1
|
Torrance BL, Cadar AN, Panier HA, Martin DE, Lorenzo EC, Jellison ER, Bartley JM, Haynes L. Cellular senescence is a double-edged sword in regulating aged immune responses to influenza. Aging Cell 2024; 23:e14162. [PMID: 38689516 PMCID: PMC11258475 DOI: 10.1111/acel.14162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 03/10/2024] [Accepted: 03/14/2024] [Indexed: 05/02/2024] Open
Abstract
Clearance of senescent cells has demonstrated therapeutic potential in the context of chronic age-related diseases. Little is known, however, how clearing senescent cells affects the ability to respond to an acute infection and form quality immunological memory. We aimed to probe the effects of clearing senescent cells in aged mice on the immune response to influenza (flu) infection. We utilized a p16 trimodality reporter mouse model (p16-3MR) to allow for identification and selective clearance of p16-expressing cells upon administration of ganciclovir (GCV). While p16-expressing cells may exacerbate dysfunctional responses to a primary infection, our data suggest they may play a role in fostering memory cell generation. We demonstrate that although clearance of p16-expressing cells enhanced viral clearance, this also severely limited antibody production in the lungs of flu-infected aged mice. 30 days later, there were fewer flu-specific CD8 memory T cells and lower levels of flu-specific antibodies in the lungs of GCV-treated mice. Furthermore, GCV-treated mice were unable to mount an optimal memory response and demonstrated increased viral load following heterosubtypic challenge. These results suggest that targeting senescent cells may potentiate primary responses while limiting the ability to form durable and protective immune memory with age.
Collapse
Affiliation(s)
- Blake L. Torrance
- UConn Center on AgingUniversity of Connecticut School of MedicineFarmingtonConnecticutUSA
- Department of ImmunologyUniversity of Connecticut School of MedicineFarmingtonConnecticutUSA
| | - Andreia N. Cadar
- UConn Center on AgingUniversity of Connecticut School of MedicineFarmingtonConnecticutUSA
- Department of ImmunologyUniversity of Connecticut School of MedicineFarmingtonConnecticutUSA
| | - Hunter A. Panier
- Department of MedicineUniversity of Connecticut School of MedicineFarmingtonConnecticutUSA
| | - Dominique E. Martin
- UConn Center on AgingUniversity of Connecticut School of MedicineFarmingtonConnecticutUSA
- Department of ImmunologyUniversity of Connecticut School of MedicineFarmingtonConnecticutUSA
| | - Erica C. Lorenzo
- UConn Center on AgingUniversity of Connecticut School of MedicineFarmingtonConnecticutUSA
| | - Evan R. Jellison
- Department of ImmunologyUniversity of Connecticut School of MedicineFarmingtonConnecticutUSA
| | - Jenna M. Bartley
- UConn Center on AgingUniversity of Connecticut School of MedicineFarmingtonConnecticutUSA
- Department of ImmunologyUniversity of Connecticut School of MedicineFarmingtonConnecticutUSA
| | - Laura Haynes
- UConn Center on AgingUniversity of Connecticut School of MedicineFarmingtonConnecticutUSA
- Department of ImmunologyUniversity of Connecticut School of MedicineFarmingtonConnecticutUSA
| |
Collapse
|
2
|
Li Y, Yang Y, Chen D, Wang Y, Zhang X, Li W, Chen S, Wong SM, Shen M, Akerley BJ, Shen H. Memory Th17 cell-mediated protection against lethal secondary pneumococcal pneumonia following influenza infection. mBio 2023; 14:e0051923. [PMID: 37222516 PMCID: PMC10470593 DOI: 10.1128/mbio.00519-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 04/11/2023] [Indexed: 05/25/2023] Open
Abstract
Streptococcus pneumoniae (Sp) frequently causes secondary pneumonia after influenza A virus (IAV) infection, leading to high morbidity and mortality worldwide. Concomitant pneumococcal and influenza vaccination improves protection against coinfection but does not always yield complete protection. Impaired innate and adaptive immune responses have been associated with attenuated bacterial clearance in influenza virus-infected hosts. In this study, we showed that preceding low-dose IAV infection caused persistent Sp infection and suppression of bacteria-specific T-helper type 17 (Th17) responses in mice. Prior Sp infection protected against subsequent IAV/Sp coinfection by improving bacterial clearance and rescuing bacteria-specific Th17 responses in the lungs. Furthermore, blockade of IL-17A by anti-IL-17A antibodies abrogated the protective effect of Sp preinfection. Importantly, memory Th17 responses induced by Sp preinfection overcame viral-driven Th17 inhibition and provided cross-protection against different Sp serotypes following coinfection with IAV. These results indicate that bacteria-specific Th17 memory cells play a key role in providing protection against IAV/Sp coinfection in a serotype-independent manner and suggest that a Th17-based vaccine would have excellent potential to mitigate disease caused by coinfection. IMPORTANCE Streptococcus pneumoniae (Sp) frequently causes secondary bacterial pneumonia after influenza A virus (IAV) infection, leading to increased morbidity and mortality worldwide. Current pneumococcal vaccines induce highly strain-specific antibody responses and provide limited protection against IAV/Sp coinfection. Th17 responses are broadly protective against Sp single infection, but whether the Th17 response, which is dramatically impaired by IAV infection in naïve mice, might be effective in immunization-induced protection against pneumonia caused by coinfection is not known. In this study, we have revealed that Sp-specific memory Th17 cells rescue IAV-driven inhibition and provide cross-protection against subsequent lethal coinfection with IAV and different Sp serotypes. These results indicate that a Th17-based vaccine would have excellent potential to mitigate disease caused by IAV/Sp coinfection.
