in modulating neutrophilic responses during lethal IAV infection. Nlrp12-/- mice are protected from lethality during IAV infection and show decreased vascular permeability, fewer pulmonary neutrophils, and a reduction in levels of neutrophil chemoattractant CXCL1 in their lungs compared with wild-type mice. Nlrp12-/- neutrophils and dendritic cells within the IAV-infected lungs produce less CXCL1 than their wild-type counterparts. Decreased CXCL1 production by Nlrp12-/- dendritic cells was not due to a difference in CXCL1 protein stability, but instead to a decrease in Cxcl1 mRNA stability. Together, these data demonstrate a previously unappreciated role for Nlrp12 in exacerbating the pathogenesis of IAV infection through the regulation of CXCL1-mediated neutrophilic responses.
Collapse
Affiliation(s)
- Emma E Hornick
- Interdisciplinary Program in Immunology, University of Iowa Carver College of Medicine, Iowa City, IA 52242
| | - Balaji Banoth
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Ann M Miller
- Interdisciplinary Program in Immunology, University of Iowa Carver College of Medicine, Iowa City, IA 52242
| | - Zeb R Zacharias
- Interdisciplinary Program in Immunology, University of Iowa Carver College of Medicine, Iowa City, IA 52242.,Department of Pathology, University of Iowa Carver College of Medicine, Iowa City, IA 52242
| | - Nidhi Jain
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Mary E Wilson
- Interdisciplinary Program in Immunology, University of Iowa Carver College of Medicine, Iowa City, IA 52242.,Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA 52242.,Veterans Affairs Medical Center, Iowa City, IA 52246; and
| | | | - Kevin L Legge
- Interdisciplinary Program in Immunology, University of Iowa Carver College of Medicine, Iowa City, IA 52242.,Department of Pathology, University of Iowa Carver College of Medicine, Iowa City, IA 52242.,Department of Microbiology and Immunology, University of Iowa Carver College of Medicine, Iowa City, IA 52242
| | - Gail A Bishop
- Interdisciplinary Program in Immunology, University of Iowa Carver College of Medicine, Iowa City, IA 52242.,Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA 52242.,Veterans Affairs Medical Center, Iowa City, IA 52246; and.,Department of Microbiology and Immunology, University of Iowa Carver College of Medicine, Iowa City, IA 52242
| | - Fayyaz S Sutterwala
- Interdisciplinary Program in Immunology, University of Iowa Carver College of Medicine, Iowa City, IA 52242.,Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Suzanne L Cassel
- Interdisciplinary Program in Immunology, University of Iowa Carver College of Medicine, Iowa City, IA 52242; .,Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| |
Collapse
|
16
|
Moriyama M, Takeyama H, Hasegawa H, Ichinohe T. Induction of lung CD8 + T cell responses by consecutive inoculations of a poly(I:C) influenza vaccine. Vaccine 2017; 35:6620-6626. [PMID: 29079103 DOI: 10.1016/j.vaccine.2017.10.038] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2017] [Revised: 09/05/2017] [Accepted: 10/13/2017] [Indexed: 01/24/2023]
Abstract
The cytotoxic T lymphocyte (CTL) response plays a key role in host recovery from influenza virus infection and in subsequent immunity. Compared to natural infection with influenza virus, however, intranasal vaccination with adjuvant-combined inactivated vaccine elicits only moderate CTL responses. Here we demonstrate that 5 days of consecutive, intranasal vaccination with a combination of inactivated influenza vaccine and poly(I:C) elicits a strong CTL response in the lung. Antigen-captured respiratory DCs did efficiently migrate from the lung to the mediastinal lymph node (mLN) after the 5 day series of inoculations with vaccine and poly(I:C). Importantly, formalin-inactivated whole virus vaccine and poly(I:C) adjuvant have synergic effects on consecutive vaccinations to elicit a strong CTL response in the lung. Although the CTL response was less effective against heterologous influenza virus, we show for the first time that intranasal administration of inactivated influenza virus vaccine and poly(I:C) for 5 consecutive days can elicit high levels of influenza virus-specific CD8+ T cells in the lung.
Collapse
Affiliation(s)
- Miyu Moriyama
- Division of Viral Infection, Department of Infectious Disease Control, International Research Center for Infectious Diseases, Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo 108-8639, Japan; Department of Life Science and Medical Bioscience, Waseda University, Shinjuku-ku, Tokyo 162-8480, Japan; Department of Pathology, National Institute of Infectious Diseases, Shinjuku-ku, Tokyo 162-8640, Japan
| | - Haruko Takeyama
- Department of Life Science and Medical Bioscience, Waseda University, Shinjuku-ku, Tokyo 162-8480, Japan
| | - Hideki Hasegawa
- Department of Pathology, National Institute of Infectious Diseases, Shinjuku-ku, Tokyo 162-8640, Japan
| | - Takeshi Ichinohe
- Division of Viral Infection, Department of Infectious Disease Control, International Research Center for Infectious Diseases, Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo 108-8639, Japan.
| |
Collapse
|
17
|
Orlowski P, Pardecka M, Cymerys J, Krzyzowska M. Dendritic cells during mousepox: The role of delayed apoptosis in the pathogenesis of infection. Microb Pathog 2017; 109:99-109. [PMID: 28554653 DOI: 10.1016/j.micpath.2017.05.037] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 05/14/2017] [Accepted: 05/19/2017] [Indexed: 10/19/2022]
Abstract
Dendritic cells (DCs) are effector cells linking the innate immune system with the adaptive immune response. Many viruses eliminate DCs to prevent host response, induce immunosuppression and to maintain chronic infection. In this study, we examined apoptotic response of dendritic cells during in vitro and in vivo infection with ectromelia virus (ECTV), the causative agent of mousepox. ECTV-infected bone marrow dendritic cells (BMDCs) from BALB/c mice underwent apoptosis through mitochondrial pathway at 48 h post infection, up-regulated FasL and decreased expression of anti-apoptotic Bcl-2 and pro-apoptotic Fas. Similar pattern of Bcl-2, Fas and FasL expression was observed for DCs early during in vivo infection of BALB/c mice. Both BMDCs and DCs from BALB/c mice showed no maturation upon ECTV infection. We conclude that ECTV-infected DCs from BALB/c mouse strain help the virus to spread and to maintain infection.
Collapse
Affiliation(s)
- Piotr Orlowski
- Military Institute of Hygiene and Epidemiology, Kozielska 4, 01-163 Warsaw, Poland.
| | - Maja Pardecka
- Department of Preclinical Sciences, Faculty of Veterinary Medicine, Ciszewskiego 8, 02-786, Warsaw University of Life Sciences, Warsaw, Poland
| | - Joanna Cymerys
- Department of Preclinical Sciences, Faculty of Veterinary Medicine, Ciszewskiego 8, 02-786, Warsaw University of Life Sciences, Warsaw, Poland
| | | |
Collapse
|
18
|
Haller S, Duval A, Migliorini R, Stevanin M, Mack V, Acha-Orbea H. Interleukin-35-Producing CD8α + Dendritic Cells Acquire a Tolerogenic State and Regulate T Cell Function. Front Immunol 2017; 8:98. [PMID: 28228759 PMCID: PMC5296329 DOI: 10.3389/fimmu.2017.00098] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 01/19/2017] [Indexed: 01/02/2023] Open
Abstract
Dendritic cells (DCs) play a central role in shaping immunogenic as well as tolerogenic adaptive immune responses and thereby dictate the outcome of adaptive immunity. Here, we report the generation of a CD8α+ DC line constitutively secreting the tolerogenic cytokine interleukin (IL)-35. IL-35 secretion led to impaired CD4+ and CD8+ T lymphocyte proliferation and interfered with their function in vitro and also in vivo. IL-35 was furthermore found to induce a tolerogenic phenotype on CD8α+ DCs, characterized by the upregulation of CD11b, downregulation of MHC class II, a reduced costimulatory potential as well as production of the immunomodulatory molecule IL-10. Vaccination of mice with IL-35-expressing DCs promoted tumor growth and reduced the severity of autoimmune encephalitis not only in a preventive but also after induction of encephalitogenic T cells. The reduction in experimental autoimmune encephalitis severity was significantly more pronounced when antigen-pulsed IL-35+ DCs were used. These findings suggest a new, indirect effector mechanism by which IL-35-responding antigen-presenting cells contribute to immune tolerance. Furthermore, IL-35-transfected DCs may be a promising approach for immunotherapy in the context of autoimmune diseases.