Collapse
Affiliation(s)
- Yong Li
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
- Shanghai Institute of Immunology, Shanghai Jiaotong University, Shanghai, China
| | - Ying Yang
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Dafan Chen
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
- Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan Wang
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Xinyun Zhang
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
| | - Wenchao Li
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Shengsen Chen
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
- Department of Endoscopy, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Cancer and Basic Medicine (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Sandy M. Wong
- Department of Microbiology and Immunology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Mengwen Shen
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
- Department of Emergency Medical, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Brian J. Akerley
- Department of Cell and Molecular Biology, Center for Immunology and Microbial Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Hao Shen
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
3
|
Torrance BL, Cadar AN, Martin DE, Panier HA, Lorenzo EC, Bartley JM, Xu M, Haynes L. Senolytic treatment with dasatinib and quercetin does not improve overall influenza responses in aged mice. FRONTIERS IN AGING 2023; 4:1212750. [PMID: 37396956 PMCID: PMC10313122 DOI: 10.3389/fragi.2023.1212750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 06/09/2023] [Indexed: 07/04/2023]
Abstract
Age is the greatest risk factor for adverse outcomes following influenza (flu) infection. The increased burden of senescent cells with age has been identified as a root cause in many diseases of aging and targeting these cells with drugs termed senolytics has shown promise in alleviating many age-related declines across organ systems. However, there is little known whether targeting these cells will improve age-related deficits in the immune system. Here, we utilized a well characterized senolytic treatment with a combination of dasatinib and quercetin (D + Q) to clear aged (18-20 months) mice of senescent cells prior to a flu infection. We comprehensively profiled immune responses during the primary infection as well as development of immune memory and protection following pathogen reencounter. Senolytic treatment did not improve any aspects of the immune response that were assayed for including: weight loss, viral load, CD8 T-cell infiltration, antibody production, memory T cell development, or recall ability. These results indicate that D + Q may not be an appropriate senolytic to improve aged immune responses to flu infection.
Collapse
Affiliation(s)
- Blake L. Torrance
- University of Connecticut Center on Aging, Farmington, CT, United States
- Department of Immunology, Farmington, CT, United States
| | - Andreia N. Cadar
- University of Connecticut Center on Aging, Farmington, CT, United States
- Department of Immunology, Farmington, CT, United States
| | - Dominique E. Martin
- University of Connecticut Center on Aging, Farmington, CT, United States
- Department of Immunology, Farmington, CT, United States
| | | | - Erica C. Lorenzo
- University of Connecticut Center on Aging, Farmington, CT, United States
| | - Jenna M. Bartley
- University of Connecticut Center on Aging, Farmington, CT, United States
- Department of Immunology, Farmington, CT, United States
| | - Ming Xu
- University of Connecticut Center on Aging, Farmington, CT, United States
- Department of Genetics and Genome Sciences, Farmington, CT, United States
| | - Laura Haynes
- University of Connecticut Center on Aging, Farmington, CT, United States
- Department of Immunology, Farmington, CT, United States
| |
Collapse
|
4
|
Torrance BL, Panier HA, Cadar AN, Martin DE, Lorenzo EC, Jellison ER, Bartley JM, Haynes L. Cellular Senescence is a Double-Edged Sword in Regulating Aged Immune Responses to Influenza. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.10.536027. [PMID: 37090607 PMCID: PMC10120665 DOI: 10.1101/2023.04.10.536027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
Clearance of senescent cells has demonstrated therapeutic potential in the context of chronic age-related diseases. Little is known, however, how clearing senescent cells affects the ability to respond to an acute infection and form quality immunological memory. We aimed to probe the effects of clearing senescent cells in aged mice on the immune response to influenza (flu) infection. We utilized a p16 trimodality reporter mouse model (p16-3MR) to allow for identification and selective deletion of p16-expressing senescent cells upon administration of ganciclovir (GCV). While p16-expressing senescent cells may exacerbate dysfunctional responses to a primary infection, our data suggest they may play a role in fostering memory cell generation. We demonstrate that although deletion of p16-expressing cells enhanced viral clearance, this also severely limited antibody production in the lungs of flu-infected aged mice. 30 days later, there were fewer flu-specific CD8 memory T cells and lower levels of flu-specific antibodies in the lungs of GCV treated mice. GCV treated mice were unable to mount an optimal memory response and demonstrated increased viral load following a heterosubtypic challenge. These results suggest that targeting senescent cells may potentiate primary responses while limiting the ability to form durable and protective immune memory with age.
Collapse
|
5
|
Keilich SR, Cadar AN, Ahern DT, Torrance BL, Lorenzo EC, Martin DE, Haynes L, Bartley JM. Altered T cell infiltration and enrichment of leukocyte regulating pathways within aged skeletal muscle are associated impaired muscle function following influenza infection. GeroScience 2023; 45:1197-1213. [PMID: 36580167 PMCID: PMC9886695 DOI: 10.1007/s11357-022-00715-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 12/09/2022] [Indexed: 12/30/2022] Open
Abstract
Older adults have diminished immune responses that increase susceptibility to infectious diseases, such as influenza (flu). In older adults, flu infection can lead to hospitalization, catastrophic disability, and mortality. We previously demonstrated severe and prolonged muscle degradation and atrophy in aged mice during flu infection. Here, we utilized an unbiased transcriptomic analysis to elucidate mechanisms of flu-induced muscular declines in a mouse model. Our results showed age-related gene expression differences including downregulation of genes associated with muscle regeneration and organization and upregulation of genes associated with pro-inflammatory cytokines and migratory immune pathways in aged mice when compared to young. Pathway analysis revealed significant enrichment of leukocyte migration and T cell activation pathways in the aged muscle during infection. Intramuscular CD4 T cells increased in both young and aged mice during infection, while intramuscular CD8 T cells increased exclusively in aged muscle. CD4 T cells in young muscle were regulatory T cells (Treg), while those in aged were T follicular helper (Tfh) and Th2 cells. Correspondingly, IL-33, an important cytokine for Treg accumulation within tissue, increased only in young flu-infected muscle. Conversely, CXCL10 (IP-10) increased only in aged muscle suggesting a continued recruitment of CD8 T cells into the aged muscle during flu infection. Overall, our findings elucidate a link between flu-induced disability and dysregulated intracellular T cell recruitment into flu-injured muscle with aging. Furthermore, we uncovered potential pathways involved that can be targeted to develop preventative and therapeutic interventions to avert disability and maintain independence following infection.
Collapse
Affiliation(s)
- Spencer R Keilich
- UConn Center On Aging, University of Connecticut School of Medicine, Farmington, CT, 06030, USA
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT, 06030, USA
- Millipore Sigma, 400 Summit Drive, Burlington, MA, 01803, USA
| | - Andreia N Cadar
- UConn Center On Aging, University of Connecticut School of Medicine, Farmington, CT, 06030, USA
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT, 06030, USA
| | - Darcy T Ahern
- Department of Genetics and Genome Sciences, University of Connecticut School of Medicine, Farmington, CT, 06030, USA
- Intellia Therapeutics, 40 Erie St, Cambridge, MA, 02139, USA
| | - Blake L Torrance
- UConn Center On Aging, University of Connecticut School of Medicine, Farmington, CT, 06030, USA
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT, 06030, USA
| | - Erica C Lorenzo
- UConn Center On Aging, University of Connecticut School of Medicine, Farmington, CT, 06030, USA
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT, 06030, USA
| | - Dominique E Martin
- UConn Center On Aging, University of Connecticut School of Medicine, Farmington, CT, 06030, USA
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT, 06030, USA
| | - Laura Haynes
- UConn Center On Aging, University of Connecticut School of Medicine, Farmington, CT, 06030, USA.
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT, 06030, USA.
| | - Jenna M Bartley
- UConn Center On Aging, University of Connecticut School of Medicine, Farmington, CT, 06030, USA.
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT, 06030, USA.
| |
Collapse
|
6
|
Zhang X, Yang Y, Chen S, Li W, Li Y, Akerley BJ, Shao L, Zhang W, Shen H, Abt MC. Antigen-specific memory Th17 cells promote cross-protection against nontypeable Haemophilus influenzae after mild influenza A virus infection. Mucosal Immunol 2023; 16:153-166. [PMID: 36736665 DOI: 10.1016/j.mucimm.2023.01.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 01/22/2023] [Indexed: 02/05/2023]
Abstract
Secondary bacterial pneumonia after influenza A virus (IAV) infection is the leading cause of hospitalization and death associated with IAV infection worldwide. Nontypeable Haemophilus influenzae (NTHi) is one of the most common causes of secondary bacterial pneumonia. Current efforts to develop vaccines against NTHi infection focus on inducing antibodies but are hindered by antigenic diversity among NTHi strains. Therefore, we investigated the contribution of the memory T helper type 17 (Th17) response in protective immunity against IAV/NTHi coinfection. We observed that even a mild IAV infection impaired the NTHi-specific Th17 response and increased morbidity and mortality compared with NTHi monoinfected mice. However, pre-existing memory NTHi-specific Th17 cells induced by a previous NTHi infection overcame IAV-driven Th17 inhibition and were cross-protective against different NTHi strains. Last, mice immunized with a NTHi protein that induced a strong Th17 memory response were broadly protected against diverse NTHi strains after challenge with coinfection. These results indicate that vaccination that limits IAV infection to mild disease may be insufficient to eliminate the risk of a lethal secondary bacterial pneumonia. However, NTHi-specific memory Th17 cells provide serotype-independent protection despite an ongoing IAV infection and demonstrate the advantage of developing broadly protective Th17-inducing vaccines against secondary bacterial pneumonia.