Collapse
Affiliation(s)
- Sergio Haller
- Department of Biochemistry, Center for Immunity and Infection Lausanne (CIIL), University of Lausanne , Lausanne , Switzerland
| | - Anaïs Duval
- Institut de Pharmacologie et de Biologie Structurale, Centre National de la Recherche Scientifique (CNRS), Université Paul Sabatier, Université de Toulouse , Toulouse , France
| | - Romain Migliorini
- Department of Biochemistry, Center for Immunity and Infection Lausanne (CIIL), University of Lausanne , Lausanne , Switzerland
| | - Mathias Stevanin
- Department of Biochemistry, Center for Immunity and Infection Lausanne (CIIL), University of Lausanne , Lausanne , Switzerland
| | - Vanessa Mack
- Department of Biochemistry, Center for Immunity and Infection Lausanne (CIIL), University of Lausanne , Lausanne , Switzerland
| | - Hans Acha-Orbea
- Department of Biochemistry, Center for Immunity and Infection Lausanne (CIIL), University of Lausanne , Lausanne , Switzerland
| |
Collapse
|
19
|
Channappanavar R, Fehr AR, Vijay R, Mack M, Zhao J, Meyerholz DK, Perlman S. Dysregulated Type I Interferon and Inflammatory Monocyte-Macrophage Responses Cause Lethal Pneumonia in SARS-CoV-Infected Mice. Cell Host Microbe 2016; 19:181-93. [PMID: 26867177 PMCID: PMC4752723 DOI: 10.1016/j.chom.2016.01.007] [Citation(s) in RCA: 1160] [Impact Index Per Article: 128.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Revised: 11/30/2015] [Accepted: 01/22/2016] [Indexed: 02/08/2023]
Abstract
Highly pathogenic human respiratory coronaviruses cause acute lethal disease characterized by exuberant inflammatory responses and lung damage. However, the factors leading to lung pathology are not well understood. Using mice infected with SARS (severe acute respiratory syndrome)-CoV, we show that robust virus replication accompanied by delayed type I interferon (IFN-I) signaling orchestrates inflammatory responses and lung immunopathology with diminished survival. IFN-I remains detectable until after virus titers peak, but early IFN-I administration ameliorates immunopathology. This delayed IFN-I signaling promotes the accumulation of pathogenic inflammatory monocyte-macrophages (IMMs), resulting in elevated lung cytokine/chemokine levels, vascular leakage, and impaired virus-specific T cell responses. Genetic ablation of the IFN-αβ receptor (IFNAR) or IMM depletion protects mice from lethal infection, without affecting viral load. These results demonstrate that IFN-I and IMM promote lethal SARS-CoV infection and identify IFN-I and IMMs as potential therapeutic targets in patients infected with pathogenic coronavirus and perhaps other respiratory viruses. SARS-CoV causes a lethal respiratory infection in BALB/c mice Robust SARS-CoV replication and delayed IFN-I signaling promote disease IFN-I induces influx of pathogenic inflammatory monocytes and vascular leakage Disease severity is ameliorated in the absence of IFN signaling
Collapse
Affiliation(s)
| | - Anthony R Fehr
- Department of Microbiology, University of Iowa, Iowa City, IA 52242, USA
| | - Rahul Vijay
- Interdisciplinary Program in Immunology, University of Iowa, Iowa City, IA 52242, USA
| | - Matthias Mack
- Department of Internal Medicine, University Hospital Regensburg, Regensburg 93042, Germany
| | - Jincun Zhao
- Department of Microbiology, University of Iowa, Iowa City, IA 52242, USA; State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| | - David K Meyerholz
- Department of Pathology, University of Iowa, Iowa City, IA 52242, USA
| | - Stanley Perlman
- Department of Microbiology, University of Iowa, Iowa City, IA 52242, USA; Interdisciplinary Program in Immunology, University of Iowa, Iowa City, IA 52242, USA.
| |
Collapse
|
20
|
Hartwig SM, Ketterer M, Apicella MA, Varga SM. Non-typeable Haemophilus influenzae protects human airway epithelial cells from a subsequent respiratory syncytial virus challenge. Virology 2016; 498:128-135. [PMID: 27573069 DOI: 10.1016/j.virol.2016.08.020] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Revised: 07/27/2016] [Accepted: 08/19/2016] [Indexed: 02/03/2023]
Abstract
Respiratory syncytial virus (RSV) and the common commensal and opportunistic pathogen, non-typeable Haemophilus influenzae (NTHi) both serve as a frequent cause of respiratory infection in children. Although it is well established that some respiratory viruses can increase host susceptibility to secondary bacterial infections, few studies have examined how commensal bacteria could influence a secondary viral response. Here, we examined the impact of NTHi exposure on a subsequent RSV infection of human bronchial epithelial cells (16HBE14o-). Co-culture of 16HBE14o- cells with NTHi resulted in inhibition of viral gene expression following RSV infection. 16HBE14o- cells co-cultured with heat-killed NTHi failed to protect against an RSV infection, indicating that protection requires live bacteria. However, NTHi did not inhibit influenza A virus replication, indicating that NTHi-mediated protection was RSV-specific. Our data demonstrates that prior exposure to a commensal bacterium such as NTHi can elicit protection against a subsequent RSV infection.
Collapse
Affiliation(s)
- Stacey M Hartwig
- Department of Microbiology, University of Iowa, Iowa City, IA, United States
| | - Margaret Ketterer
- Department of Microbiology, University of Iowa, Iowa City, IA, United States
| | - Michael A Apicella
- Department of Microbiology, University of Iowa, Iowa City, IA, United States
| | - Steven M Varga
- Department of Microbiology, University of Iowa, Iowa City, IA, United States; Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA, United States; Department of Pathology, University of Iowa, Iowa City, IA, United States.
| |
Collapse
|
21
|
Shaikh SR, Fessler MB, Gowdy KM. Role for phospholipid acyl chains and cholesterol in pulmonary infections and inflammation. J Leukoc Biol 2016; 100:985-997. [PMID: 27286794 PMCID: PMC5069085 DOI: 10.1189/jlb.4vmr0316-103r] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 05/19/2016] [Indexed: 12/15/2022] Open
Abstract
Review on how complex mixtures of bioactive lipids and cholesterol may influence the pulmonary immune response during infection. Bacterial and viral respiratory tract infections result in millions of deaths worldwide and are currently the leading cause of death from infection. Acute inflammation is an essential element of host defense against infection, but can be damaging to the host when left unchecked. Effective host defense requires multiple lipid mediators, which collectively have proinflammatory and/or proresolving effects on the lung. During pulmonary infections, phospholipid acyl chains and cholesterol can be chemically and enzymatically oxidized, as well as truncated and modified, producing complex mixtures of bioactive lipids. We review recent evidence that phospholipids and cholesterol and their derivatives regulate pulmonary innate and adaptive immunity during infection. We first highlight data that oxidized phospholipids generated in the lung during infection stimulate pattern recognition receptors, such as TLRs and scavenger receptors, thereby amplifying the pulmonary inflammatory response. Next, we discuss evidence that oxidation of endogenous pools of cholesterol during pulmonary infections produces oxysterols that also modify the function of both innate and adaptive immune cells. Last, we conclude with data that n‐3 polyunsaturated fatty acids, both in the form of phospholipid acyl chains and through enzymatic processing into endogenous proresolving lipid mediators, aid in the resolution of lung inflammation through distinct mechanisms. Unraveling the complex mechanisms of induction and function of distinct classes of bioactive lipids, both native and modified, may hold promise for developing new therapeutic strategies for improving pulmonary outcomes in response to infection.
Collapse
Affiliation(s)
- Saame Raza Shaikh
- Department of Biochemistry and Molecular Biology, East Carolina Diabetes and Obesity Institute, East Carolina Heart Institute, Brody School of Medicine, East Carolina University (ECU), Greenville, North Carolina, USA
| | - Michael B Fessler
- Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health (NIEHS/NIH), Research Triangle Park, North Carolina, USA
| | - Kymberly M Gowdy
- Department of Pharmacology and Toxicology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, USA;
| |
Collapse
|
22
|
Abstract
The T cell response is an integral and essential part of the host immune response to acute virus infection. Each viral pathogen has unique, frequently nuanced, aspects to its replication, which affects the host response and as a consequence the capacity of the virus to produce disease. There are, however, common features to the T cell response to viruses, which produce acute limited infection. This is true whether virus replication is restricted to a single site, for example, the respiratory tract (RT), CNS etc., or replication is in multiple sites throughout the body. In describing below the acute T cell response to virus infection, we employ acute virus infection of the RT as a convenient model to explore this process of virus infection and the host response. We divide the process into three phases: the induction (initiation) of the response, the expression of antiviral effector activity resulting in virus elimination, and the resolution of inflammation with restoration of tissue homeostasis.