Collapse
Affiliation(s)
- Xinyun Zhang
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China; Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, USA
| | - Ying Yang
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, USA
| | - ShengSen Chen
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China; Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, USA; Department of Endoscopy, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Cancer and Basic Medicine (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Wenchao Li
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, USA; Shanghai Institute of Immunology, Shanghai Jiaotong University, Shanghai, China; Department of Immunology and Rheumatology, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Yong Li
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, USA; Shanghai Institute of Immunology, Shanghai Jiaotong University, Shanghai, China
| | - Brian J Akerley
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Linyun Shao
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
| | - Wenhong Zhang
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China; National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China; Key Laboratory of Medical Molecular Virology (MOE/MOH), Shanghai Medical College, Fudan University, Shanghai, China
| | - Hao Shen
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, USA.
| | - Michael C Abt
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, USA.
| |
Collapse
|
7
|
Doan TA, Forward T, Tamburini BAJ. Trafficking and retention of protein antigens across systems and immune cell types. Cell Mol Life Sci 2022; 79:275. [PMID: 35505125 PMCID: PMC9063628 DOI: 10.1007/s00018-022-04303-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 04/01/2022] [Accepted: 04/12/2022] [Indexed: 12/05/2022]
Abstract
In response to infection or vaccination, the immune system initially responds non-specifically to the foreign insult (innate) and then develops a specific response to the foreign antigen (adaptive). The programming of the immune response is shaped by the dispersal and delivery of antigens. The antigen size, innate immune activation and location of the insult all determine how antigens are handled. In this review we outline which specific cell types are required for antigen trafficking, which processes require active compared to passive transport, the ability of specific cell types to retain antigens and the viruses (human immunodeficiency virus, influenza and Sendai virus, vesicular stomatitis virus, vaccinia virus) and pattern recognition receptor activation that can initiate antigen retention. Both where the protein antigen is localized and how long it remains are critically important in shaping protective immune responses. Therefore, understanding antigen trafficking and retention is necessary to understand the type and magnitude of the immune response and essential for the development of novel vaccine and therapeutic targets.
Collapse
Affiliation(s)
- Thu A Doan
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Colorado School of Medicine, Aurora, USA.,Immunology Graduate Program, University of Colorado School of Medicine, Aurora, USA
| | - Tadg Forward
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Colorado School of Medicine, Aurora, USA
| | - Beth A Jirón Tamburini
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Colorado School of Medicine, Aurora, USA. .,Immunology Graduate Program, University of Colorado School of Medicine, Aurora, USA. .,Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, USA.
| |
Collapse
|
8
|
Walsh SM, Sheridan RM, Lucas ED, Doan TA, Ware BC, Schafer J, Fu R, Burchill MA, Hesselberth JR, Tamburini BAJ. Molecular tracking devices quantify antigen distribution and archiving in the murine lymph node. eLife 2021; 10:e62781. [PMID: 33843587 PMCID: PMC8116055 DOI: 10.7554/elife.62781] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 04/09/2021] [Indexed: 01/02/2023] Open
Abstract
The detection of foreign antigens in vivo has relied on fluorescent conjugation or indirect read-outs such as antigen presentation. In our studies, we found that these widely used techniques had several technical limitations that have precluded a complete picture of antigen trafficking or retention across lymph node cell types. To address these limitations, we developed a 'molecular tracking device' to follow the distribution, acquisition, and retention of antigen in the lymph node. Utilizing an antigen conjugated to a nuclease-resistant DNA tag, acting as a combined antigen-adjuvant conjugate, and single-cell mRNA sequencing, we quantified antigen abundance in the lymph node. Variable antigen levels enabled the identification of caveolar endocytosis as a mechanism of antigen acquisition or retention in lymphatic endothelial cells. Thus, these molecular tracking devices enable new approaches to study dynamic tissue dissemination of antigen-adjuvant conjugates and identify new mechanisms of antigen acquisition and retention at cellular resolution in vivo.
Collapse
Affiliation(s)
- Shannon M Walsh
- Department of Biochemistry and Molecular Genetics, University of Colorado School of MedicineAuroraUnited States
| | - Ryan M Sheridan
- RNA Bioscience Initiative, University of Colorado School of MedicineAuroraUnited States
| | - Erin D Lucas
- Immunology Graduate Program, University of Colorado School of MedicineAuroraUnited States
- Department of Immunology and Microbiology, University of Colorado School of MedicineAuroraUnited States
| | - Thu A Doan
- Immunology Graduate Program, University of Colorado School of MedicineAuroraUnited States
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Colorado School of MedicineAuroraUnited States
| | - Brian C Ware
- Immunology Graduate Program, University of Colorado School of MedicineAuroraUnited States
- Department of Immunology and Microbiology, University of Colorado School of MedicineAuroraUnited States
| | - Johnathon Schafer
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Colorado School of MedicineAuroraUnited States
| | - Rui Fu
- RNA Bioscience Initiative, University of Colorado School of MedicineAuroraUnited States
| | - Matthew A Burchill
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Colorado School of MedicineAuroraUnited States
| | - Jay R Hesselberth
- Department of Biochemistry and Molecular Genetics, University of Colorado School of MedicineAuroraUnited States
- RNA Bioscience Initiative, University of Colorado School of MedicineAuroraUnited States
| | - Beth Ann Jiron Tamburini
- Immunology Graduate Program, University of Colorado School of MedicineAuroraUnited States
- Department of Immunology and Microbiology, University of Colorado School of MedicineAuroraUnited States
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Colorado School of MedicineAuroraUnited States
| |
Collapse
|
9
|
Hutchinson JA, Kronenberg K, Riquelme P, Wenzel JJ, Glehr G, Schilling HL, Zeman F, Evert K, Schmiedel M, Mickler M, Drexler K, Bitterer F, Cordero L, Beyer L, Bach C, Koestler J, Burkhardt R, Schlitt HJ, Hellwig D, Werner JM, Spang R, Schmidt B, Geissler EK, Haferkamp S. Virus-specific memory T cell responses unmasked by immune checkpoint blockade cause hepatitis. Nat Commun 2021; 12:1439. [PMID: 33664251 PMCID: PMC7933278 DOI: 10.1038/s41467-021-21572-y] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 01/29/2021] [Indexed: 02/08/2023] Open
Abstract
Treatment of advanced melanoma with combined PD-1/CTLA-4 blockade commonly causes serious immune-mediated complications. Here, we identify a subset of patients predisposed to immune checkpoint blockade-related hepatitis who are distinguished by chronic expansion of effector memory CD4+ T cells (TEM cells). Pre-therapy CD4+ TEM cell expansion occurs primarily during autumn or winter in patients with metastatic disease and high cytomegalovirus (CMV)-specific serum antibody titres. These clinical features implicate metastasis-dependent, compartmentalised CMV reactivation as the cause of CD4+ TEM expansion. Pre-therapy CD4+ TEM expansion predicts hepatitis in CMV-seropositive patients, opening possibilities for avoidance or prevention. 3 of 4 patients with pre-treatment CD4+ TEM expansion who received αPD-1 monotherapy instead of αPD-1/αCTLA-4 therapy remained hepatitis-free. 4 of 4 patients with baseline CD4+ TEM expansion given prophylactic valganciclovir and αPD-1/αCTLA-4 therapy remained hepatitis-free. Our findings exemplify how pathogen exposure can shape clinical reactions after cancer therapy and how this insight leads to therapeutic innovations.
Collapse
Affiliation(s)
- James A. Hutchinson
- grid.411941.80000 0000 9194 7179Department of Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Katharina Kronenberg
- grid.411941.80000 0000 9194 7179Department of Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Paloma Riquelme
- grid.411941.80000 0000 9194 7179Department of Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Jürgen J. Wenzel
- grid.411941.80000 0000 9194 7179Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg, Regensburg, Germany
| | - Gunther Glehr
- grid.7727.50000 0001 2190 5763Institute of Functional Genomics and Statistical Bioinformatics, University of Regensburg, Regensburg, Germany
| | - Hannah-Lou Schilling
- grid.411941.80000 0000 9194 7179Department of Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Florian Zeman
- grid.411941.80000 0000 9194 7179Center for Clinical Studies, University Hospital Regensburg, Regensburg, Germany
| | - Katja Evert
- grid.411941.80000 0000 9194 7179Institute of Pathology, University Hospital Regensburg, Regensburg, Germany
| | - Martin Schmiedel
- grid.411941.80000 0000 9194 7179Department of Nuclear Medicine, University Hospital Regensburg, Regensburg, Germany
| | - Marion Mickler
- grid.411941.80000 0000 9194 7179Department of Dermatology, University Hospital Regensburg, Regensburg, Germany
| | - Konstantin Drexler