Collapse
|
23
|
Vijay R, Hua X, Meyerholz DK, Miki Y, Yamamoto K, Gelb M, Murakami M, Perlman S. Critical role of phospholipase A2 group IID in age-related susceptibility to severe acute respiratory syndrome-CoV infection. ACTA ACUST UNITED AC 2015; 212:1851-68. [PMID: 26392224 PMCID: PMC4612096 DOI: 10.1084/jem.20150632] [Citation(s) in RCA: 109] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Accepted: 08/19/2015] [Indexed: 12/24/2022]
Abstract
Vijay et al. show that an age-dependent increase of phospholipase A2 group IID (PLA2G2D) in the lung contributes to worse outcomes in mice infected with SARS-CoV. Mice lacking (PLA2G2D) had increased survival to lethal infection with enhanced DC migration to the dLN and augmented T cell responses. The results suggest that targeting (PLA2G2D) in elderly patients with respiratory infections could represent an attractive therapeutic strategy. Oxidative stress and chronic low-grade inflammation in the lungs are associated with aging and may contribute to age-related immune dysfunction. To maintain lung homeostasis, chronic inflammation is countered by enhanced expression of proresolving/antiinflammatory factors. Here, we show that age-dependent increases of one such factor in the lungs, a phospholipase A2 (PLA2) group IID (PLA2G2D) with antiinflammatory properties, contributed to worse outcomes in mice infected with severe acute respiratory syndrome-coronavirus (SARS-CoV). Strikingly, infection of mice lacking PLA2G2D expression (Pla2g2d−/− mice) converted a uniformly lethal infection to a nonlethal one (>80% survival), subsequent to development of enhanced respiratory DC migration to the draining lymph nodes, augmented antivirus T cell responses, and diminished lung damage. We also observed similar effects in influenza A virus–infected middle-aged Pla2g2d−/− mice. Furthermore, oxidative stress, probably via lipid peroxidation, was found to induce PLA2G2D expression in mice and in human monocyte–derived macrophages. Thus, our results suggest that directed inhibition of a single inducible phospholipase, PLA2G2D, in the lungs of older patients with severe respiratory infections is potentially an attractive therapeutic intervention to restore immune function.
Collapse
Affiliation(s)
- Rahul Vijay
- Interdisciplinary Program in Immunology and Department of Otolaryngology, Department of Pathology, and Department of Microbiology, University of Iowa, Iowa City, IA 52242
| | - Xiaoyang Hua
- Interdisciplinary Program in Immunology and Department of Otolaryngology, Department of Pathology, and Department of Microbiology, University of Iowa, Iowa City, IA 52242
| | - David K Meyerholz
- Interdisciplinary Program in Immunology and Department of Otolaryngology, Department of Pathology, and Department of Microbiology, University of Iowa, Iowa City, IA 52242
| | - Yoshimi Miki
- Lipid Metabolism Project, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
| | - Kei Yamamoto
- Lipid Metabolism Project, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
| | - Michael Gelb
- Department of Chemistry and Department of Biochemistry, University of Washington, Seattle, WA 98195 Department of Chemistry and Department of Biochemistry, University of Washington, Seattle, WA 98195
| | - Makoto Murakami
- Lipid Metabolism Project, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency, Saitama 332-0012, Japan
| | - Stanley Perlman
- Interdisciplinary Program in Immunology and Department of Otolaryngology, Department of Pathology, and Department of Microbiology, University of Iowa, Iowa City, IA 52242 Interdisciplinary Program in Immunology and Department of Otolaryngology, Department of Pathology, and Department of Microbiology, University of Iowa, Iowa City, IA 52242
| |
Collapse
|
24
|
Fas/FasL pathway participates in regulation of antiviral and inflammatory response during mousepox infection of lungs. Mediators Inflamm 2015; 2015:281613. [PMID: 25873756 PMCID: PMC4385687 DOI: 10.1155/2015/281613] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2015] [Accepted: 02/26/2015] [Indexed: 11/25/2022] Open
Abstract
Fas receptor-Fas ligand (FasL) signalling is involved in apoptosis of immune cells as well as of the virus infected target cells but increasing evidence accumulates on Fas as a mediator of apoptosis-independent processes such as induction of activating and proinflammatory signals. In this study, we examined the role of Fas/FasL pathway in inflammatory and antiviral response in lungs using a mousepox model applied to C57BL6/J, B6. MRL-Faslpr/J, and B6Smn.C3-Faslgld/J mice. Ectromelia virus (ECTV) infection of Fas- and FasL-deficient mice led to increased virus titers in lungs and decreased migration of IFN-γ expressing NK cells, CD4+ T cells, CD8+ T cells, and decreased IL-15 expression. The lungs of ECTV-infected Fas- and FasL-deficient mice showed significant inflammation during later phases of infection accompanied by decreased expression of anti-inflammatory IL-10 and TGF-β1 cytokines and disturbances in CXCL1 and CXCL9 expression. Experiments in vitro demonstrated that ECTV-infected cultures of epithelial cells, but not macrophages, upregulate Fas and FasL and are susceptible to Fas-induced apoptosis. Our study demonstrates that Fas/FasL pathway during ECTV infection of the lungs plays an important role in controlling local inflammatory response and mounting of antiviral response.
Collapse
|
25
|
Lambrecht BN, Neyt K, van Helden MJ. The Mucosal Immune Response to Respiratory Viruses. Mucosal Immunol 2015. [DOI: 10.1016/b978-0-12-415847-4.00094-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
26
|
Chiu C, Openshaw PJ. Antiviral B cell and T cell immunity in the lungs. Nat Immunol 2015; 16:18-26. [PMID: 25521681 PMCID: PMC7097128 DOI: 10.1038/ni.3056] [Citation(s) in RCA: 115] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Accepted: 11/14/2014] [Indexed: 12/13/2022]
Abstract
Respiratory viruses are frequent causes of repeated common colds, bronchitis and pneumonia, which often occur unpredictably as epidemics and pandemics. Despite those decimating effects on health and decades of intensive research, treatments remain largely supportive. The only commonly available vaccines are against influenza virus, and even these need improvement. The lung shares some features with other mucosal sites, but preservation of its especially delicate anatomical structures necessitates a fine balance of pro- and anti-inflammatory responses; well-timed, appropriately placed and tightly regulated T cell and B cell responses are essential for protection from infection and limitation of symptoms, whereas poorly regulated inflammation contributes to tissue damage and disease. Recent advances in understanding adaptive immunity should facilitate vaccine development and reduce the global effect of respiratory viruses.
Collapse
Affiliation(s)
- Christopher Chiu
- Centre for Respiratory Infection, National Heart and Lung Institute, Imperial College London, London, UK
| | - Peter J Openshaw
- Centre for Respiratory Infection, National Heart and Lung Institute, Imperial College London, London, UK
| |
Collapse
|
27
|
|
28
|
|
29
|
Khan SH, Hemann EA, Legge KL, Norian LA, Badovinac VP. Diet-induced obesity does not impact the generation and maintenance of primary memory CD8 T cells. THE JOURNAL OF IMMUNOLOGY 2014; 193:5873-82. [PMID: 25378592 DOI: 10.4049/jimmunol.1401685] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
The extent to which obesity compromises the differentiation and maintenance of protective memory CD8 T cell responses and renders obese individuals susceptible to infection remains unknown. In this study, we show that diet-induced obesity did not impact the maintenance of pre-existing memory CD8 T cells, including acquisition of a long-term memory phenotype (i.e., CD27(hi), CD62L(hi), KLRG1(lo)) and function (i.e., cytokine production, secondary expansion, and memory CD8 T cell-mediated protection). Additionally, obesity did not influence the differentiation and maintenance of newly evoked memory CD8 T cell responses in inbred and outbred hosts generated in response to different types of systemic (LCMV, L. monocytogenes) and/or localized (influenza virus) infections. Interestingly, the rate of naive-to-memory CD8 T cell differentiation after a peptide-coated dendritic cell immunization was similar in lean and obese hosts, suggesting that obesity-associated inflammation, unlike pathogen- or adjuvant-induced inflammation, did not influence the development of endogenous memory CD8 T cell responses. Therefore, our studies reveal that the obese environment does not influence the development or maintenance of memory CD8 T cell responses that are either primed before or after obesity is established, a surprising notion with important implications for future studies aiming to elucidate the role obesity plays in host susceptibility to infections.