- grid.411941.80000 0000 9194 7179Department of Dermatology, University Hospital Regensburg, Regensburg, Germany
| | - Florian Bitterer
- grid.411941.80000 0000 9194 7179Department of Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Laura Cordero
- grid.411941.80000 0000 9194 7179Department of Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Lukas Beyer
- grid.411941.80000 0000 9194 7179Institute of Radiology, University Hospital Regensburg, Regensburg, Germany
| | - Christian Bach
- grid.411668.c0000 0000 9935 6525Department of Medicine V, University Hospital Erlangen, Erlangen, Germany
| | - Josef Koestler
- grid.411941.80000 0000 9194 7179Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg, Regensburg, Germany
| | - Ralph Burkhardt
- grid.411941.80000 0000 9194 7179Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Regensburg, Germany
| | - Hans J. Schlitt
- grid.411941.80000 0000 9194 7179Department of Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Dirk Hellwig
- grid.411941.80000 0000 9194 7179Department of Nuclear Medicine, University Hospital Regensburg, Regensburg, Germany
| | - Jens M. Werner
- grid.411941.80000 0000 9194 7179Department of Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Rainer Spang
- grid.7727.50000 0001 2190 5763Institute of Functional Genomics and Statistical Bioinformatics, University of Regensburg, Regensburg, Germany
| | - Barbara Schmidt
- grid.411941.80000 0000 9194 7179Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg, Regensburg, Germany
| | - Edward K. Geissler
- grid.411941.80000 0000 9194 7179Department of Surgery, University Hospital Regensburg, Regensburg, Germany ,Personalised Tumour Therapy, Fraunhofer Institute for Experimental Medicine and Toxicology, Regensburg, Germany
| | - Sebastian Haferkamp
- grid.411941.80000 0000 9194 7179Department of Dermatology, University Hospital Regensburg, Regensburg, Germany
| |
Collapse
|
10
|
LRCH1 deficiency enhances LAT signalosome formation and CD8 + T cell responses against tumors and pathogens. Proc Natl Acad Sci U S A 2020; 117:19388-19398. [PMID: 32727906 DOI: 10.1073/pnas.2000970117] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
CD8+ T cells play pivotal roles in eradicating pathogens and tumor cells. T cell receptor (TCR) signaling is vital for the optimal activation of CD8+ T cells. Upon TCR engagement, the transmembrane adapter protein LAT (linker for activation of T cells) recruits other key signaling molecules and forms the "LAT signalosome" for downstream signal transduction. However, little is known about which functional partners could restrain the formation of the LAT signalosome and inhibit CD8+ cytotoxic T lymphocyte (CTL)-mediated cytotoxicity. Here we have demonstrated that LRCH1 (leucine-rich repeats and calponin homology domain containing 1) directly binds LAT, reduces LAT phosphorylation and interaction with GRB2, and also promotes the endocytosis of LAT. Lrch1 -/- mice display better protection against influenza virus and Listeria infection, with enhanced CD8+ T cell proliferation and cytotoxicity. Adoptive transfer of Lrch1 -/- CD8+ CTLs leads to increased B16-MO5 tumor clearance in vivo. Furthermore, knockout of LRCH1 in human chimeric antigen receptor (CAR) T cells that recognize the liver tumor-associated antigen glypican-3 could improve CAR T cell migration and proliferation in vitro. These findings suggest LRCH1 as a potential translational target to improve T cell immunotherapy against infection and tumors.
Collapse
|
11
|
Keilich SR, Lorenzo EC, Torrance BL, Harrison AG, Bartley JM, Haynes L. Vaccination mitigates influenza-induced muscular declines in aged mice. GeroScience 2020; 42:1593-1608. [PMID: 32472355 DOI: 10.1007/s11357-020-00206-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 05/19/2020] [Indexed: 10/24/2022] Open
Abstract
Influenza (flu) infection increases the risk for disability, falls, and broken bones in older adults. We have employed a preclinical model to examine the impact of flu on muscle function, which has a direct impact on fall risk. In mice, flu causes mobility and strength impairments with induction of inflammatory and muscle degradation genes that are increased and prolonged with aging. To determine if vaccination could reduce flu-induced muscle decrements, mice were vaccinated with flu nucleoprotein, infected, and muscle parameters were measured. Vaccination of aged mice resulted in significant protection from functional decrements, muscle gene expressions alterations, and morphological damage. Vaccination also improved protection from lung localized and systemic inflammation in aged mice. Despite documented decreased vaccine efficacy with aging, vaccination still provided partial protection to aged mice and represents a potential strategy to prevent flu-induced disability. These findings provide translational insight on ways to reduce flu-induced disability with aging. Graphical abstract .
Collapse
Affiliation(s)
- Spencer R Keilich
- UConn Center on Aging, University of Connecticut School of Medicine, 263 Farmington Avenue, Farmington, CT, 06030, USA.,Department of Immunology, University of Connecticut School of Medicine, 263 Farmington Avenue, Farmington, CT, 06030, USA
| | - Erica C Lorenzo
- UConn Center on Aging, University of Connecticut School of Medicine, 263 Farmington Avenue, Farmington, CT, 06030, USA.,Department of Immunology, University of Connecticut School of Medicine, 263 Farmington Avenue, Farmington, CT, 06030, USA
| | - Blake L Torrance
- UConn Center on Aging, University of Connecticut School of Medicine, 263 Farmington Avenue, Farmington, CT, 06030, USA.,Department of Immunology, University of Connecticut School of Medicine, 263 Farmington Avenue, Farmington, CT, 06030, USA
| | - Andrew G Harrison
- Department of Immunology, University of Connecticut School of Medicine, 263 Farmington Avenue, Farmington, CT, 06030, USA
| | - Jenna M Bartley
- UConn Center on Aging, University of Connecticut School of Medicine, 263 Farmington Avenue, Farmington, CT, 06030, USA.,Department of Immunology, University of Connecticut School of Medicine, 263 Farmington Avenue, Farmington, CT, 06030, USA
| | - Laura Haynes
- UConn Center on Aging, University of Connecticut School of Medicine, 263 Farmington Avenue, Farmington, CT, 06030, USA. .,Department of Immunology, University of Connecticut School of Medicine, 263 Farmington Avenue, Farmington, CT, 06030, USA.
| |
Collapse
|
12
|
Lauron EJ, Yang L, Harvey IB, Sojka DK, Williams GD, Paley MA, Bern MD, Park E, Victorino F, Boon ACM, Yokoyama WM. Viral MHCI inhibition evades tissue-resident memory T cell formation and responses. J Exp Med 2019; 216:117-132. [PMID: 30559127 PMCID: PMC6314518 DOI: 10.1084/jem.20181077] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 09/14/2018] [Accepted: 11/07/2018] [Indexed: 01/06/2023] Open
Abstract
Tissue-resident memory CD8+ T cells (TRMs) confer rapid protection and immunity against viral infections. Many viruses have evolved mechanisms to inhibit MHCI presentation in order to evade CD8+ T cells, suggesting that these mechanisms may also apply to TRM-mediated protection. However, the effects of viral MHCI inhibition on the function and generation of TRMs is unclear. Herein, we demonstrate that viral MHCI inhibition reduces the abundance of CD4+ and CD8+ TRMs, but its effects on the local microenvironment compensate to promote antigen-specific CD8+ TRM formation. Unexpectedly, local cognate antigen enhances CD8+ TRM development even in the context of viral MHCI inhibition and CD8+ T cell evasion, strongly suggesting a role for in situ cross-presentation in local antigen-driven TRM differentiation. However, local cognate antigen is not required for CD8+ TRM maintenance. We also show that viral MHCI inhibition efficiently evades CD8+ TRM effector functions. These findings indicate that viral evasion of MHCI antigen presentation has consequences on the development and response of antiviral TRMs.
Collapse
Affiliation(s)
- Elvin J Lauron
- Division of Rheumatology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Liping Yang
- Division of Rheumatology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Ian B Harvey
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
| | - Dorothy K Sojka
- Division of Rheumatology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Graham D Williams
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO
| | - Michael A Paley
- Division of Rheumatology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Michael D Bern
- Division of Rheumatology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Eugene Park
- Division of Rheumatology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Francisco Victorino
- Division of Rheumatology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Adrianus C M Boon
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO
| | - Wayne M Yokoyama
- Division of Rheumatology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
13
|
Rojas-Quintero J, Wang X, Tipper J, Burkett PR, Zuñiga J, Ashtekar AR, Polverino F, Rout A, Yambayev I, Hernández C, Jimenez L, Ramírez G, Harrod KS, Owen CA. Matrix metalloproteinase-9 deficiency protects mice from severe influenza A viral infection. JCI Insight 2018; 3:99022. [PMID: 30568032 DOI: 10.1172/jci.insight.99022] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 11/06/2018] [Indexed: 02/06/2023] Open
Abstract
Matrix metalloproteinase-9 (MMP-9) cleaves various proteins to regulate inflammatory and injury responses. However, MMP-9's activities during influenza A viral (IAV) infections are incompletely understood. Herein, plasma MMP-9 levels were increased in patients with pandemic H1N1 and seasonal IAV infections. MMP-9 lung levels were increased and localized to airway epithelial cells and leukocytes in H1N1-infected WT murine lungs. H1N1-infected Mmp-9-/- mice had lower mortality rates, reduced weight loss, lower lung viral titers, and reduced lung injury, along with lower E-cadherin shedding in bronchoalveolar lavage fluid (BALF) samples than WT mice. H1N1-infected Mmp-9-/- mice had an altered immune response to IAV with lower BALF PMN and macrophage counts, higher Th1-like CD4+ and CD8+ T cell subsets, lower T regulatory cell counts, reduced lung type I interferon levels, and higher lung interferon-γ levels. Mmp-9 bone marrow-chimera studies revealed that Mmp-9 deficiency in lung parenchymal cells protected mice from IAV-induced mortality. H1N1-infected Mmp-9-/- lung epithelial cells had lower viral titers than H1N1-infected WT cells in vitro. Thus, H1N1-infected Mmp-9-/- mice are protected from IAV-induced lung disease due to a more effective adaptive immune response to IAV and reduced epithelial barrier injury due partly to reduced E-cadherin shedding. Thus, we believe that MMP-9 is a novel therapeutic target for IAV infections.