Collapse
Affiliation(s)
- Shaniya H Khan
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA 52242
| | - Emily A Hemann
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA 52242
| | - Kevin L Legge
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA 52242; Department of Pathology, University of Iowa, Iowa City, IA 52242; and
| | - Lyse A Norian
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA 52242; Department of Urology, University of Iowa, Iowa City, IA 52242
| | - Vladimir P Badovinac
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA 52242; Department of Pathology, University of Iowa, Iowa City, IA 52242; and
| |
Collapse
|
30
|
Batich KA, Swartz AM, Sampson JH. Enhancing dendritic cell-based vaccination for highly aggressive glioblastoma. Expert Opin Biol Ther 2014; 15:79-94. [PMID: 25327832 DOI: 10.1517/14712598.2015.972361] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Patients with primary glioblastoma (GBM) have a dismal prognosis despite standard therapy, which can induce potentially deleterious side effects. Arming the immune system is an alternative therapeutic approach, as its cellular effectors and inherent capacity for memory can be utilized to specifically target invasive tumor cells, while sparing collateral damage to otherwise healthy brain parenchyma. AREAS COVERED Active immunotherapy is aimed at eliciting a specific immune response against tumor antigens. Dendritic cells (DCs) are one of the most potent activators of de novo and recall immune responses and are thus a vehicle for successful immunotherapy. Currently, investigators are optimizing DC vaccines by enhancing maturation status and migratory potential to induce more potent antitumor responses. An update on the most recent DC immunotherapy trials is provided. EXPERT OPINION Targeting of unique antigens restricted to the tumor itself is the most important parameter in advancing DC vaccines. In order to overcome intrinsic mechanisms of immune evasion observed in GBM, the future of DC-based therapy lies in a multi-antigenic vaccine approach. Successful targeting of multiple antigens will require a comprehensive understanding of all immunologically relevant oncological epitopes present in each tumor, thereby permitting a rational vaccine design.
Collapse
Affiliation(s)
- Kristen A Batich
- Duke Brain Tumor Immunotherapy Program, Division of Neurosurgery, Department of Surgery ; Durham, NC 27710 , USA
| | | | | |
Collapse
|
31
|
Hemann EA, McGill JL, Legge KL. Chronic ethanol exposure selectively inhibits the influenza-specific CD8 T cell response during influenza a virus infection. Alcohol Clin Exp Res 2014; 38:2403-13. [PMID: 25160044 DOI: 10.1111/acer.12522] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Accepted: 06/27/2014] [Indexed: 12/18/2022]
Abstract
BACKGROUND It is well established that chronic ethanol (EtOH) consumption is associated with increased incidence and disease severity of respiratory infections. Our recent work demonstrates this increase in disease severity to influenza A virus (IAV) infections is due, in part, to a failure to mount a robust IAV-specific CD8 T cell response along with a specific impairment in the ability of these T cells to produce interferon γ (IFNγ). However, the full extent of the lesion in the effector CD8 T cell compartment during chronic EtOH consumption remains unknown. METHODS Utilizing the Meadows-Cook murine model of chronic alcohol consumption, mice received EtOH in their drinking water for 8 or 12 weeks. Mice were challenged intranasally with IAV, and the activation and effector functions of IAV-specific CD8 T cells were determined in both the lung-draining lymph nodes (dLN) and lungs. RESULTS Our results confirm the defect in IFNγ production; however, the ability of IAV-specific T cells to produce tumor necrosis factor α (TNFα) and interleukin-2 (IL-2) in EtOH-consuming mice remains unaltered. In contrast, EtOH consumption significantly reduces the ability of CD8 T cells to degranulate and kill IAV-specific targets. Finally, our findings suggest the lesion begins during the initial activation of CD8 T cells, as we observe early defects in proliferation in the dLN of IAV-infected, EtOH-consuming mice. CONCLUSIONS These findings highlight the previously unrecognized depth of the lesion in the IAV-specific CD8 T cell response during chronic EtOH consumption. Given the important role CD8 T cell immunity plays in control of IAV, these findings may aid in the development of vaccination and/or therapeutic strategies to reverse these defects in the CD8 T cell response and reduce serious disease outcomes associated with IAV infections in alcoholics.
Collapse
Affiliation(s)
- Emily A Hemann
- Department of Pathology and Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, Iowa
| | | | | |
Collapse
|
32
|
Boonnak K, Vogel L, Feldmann F, Feldmann H, Legge KL, Subbarao K. Lymphopenia associated with highly virulent H5N1 virus infection due to plasmacytoid dendritic cell-mediated apoptosis of T cells. THE JOURNAL OF IMMUNOLOGY 2014; 192:5906-12. [PMID: 24829418 DOI: 10.4049/jimmunol.1302992] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Although lymphopenia is a hallmark of severe infection with highly pathogenic H5N1 and the newly emerged H7N9 influenza viruses in humans, the mechanism(s) by which lethal H5N1 viruses cause lymphopenia in mammalian hosts remains poorly understood. Because influenza-specific T cell responses are initiated in the lung draining lymph nodes (LNs), and lymphocytes subsequently traffic to the lungs or peripheral circulation, we compared the immune responses in the lung draining LNs postinfection with a lethal A/HK/483/97 or nonlethal A/HK/486/97 (H5N1) virus in a mouse model. We found that lethal H5N1, but not nonlethal H5N1, virus infection in mice enhances Fas ligand (FasL) expression on plasmacytoid dendritic cells (pDCs), resulting in apoptosis of influenza-specific CD8(+) T cells via a Fas-FasL-mediated pathway. We also found that pDCs, but not other DC subsets, preferentially accumulate in the lung draining LNs of lethal H5N1 virus-infected mice, and that the induction of FasL expression on pDCs correlates with high levels of IL-12p40 monomer/homodimer in the lung draining LNs. Our data suggest that one of the mechanisms of lymphopenia associated with lethal H5N1 virus infection involves a deleterious role for pDCs.
Collapse
Affiliation(s)
- Kobporn Boonnak
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Leatrice Vogel
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Friederike Feldmann
- Veterinary Branch, Rocky Mountain Laboratory, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840
| | - Heinz Feldmann
- Laboratory of Virology, Rocky Mountain Laboratory, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840; and
| | - Kevin L Legge
- Department of Pathology, University of Iowa, Iowa City, IA 52242
| | - Kanta Subbarao
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892;
| |
Collapse
|
33
|
Neyt K, Lambrecht BN. The role of lung dendritic cell subsets in immunity to respiratory viruses. Immunol Rev 2014; 255:57-67. [PMID: 23947347 DOI: 10.1111/imr.12100] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Viral infections are a common cause of acute respiratory disease. The clinical course of infection and symptoms depend on the viral strain, the health status of the host, and the immunological status of the host. Dendritic cells (DCs) play a crucial role in recognizing and presenting viral antigens and in inducing adaptive immune responses that clear the virus. Because the lung is continuously exposed to the air, the lung is equipped with an elaborate network of DCs to sense incoming foreign pathogens. Increasing knowledge on DC biology has informed us that DCs are not a single cell type. In the steady state lung, three DC subsets can be defined: CD11b(+) or CD103(+) conventional DCs and plasmacytoid DCs. Upon inflammation, inflammatory monocyte-derived DCs are recruited to the lung. It is only recently that tools became available to allow DC subsets to be clearly studied. This review focuses on the activation of DCs and the function of lung DCs in the context of respiratory virus infection and highlights some cautionary points for interpreting older experiments.
Collapse
Affiliation(s)
- Katrijn Neyt
- VIB Inflammation Research Center, Laboratory of Immunoregulation, Ghent, Belgium
| | | |
Collapse
|
34
|
Slütter B, Pewe LL, Kaech SM, Harty JT. Lung airway-surveilling CXCR3(hi) memory CD8(+) T cells are critical for protection against influenza A virus. Immunity 2014; 39:939-48. [PMID: 24238342 DOI: 10.1016/j.immuni.2013.09.013] [Citation(s) in RCA: 178] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2013] [Accepted: 09/18/2013] [Indexed: 01/06/2023]
Abstract
Inducing memory CD8(+) T cells specific for conserved antigens from influenza A virus (IAV) is a potential strategy for broadly protective vaccines. Here we show that memory CD8(+) T cells in the airways played an important role in early control of IAV. Expression of the chemokine receptor CXCR3 was critical for memory CD8(+) T cells to populate the airways during the steady state and vaccination approaches were designed to favor the establishment of memory CD8(+) T cells in the airways. Specifically, we found that interleukin-12 (IL-12) signaling shortly after immunization limited CXCR3 expression on memory CD8(+) T cells. Neutralization of IL-12 or adjuvants that did not induce high amounts of IL-12 enhanced CXCR3 expression, sustained airway localization of memory CD8(+) T cells, and resulted in superior protection against IAV.
Collapse
Affiliation(s)
- Bram Slütter
- Department of Microbiology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | | | | | | |
Collapse
|
35
|
Handel A, Akin V, Pilyugin SS, Zarnitsyna V, Antia R. How sticky should a virus be? The impact of virus binding and release on transmission fitness using influenza as an example. J R Soc Interface 2014; 11:20131083. [PMID: 24430126 DOI: 10.1098/rsif.2013.1083] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Budding viruses face a trade-off: virions need to efficiently attach to and enter uninfected cells while newly generated virions need to efficiently detach from infected cells. The right balance between attachment and detachment-the right amount of stickiness-is needed for maximum fitness. Here, we design and analyse a mathematical model to study in detail the impact of attachment and detachment rates on virus fitness. We apply our model to influenza, where stickiness is determined by a balance of the haemagglutinin (HA) and neuraminidase (NA) proteins. We investigate how drugs, the adaptive immune response and vaccines impact influenza stickiness and fitness. Our model suggests that the location in the 'stickiness landscape' of the virus determines how well interventions such as drugs or vaccines are expected to work. We discuss why hypothetical NA enhancer drugs might occasionally perform better than the currently available NA inhibitors in reducing virus fitness. We show that an increased antibody or T-cell-mediated immune response leads to maximum fitness at higher stickiness. We further show that antibody-based vaccines targeting mainly HA or NA, which leads to a shift in stickiness, might reduce virus fitness above what can be achieved by the direct immunological action of the vaccine. Overall, our findings provide potentially useful conceptual insights for future vaccine and drug development and can be applied to other budding viruses beyond influenza.
Collapse
Affiliation(s)
- Andreas Handel
- Department of Epidemiology and Biostatistics, College of Public Health, University of Georgia, , Athens, GA 30602, USA
| | | | | | | | | |
Collapse
|
36
|
Li Y, Handel A. Modeling inoculum dose dependent patterns of acute virus infections. J Theor Biol 2014; 347:63-73. [PMID: 24440713 DOI: 10.1016/j.jtbi.2014.01.008] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2013] [Revised: 12/31/2013] [Accepted: 01/06/2014] [Indexed: 12/24/2022]
Abstract
Inoculum dose, i.e. the number of pathogens at the beginning of an infection, often affects key aspects of pathogen and immune response dynamics. These in turn determine clinically relevant outcomes, such as morbidity and mortality. Despite the general recognition that inoculum dose is an important component of infection outcomes, we currently do not understand its impact in much detail. This study is intended to start filling this knowledge gap by analyzing inoculum dependent patterns of viral load dynamics in acute infections. Using experimental data for adenovirus and infectious bronchitis virus infections as examples, we demonstrate inoculum dose dependent patterns of virus dynamics. We analyze the data with the help of mathematical models to investigate what mechanisms can reproduce the patterns observed in experimental data. We find that models including components of both the innate and adaptive immune response are needed to reproduce the patterns found in the data. We further analyze which types of innate or adaptive immune response models agree with observed data. One interesting finding is that only models for the adaptive immune response that contain growth terms partially independent of viral load can properly reproduce observed patterns. This agrees with the idea that an antigen-independent, programmed response is part of the adaptive response. Our analysis provides useful insights into the types of model structures that are required to properly reproduce observed virus dynamics for varying inoculum doses. We suggest that such models should be taken as basis for future models of acute viral infections.
Collapse
Affiliation(s)
- Yan Li
- Institute of Bioinformatics, The University of Georgia, Athens, GA, USA
| | - Andreas Handel
- Department of Epidemiology and Biostatistics, The University of Georgia, Athens, GA, USA.
| |
Collapse
|
37
|
Abstract
Influenza virus infection induces a potent initial innate immune response, which serves to limit the extent of viral replication and virus spread. However, efficient (and eventual) viral clearance within the respiratory tract requires the subsequent activation, rapid proliferation, recruitment, and expression of effector activities by the adaptive immune system, consisting of antibody producing B cells and influenza-specific T lymphocytes with diverse functions. The ensuing effector activities of these T lymphocytes ultimately determine (along with antibodies) the capacity of the host to eliminate the viruses and the extent of tissue damage. In this review, we describe this effector T cell response to influenza virus infection. Based on information largely obtained in experimental settings (i.e., murine models), we will illustrate the factors regulating the induction of adaptive immune T cell responses to influenza, the effector activities displayed by these activated T cells, the mechanisms underlying the expression of these effector mechanisms, and the control of the activation/differentiation of these T cells, in situ, in the infected lungs.
Collapse
|
38
|
Gao R, Bhatnagar J, Blau DM, Greer P, Rollin DC, Denison AM, Deleon-Carnes M, Shieh WJ, Sambhara S, Tumpey TM, Patel M, Liu L, Paddock C, Drew C, Shu Y, Katz JM, Zaki SR. Cytokine and chemokine profiles in lung tissues from fatal cases of 2009 pandemic influenza A (H1N1): role of the host immune response in pathogenesis. THE AMERICAN JOURNAL OF PATHOLOGY 2013; 183:1258-1268. [PMID: 23938324 PMCID: PMC7119452 DOI: 10.1016/j.ajpath.2013.06.023] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/23/2013] [Revised: 06/21/2013] [Accepted: 06/24/2013] [Indexed: 12/18/2022]
Abstract
Pathological studies on fatal cases caused by 2009 pandemic influenza H1N1 virus (2009 pH1N1) reported extensive diffuse alveolar damage and virus infection predominantly in the lung parenchyma. However, the host immune response after severe 2009 pH1N1 infection is poorly understood. Herein, we investigated viral load, the immune response, and apoptosis in lung tissues from 50 fatal cases with 2009 pH1N1 virus infection. The results suggested that 7 of the 27 cytokines/chemokines showed remarkably high expression, including IL-1 receptor antagonist protein, IL-6, tumor necrosis factor-α, IL-8, monocyte chemoattractant protein-1, macrophage inflammatory protein 1-β, and interferon-inducible protein-10 in lung tissues of 2009 pH1N1 fatal cases. Viral load, which showed the highest level on day 7 of illness onset and persisted until day 17 of illness, was positively correlated with mRNA levels of IL-1 receptor antagonist protein, monocyte chemoattractant protein-1, macrophage inflammatory protein 1-β, interferon-inducible protein-10, and regulated on activation normal T-cell expressed and secreted. Apoptosis was evident in lung tissues stained by the TUNEL assay. Decreased Fas and elevated FasL mRNA levels were present in lung tissues, and cleaved caspase-3 was frequently seen in pneumocytes, submucosal glands, and lymphoid tissues. The pathogenesis of the 2009 pH1N1 virus infection is associated with viral replication and production of proinflammatory mediators. FasL and caspase-3 are involved in the pathway of 2009 pH1N1 virus-induced apoptosis in lung tissues, and the disequilibrium between the Fas and FasL level in lung tissues could contribute to delayed clearance of the virus and subsequent pathological damages.
Collapse
Affiliation(s)
- Rongbao Gao
- Department of Influenza, State Key Laboratory for Molecular Virology and Genetic Engineering, Chinese National Influenza Center, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Julu Bhatnagar
- Infectious Diseases Pathology Branch, the Division of High-Consequence Pathogens and Pathology, the Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Dianna M Blau
- Infectious Diseases Pathology Branch, the Division of High-Consequence Pathogens and Pathology, the Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Patricia Greer
- Infectious Diseases Pathology Branch, the Division of High-Consequence Pathogens and Pathology, the Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Dominique C Rollin
- Infectious Diseases Pathology Branch, the Division of High-Consequence Pathogens and Pathology, the Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Amy M Denison
- Infectious Diseases Pathology Branch, the Division of High-Consequence Pathogens and Pathology, the Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Marlene Deleon-Carnes
- Infectious Diseases Pathology Branch, the Division of High-Consequence Pathogens and Pathology, the Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Wun-Ju Shieh
- Infectious Diseases Pathology Branch, the Division of High-Consequence Pathogens and Pathology, the Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Suryaprakash Sambhara
- Immunology and Pathogenesis Branch, the Influenza Division, the Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Terrence M Tumpey
- Immunology and Pathogenesis Branch, the Influenza Division, the Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Mitesh Patel
- Infectious Diseases Pathology Branch, the Division of High-Consequence Pathogens and Pathology, the Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Lindy Liu
- Infectious Diseases Pathology Branch, the Division of High-Consequence Pathogens and Pathology, the Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Christopher Paddock
- Infectious Diseases Pathology Branch, the Division of High-Consequence Pathogens and Pathology, the Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Clifton Drew
- Infectious Diseases Pathology Branch, the Division of High-Consequence Pathogens and Pathology, the Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Yuelong Shu
- Department of Influenza, State Key Laboratory for Molecular Virology and Genetic Engineering, Chinese National Influenza Center, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Jacqueline M Katz
- Immunology and Pathogenesis Branch, the Influenza Division, the Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Sherif R Zaki
- Department of Influenza, State Key Laboratory for Molecular Virology and Genetic Engineering, Chinese National Influenza Center, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China; Infectious Diseases Pathology Branch, the Division of High-Consequence Pathogens and Pathology, the Centers for Disease Control and Prevention, Atlanta, Georgia.
| |
Collapse
|
39
|
Hemann EA, Kang SM, Legge KL. Protective CD8 T cell-mediated immunity against influenza A virus infection following influenza virus-like particle vaccination. THE JOURNAL OF IMMUNOLOGY 2013; 191:2486-94. [PMID: 23885108 DOI: 10.4049/jimmunol.1300954] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The development of influenza A virus (IAV) vaccines capable of inducing cytotoxic CD8 T cell responses could potentially provide superior, long-term protection against multiple, heterologous strains of IAV. Although prior studies demonstrated the effectiveness of baculovirus-derived virus-like particle (VLP) vaccination in generating Ab-mediated protection, the role that CD8 T cell immunity plays in overall VLP-mediated protection is less-well understood. In this article, we demonstrate that intranasal vaccination of mice with a VLP containing the hemagglutinin and matrix 1 proteins of IAV/PR/8/34 leads to a significant increase in hemagglutinin 533-specific CD8 T cells in the lungs and protection following subsequent homologous challenge with IAV. VLP-mediated protection was significantly reduced by CD8 T cell depletion, indicating a critical role for CD8 T cells in protective immunity. Importantly, our results show that VLP vaccine-induced CD8 T cell-mediated protection is not limited to homologous IAV strains. VLP vaccination leads to an increase in protection following heterosubtypic challenge with a strain of IAV that avoids vaccine-induced neutralizing Abs but contains conserved, immunodominant CD8 T cell epitopes. Overall, our results demonstrate the ability of influenza protein-containing VLPs to prime IAV-specific CD8 T cell responses that contribute to protection from homo- and heterosubtypic IAV infections. These results further suggest that vaccination strategies focused on the development of cross-protective CD8 T cell responses may contribute to the development of "universal" IAV vaccines.
Collapse
Affiliation(s)
- Emily A Hemann
- Department of Pathology and Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA 52242, USA
| | | | | |
Collapse
|
40
|
Slütter B, Pewe LL, Lauer P, Harty JT. Cutting edge: rapid boosting of cross-reactive memory CD8 T cells broadens the protective capacity of the Flumist vaccine. THE JOURNAL OF IMMUNOLOGY 2013; 190:3854-8. [PMID: 23467935 DOI: 10.4049/jimmunol.1202790] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Memory CD8 T cells recognizing conserved proteins from influenza A virus (IAV), such as nucleoprotein, have the potential to provide protection in individuals who lack the proper neutralizing Abs. In this study, we show that the most potent CD8 T cell-inducing influenza vaccine on the market (Flumist) does not induce sufficient numbers of cross-reactive CD8 T cells to provide substantial protection against lethal nonhomologous IAV challenge. However, Flumist-primed CD8 T cells rapidly acquire memory characteristics and can respond to short-interval boosting to greatly enlarge the IAV-specific memory pool, which is sufficient to protect mice from nonhomologous IAV challenge. Thus, a current vaccine strategy, Flumist, may serve as a priming platform for the rapid induction of large numbers of memory CD8 T cells with the capacity for broad protection against influenza.
Collapse
Affiliation(s)
- Bram Slütter
- Department of Microbiology, University of Iowa, Iowa City, IA 52242, USA
| | | | | | | |
Collapse
|
41
|
Fujikura D, Chiba S, Muramatsu D, Kazumata M, Nakayama Y, Kawai T, Akira S, Kida H, Miyazaki T. Type-I interferon is critical for FasL expression on lung cells to determine the severity of influenza. PLoS One 2013; 8:e55321. [PMID: 23408968 PMCID: PMC3568138 DOI: 10.1371/journal.pone.0055321] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2012] [Accepted: 12/21/2012] [Indexed: 01/09/2023] Open
Abstract
Infection of influenza A virus in mammals induces hyper lung pneumonia, which often causes lethal diseases. FasL is a specific ligand of Fas, which is a type-I transmembrane protein to induce cell death. Previously, it has been reported that the hyper induction of gene expression associated with Fas signal is observed in lethal influenza A virus infection. More importantly, it was also reported that functional mutation of the FasL gene protects the host against influenza A virus infection. These observations suggest that induction of FasL signal is functionally associated with the severity of influenza. However, regulation of the induction of FasL or Fas by influenza A virus infection is still unknown. Here, we demonstrated that FasL is induced after the viral infection, and inhibition of the Fas/FasL signal by treatment with a recombinant decoy receptor for FasL (Fas-Fc) increases the survival rate of mice after lethal infection of influenza A virus as well as functional mutation of the FasL gene in gld/gld mice. In addition, the induction level of FasL gene expression in the lung was correlated with the severity of influenza. We also showed that a variety of types of cells in the lung express FasL after the viral infection. Furthermore, type-I interferon induced by the viral infection was shown to be critical for induction of FasL protein expression in the lung. These findings suggested that expression of FasL protein induced by type-I IFN on the lung cell surface is critical to determine the severity of influenza.
Collapse
Affiliation(s)
- Daisuke Fujikura
- Department of Bioresources, Hokkaido University Research Center for Zoonosis Control, Kita-ku, Sapporo, Hokkaido, Japan
- Japan Science and Technology Agency, Innovation Plaza Hokkaido, Kita-ku, Sapporo, Hokkaido, 060-0819, Japan
| | - Satoko Chiba
- Department of Bioresources, Hokkaido University Research Center for Zoonosis Control, Kita-ku, Sapporo, Hokkaido, Japan
| | - Daisuke Muramatsu
- Department of Bioresources, Hokkaido University Research Center for Zoonosis Control, Kita-ku, Sapporo, Hokkaido, Japan
- Aureo Science Co., Ltd., Kita-ku, Sapporo, Hokkaido, Japan
| | - Mika Kazumata
- Department of Bioresources, Hokkaido University Research Center for Zoonosis Control, Kita-ku, Sapporo, Hokkaido, Japan
- Aureo Science Co., Ltd., Kita-ku, Sapporo, Hokkaido, Japan
| | - Yosuke Nakayama
- Department of Bioresources, Hokkaido University Research Center for Zoonosis Control, Kita-ku, Sapporo, Hokkaido, Japan
| | - Taro Kawai
- Laboratory of Host Defense, World Premier International Research Center, Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
- Department of Host Defense, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Shizuo Akira
- Laboratory of Host Defense, World Premier International Research Center, Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
- Department of Host Defense, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Hiroshi Kida
- Japan Science and Technology Agency, Innovation Plaza Hokkaido, Kita-ku, Sapporo, Hokkaido, 060-0819, Japan
- Hokkaido University Research Center for Zoonosis Control, Kita-ku, Sapporo, Hokkaido, Japan
- Graduate School of Veterinary Medicine, Hokkaido University, Kita-ku, Sapporo, Hokkaido, Japan
- Office International des Epizooties (OIE) Reference Laboratory for Highly Pathogenic Avian Influenza, Sapporo, Hokkaido, Japan
| | - Tadaaki Miyazaki
- Department of Bioresources, Hokkaido University Research Center for Zoonosis Control, Kita-ku, Sapporo, Hokkaido, Japan
- Japan Science and Technology Agency, Innovation Plaza Hokkaido, Kita-ku, Sapporo, Hokkaido, 060-0819, Japan
- * E-mail:
| |
Collapse
|
42
|
Zhao L, Tyrrell DL. Myeloid dendritic cells can kill T cells during chronic hepatitis C virus infection. Viral Immunol 2013; 26:25-39. [PMID: 23374153 DOI: 10.1089/vim.2012.0058] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Myeloid dendritic cells (mDCs) are the most potent professional antigen-presenting cells that regulate specific T-cell responses. Here we studied the ability of mDCs to kill T cells during HCV infection. We found that mDCs from chronic hepatitis C (CHC) patients expressed upregulated levels of two inhibitory ligands, Fas ligand and the ligand 2 of PD-1 (PD-L2), compared to healthy mDCs. However, their expression of the ligand 1 of PD-1 (PD-L1), tumor necrosis factor-related apoptosis inducing ligand (TRAIL), and B lymphocyte stimulator (BLyS) on the cell surface was comparable to healthy mDCs. CHC patient mDCs had cytotoxic effects on autologous patient T cells and allogeneic healthy T cells. CHC patient T cells had increased expression of PD-1 compared to healthy T cells. These results indicate that the cytotoxic activity of mDCs is upregulated to kill T cells during chronic HCV infection, which represents a novel mechanism of HCV immune evasion.
Collapse
Affiliation(s)
- Li Zhao
- Li KaShing Institute of Virology, Department of Medical Microbiology and Immunology, University of Alberta , Edmonton, Alberta, Canada.
| | | |
Collapse
|
43
|
Abstract
Influenza A virus (IAV) is a dangerous virus equipped with the potential to evoke widespread pandemic disease. The 2009 H1N1 pandemic highlights the urgency for developing effective therapeutics against IAV infection. Vaccination is a major weapon to combat IAV and efforts to improve current regimes are critically important. Here, we will review the role of dendritic cells (DCs), a pivotal cell type in the initiation of robust IAV immunity. The complexity of DC subset heterogeneity in the respiratory tract and lymph node that drains the IAV infected lung will be discussed, together with the varied and in some cases, conflicting contributions of individual DC populations to presenting IAV associated antigen to T cells.
Collapse
Affiliation(s)
- Jason Waithman
- Telethon Institute for Child Health Research, Centre for Child Health Research, The University of Western Australia, West Perth, WA, Australia
| | | |
Collapse
|
44
|
Intranasal treatment with poly(I•C) protects aged mice from lethal respiratory virus infections. J Virol 2012; 86:11416-24. [PMID: 22915814 DOI: 10.1128/jvi.01410-12] [Citation(s) in RCA: 114] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
In the 2002-2003 severe acute respiratory syndrome coronavirus (SARS-CoV) epidemic, no patients under 24 years of age died, while mortality was greater than 50% in those over 65 years. Greater than 90% of all deaths from influenza A virus (IAV) occur in the elderly (>65 years of age). To address this age-related susceptibility to SARS-CoV and IAV, we infected C57BL/6 (B6) mice with mouse-adapted SARS-CoV (MA15) or IAV (PR8), both of which cause severe disease in aged mice. Intranasal pretreatment of aged mice with poly(I·C) (a TLR3 agonist) and, to a lesser extent, CpG, R848, or lipopolysaccharide (TLR9, TLR7/8, or TLR4 agonists), provided a high level of protection [90% to 100% survival rate after poly(I·C) treatment] against lethal MA15 or IAV challenge and reduced pathological changes and virus loads in the lungs at early times after infection. Poly(I·C) pretreatment upregulated beta interferon (IFN-β), IFN-γ, IL-1β, and tumor necrosis factor (TNF) gene expression in the lungs. Intranasal pretreatment with IFN-β or IFN-γ but not IL-1β or TNF also protected aged mice, consistent with the notion that poly(I·C) pretreatment functioned, at least in part, by inducing IFN-β and IFN-γ. We also identified a potential cellular target for poly(I·C) by showing that treatment inhibited virus replication in primary human airway epithelial cells. These results suggest that intranasal poly(I·C) should be evaluated as a prophylactic agent in aged individuals at high risk for contracting SARS-CoV or IAV infections.
Collapse
|
45
|
Increased protection from vaccinia virus infection in mice genetically prone to lymphoproliferative disorders. J Virol 2012; 86:6010-22. [PMID: 22438562 DOI: 10.1128/jvi.07176-11] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Mutations in the genes that encode Fas or Fas ligand (FasL) can result in poor restraints on lymphocyte activation and in increased susceptibility to autoimmune disorders. Because these mutations portend a continuously activated immune state, we hypothesized that they might in some cases confer resistance to infection. To examine this possibility, the immune response to, morbidity caused by, and clearance of vaccinia virus (VACV) Western Reserve was examined in 5- to 7-week-old Fas mutant (lpr) mice, before an overt lymphoproliferative disorder was observable. On day 6 after VACV infection, C57BL/6-lpr (B6-lpr) mice had decreased morbidity, decreased viral titers, and an increased percentage and number of CD4(+) and CD8(+) T cells. As early as day 2 after infection, B6-lpr mice had decreased liver and spleen viral titers and increased numbers of and increased gamma interferon (IFN-γ) production by several different effector cell populations. Depletion of individual effector cell subsets did not inhibit the resistance of B6-lpr mice. Uninfected B6-lpr mice also had increased numbers of NK cells, γδ(+) T cells, and CD44(+) CD4(+) and CD44(+) CD8(+) T cells compared to uninfected B6 mice. Antibody to IFN-γ resulted in increased virus load in both B6 and B6-lpr mice and eliminated the differences in viral titers between them. These results suggest that IFN-γ produced by multiple activated leukocyte populations in Fas-deficient hosts enhances resistance to some viral infections.
Collapse
|
46
|
Herold S, Ludwig S, Pleschka S, Wolff T. Apoptosis signaling in influenza virus propagation, innate host defense, and lung injury. J Leukoc Biol 2012; 92:75-82. [PMID: 22345705 DOI: 10.1189/jlb.1011530] [Citation(s) in RCA: 96] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Programmed cell death is a crucial cellular response frequently observed in IV-infected tissue. This article reviews the current knowledge on the molecular virus-host interactions that induce apoptosis pathways in an IV-infected cell and the functional implications of these cellular signaling events on viral propagation at distinct steps during the viral replication cycle. Furthermore, it summarizes the role of IV-induced apoptosis pathways in equilibrating the host's antiviral immune response between effective viral clearance and development of severe apoptotic lung injury.
Collapse
Affiliation(s)
- Susanne Herold
- University of Giessen Lung Center, Department of Internal Medicine II, Giessen, Germany.
| | | | | | | |
Collapse
|
47
|
LaCasse CJ, Janikashvili N, Larmonier CB, Alizadeh D, Hanke N, Kartchner J, Situ E, Centuori S, Har-Noy M, Bonnotte B, Katsanis E, Larmonier N. Th-1 lymphocytes induce dendritic cell tumor killing activity by an IFN-γ-dependent mechanism. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2011; 187:6310-7. [PMID: 22075702 PMCID: PMC3297475 DOI: 10.4049/jimmunol.1101812] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Dendritic cells (DCs) encompass a heterogeneous population of cells capable of orchestrating innate and adaptive immune responses. The ability of DCs to act as professional APCs has been the foundation for the development and use of these cells as vaccines in cancer immunotherapy. DCs are also endowed with the nonconventional property of directly killing tumor cells. The current study investigates the regulation of murine DC cytotoxic function by T lymphocytes. We provide evidence that CD4(+) Th-1, but not Th-2, Th-17 cells, or regulatory T cells, are capable of inducing DC cytotoxic function. IFN-γ was identified as the major factor responsible for Th-1-induced DC tumoricidal activity. Tumor cell killing mediated by Th-1-activated killer DCs was dependent on inducible NO synthase expression and NO production. Importantly, Th-1-activated killer DCs were capable of presenting the acquired Ags from the killed tumor cells to T lymphocytes in vitro or in vivo. These observations offer new possibilities for the application of killer DCs in cancer immunotherapy.
Collapse
MESH Headings
- Animals
- Antigen Presentation/genetics
- Antigen Presentation/immunology
- Bone Marrow Cells/cytology
- Bone Marrow Cells/immunology
- Bone Marrow Cells/metabolism
- Cell Line, Tumor
- Coculture Techniques
- Cytotoxicity, Immunologic/genetics
- Dendritic Cells/immunology
- Dendritic Cells/metabolism
- Female
- Interferon-gamma/metabolism
- Interferon-gamma/physiology
- Mammary Neoplasms, Experimental/immunology
- Mammary Neoplasms, Experimental/pathology
- Mammary Neoplasms, Experimental/therapy
- Melanoma, Experimental/immunology
- Melanoma, Experimental/pathology
- Melanoma, Experimental/therapy
- Mice
- Mice, 129 Strain
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Receptors, Interferon/deficiency
- Receptors, Interferon/genetics
- Receptors, Interferon/physiology
- Th1 Cells/immunology
- Th1 Cells/metabolism
- Interferon gamma Receptor
Collapse
Affiliation(s)
- Collin J. LaCasse
- Department of Pediatrics, University of Arizona, Tucson, AZ 85724-5073
- Department of Immunobiology, University of Arizona, Tucson, AZ 85724-5073
| | - Nona Janikashvili
- Department of Pediatrics, University of Arizona, Tucson, AZ 85724-5073
| | | | - Darya Alizadeh
- Department of Pediatrics, University of Arizona, Tucson, AZ 85724-5073
- Cancer Biology Graduate Interdisciplinary Program, University of Arizona, Tucson, AZ 85724-5073
| | - Neale Hanke
- Department of Pediatrics, University of Arizona, Tucson, AZ 85724-5073
- Cancer Biology Graduate Interdisciplinary Program, University of Arizona, Tucson, AZ 85724-5073
| | - Jessica Kartchner
- Department of Pediatrics, University of Arizona, Tucson, AZ 85724-5073
| | - Elaine Situ
- Department of Pediatrics, University of Arizona, Tucson, AZ 85724-5073
| | - Sara Centuori
- Department of Pediatrics, University of Arizona, Tucson, AZ 85724-5073
- Cancer Biology Graduate Interdisciplinary Program, University of Arizona, Tucson, AZ 85724-5073
| | - Michael Har-Noy
- Immunovative Therapies Ltd, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | | | - Emmanuel Katsanis
- Department of Pediatrics, University of Arizona, Tucson, AZ 85724-5073
- Department of Immunobiology, University of Arizona, Tucson, AZ 85724-5073
- Cancer Biology Graduate Interdisciplinary Program, University of Arizona, Tucson, AZ 85724-5073
- BIO5 Institute and Arizona Cancer Center, University of Arizona, Tucson, AZ 85724-5073
| | - Nicolas Larmonier
- Department of Pediatrics, University of Arizona, Tucson, AZ 85724-5073
- Department of Immunobiology, University of Arizona, Tucson, AZ 85724-5073
- Cancer Biology Graduate Interdisciplinary Program, University of Arizona, Tucson, AZ 85724-5073
- BIO5 Institute and Arizona Cancer Center, University of Arizona, Tucson, AZ 85724-5073
| |
Collapse
|
48
|
Zhao J, Zhao J, Legge K, Perlman S. Age-related increases in PGD(2) expression impair respiratory DC migration, resulting in diminished T cell responses upon respiratory virus infection in mice. J Clin Invest 2011; 121:4921-30. [PMID: 22105170 DOI: 10.1172/jci59777] [Citation(s) in RCA: 215] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2011] [Accepted: 10/05/2011] [Indexed: 01/19/2023] Open
Abstract
The morbidity and mortality associated with respiratory virus infection is felt most keenly among the elderly. T cells are necessary for viral clearance, and many age-dependent intrinsic T cell defects have been documented. However, the development of robust T cell responses in the lung also requires respiratory DCs (rDCs), which must process antigen and migrate to draining LNs (DLNs), and little is known about age-related defects in these T cell-extrinsic functions. Here, we show that increases in prostaglandin D(2) (PGD(2)) expression in mouse lungs upon aging correlate with a progressive impairment in rDC migration to DLNs. Decreased rDC migration resulted in diminished T cell responses and more severe clinical disease in older mice infected with respiratory viruses. Diminished rDC migration associated with virus-specific defects in T cell responses and was not a result of cell-intrinsic defect, rather it reflected the observed age-dependent increases in PGD(2) expression. Blocking PGD(2) function with small-molecule antagonists enhanced rDC migration, T cell responses, and survival. This effect correlated with upregulation on rDCs of CCR7, a chemokine receptor involved in DC chemotaxis. Our results suggest that inhibiting PGD(2) function may be a useful approach to enhance T cell responses against respiratory viruses in older humans.
Collapse
Affiliation(s)
- Jincun Zhao
- Department of Microbiology, University of Iowa, Iowa City, Iowa 52242, USA
| | | | | | | |
Collapse
|
49
|
Gong D, Farley K, White M, Hartshorn KL, Benarafa C, Remold-O'Donnell E. Critical role of serpinB1 in regulating inflammatory responses in pulmonary influenza infection. J Infect Dis 2011; 204:592-600. [PMID: 21791661 DOI: 10.1093/infdis/jir352] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Excessive inflammatory host response increases morbidity and mortality associated with seasonal respiratory influenza, and highly pathogenic virus strains are characterized by massive infiltration of monocytes and/or macrophages that produce a storm of injurious cytokines. METHODS Here, we examined the role in respiratory influenza of serpinB1, an endogenous inhibitor of the serine proteases elastase, cathepsin G, and proteinase-3, increasingly recognized as regulators of inflammation. RESULTS After challenge with high-dose surfactant protein-D (SP-D)-sensitive influenza A/Philadelphia/82 (H3N2), serpinB1(-/-) mice died earlier and in greater numbers than did wild-type mice. Sublethally infected animals suffered increased morbidity, delayed resolution of epithelial injury, and increased immune cell death. Viral clearance and SP-D/SP-A upregulation were unimpaired and so were early virus-induced cytokine and chemokine burst and influx of large numbers of neutrophils and monocytes. Whereas initial cytokines and chemokines rapidly cleared in wild-type mice, TNF-α, IL-6, KC/CXCL1, G-CSF, IL-17A, and MCP-1/CCL2 remained elevated in serpinB1(-/-) mice. Monocyte-derived cells were the dominant immune cells in influenza-infected lungs, and those from serpinB1(-/-) mice produced excessive IL-6 and TNF-α when tested ex vivo. Pulmonary γδ T-cells that produced IL-17A were also increased. CONCLUSIONS Because viral clearance was unimpaired, the study highlights the critical role of serpinB1 in mitigating inflammation and restricting pro-inflammatory cytokine production in influenza infection.
Collapse
Affiliation(s)
- Dapeng Gong
- Immune Disease Institute and Program in Cellular and Molecular Medicine at Children's Hospital and Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | | | | | | | | | | |
Collapse
|
50
|
Brincks EL, Gurung P, Langlois RA, Hemann EA, Legge KL, Griffith TS. The magnitude of the T cell response to a clinically significant dose of influenza virus is regulated by TRAIL. THE JOURNAL OF IMMUNOLOGY 2011; 187:4581-8. [PMID: 21940678 DOI: 10.4049/jimmunol.1002241] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
An immune response of appropriate magnitude should be robust enough to control pathogen spread but not simultaneously lead to immunopathology. Primary infection with influenza A virus (IAV) results in a localized pulmonary infection and inflammation and elicits an IAV-specific CD8 T cell immune response necessary for viral clearance. Clearance of IAV-infected cells, and recovery from infection, is mediated by perforin/granzyme B- and Fas/FasL-mediated mechanisms. We recently reported that TRAIL is another means by which IAV-specific CD8 T cells can kill IAV-infected cells. The current study examined the role of TRAIL in the pulmonary CD8 T cell response to a clinically significant IAV [A/PR/8/34 (PR8; H1N1)] infection (i.e., leads to observable, but limited, morbidity and mortality in wild-type [WT] mice). Compared with WT mice, IAV-infected Trail(-/-) mice experienced increased morbidity and mortality despite similar rates of viral clearance from the lungs. The increased morbidity and mortality in Trail(-/-) mice correlated with increased pulmonary pathology and inflammatory chemokine production. Analysis of lung-infiltrating lymphocytes revealed increased numbers of IAV-specific CD8 T cells in infected Trail(-/-) mice, which correlated with increased pulmonary cytotoxic activity and increased pulmonary expression of MIG and MIP-1α. In addition, there was decreased apoptosis and increased proliferation of IAV-specific CD8 T cells in the lungs of Trail(-/-) mice compared with WT mice. Together, these data suggest that TRAIL regulates the magnitude of the IAV-specific CD8 T cell response during a clinically significant IAV infection to decrease the chance for infection-induced immunopathology.
Collapse
Affiliation(s)
- Erik L Brincks
- Department of Urology, University of Iowa, Iowa City, IA 52242, USA
| | | | | | | | | | | |
Collapse
|