Collapse
Affiliation(s)
- Joselyn Rojas-Quintero
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, and Harvard Medical School, Boston, Massachusetts, USA
| | - Xiaoyun Wang
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, and Harvard Medical School, Boston, Massachusetts, USA
| | - Jennifer Tipper
- Department of Anesthesiology and Perioperative Medicine, Division of Molecular and Translational Biomedicine, School of Medicine, University of Alabama-Birmingham, Birmingham, Alabama, USA
| | - Patrick R Burkett
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, and Harvard Medical School, Boston, Massachusetts, USA
| | - Joaquin Zuñiga
- Laboratory of Immunobiology and Genetics, and Intensive Care Unit, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
| | - Amit R Ashtekar
- Department of Anesthesiology and Perioperative Medicine, Division of Molecular and Translational Biomedicine, School of Medicine, University of Alabama-Birmingham, Birmingham, Alabama, USA
| | - Francesca Polverino
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, and Harvard Medical School, Boston, Massachusetts, USA.,Lovelace Respiratory Research Institute, Albuquerque, New Mexico, USA
| | - Amit Rout
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, and Harvard Medical School, Boston, Massachusetts, USA
| | - Ilyas Yambayev
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, and Harvard Medical School, Boston, Massachusetts, USA
| | - Carmen Hernández
- Laboratory of Immunobiology and Genetics, and Intensive Care Unit, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico.,Escuela de Medicina y Ciencias de la Salud, Tecnologico de Monterrey, Mexico City, Mexico
| | - Luis Jimenez
- Laboratory of Immunobiology and Genetics, and Intensive Care Unit, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
| | - Gustavo Ramírez
- Laboratory of Immunobiology and Genetics, and Intensive Care Unit, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
| | - Kevin S Harrod
- Department of Anesthesiology and Perioperative Medicine, Division of Molecular and Translational Biomedicine, School of Medicine, University of Alabama-Birmingham, Birmingham, Alabama, USA
| | - Caroline A Owen
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, and Harvard Medical School, Boston, Massachusetts, USA.,Lovelace Respiratory Research Institute, Albuquerque, New Mexico, USA
| |
Collapse
|
14
|
Ather JL, Dienz O, Boyson JE, Anathy V, Amiel E, Poynter ME. Serum Amyloid A3 is required for normal lung development and survival following influenza infection. Sci Rep 2018; 8:16571. [PMID: 30410021 PMCID: PMC6224415 DOI: 10.1038/s41598-018-34901-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 10/24/2018] [Indexed: 02/06/2023] Open
Abstract
Serum amyloid A (SAA) proteins are a family of acute phase apolipoproteins implicated to directly modulate innate and adaptive immune responses. However, new studies comparing endogenous SAAs and recombinant forms of these proteins have questioned the function of SAA in inflammation and immunity. We generated SAA3 knockout mice to evaluate the contribution of SAA3 to lung development and immune-mediated lung disease. While SAA3 deficiency does not affect the generation of house dust mite-induced allergic asthma, mice lacking SAA3 develop adult-onset obesity, intrinsic airway hyperresponsiveness, increased inflammatory and fibrotic gene expression in the lung, and elevated levels of lung citrullinated proteins. Polyclonally stimulated CD4+ T cells from SAA3-/- mice exhibit impaired glycolytic activity, decreased TH2 and TH1 cytokine secretion, and elevated IL-17A production compared to wild type cells. Polyclonally stimulated CD8+ T cells from SAA3-/- mice also exhibit impaired glycolytic activity as well as a diminished capacity to produce IL-2 and IFNγ. Finally, SAA3-/- mice demonstrate increased mortality in response to H1N1 influenza infection, along with higher copy number of viral RNAs in the lung, a lack of CD8+ T cell IFNγ secretion, and decreased flu-specific antibodies. Our findings indicate that endogenous SAA3 regulates lung development and homeostasis, and is required for protection against H1N1 influenza infection.
Collapse
Affiliation(s)
- Jennifer L Ather
- Vermont Lung Center, Division of Pulmonary Disease and Critical Care, Department of Medicine, University of Vermont, Burlington, VT, 05405, USA
| | - Oliver Dienz
- Department of Surgery, University of Vermont, Burlington, VT, 05405, USA
| | - Jonathan E Boyson
- Department of Surgery, University of Vermont, Burlington, VT, 05405, USA
| | - Vikas Anathy
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, 05405, USA
| | - Eyal Amiel
- Department of Biomedical and Health Sciences, University of Vermont, Burlington, VT, 05405, USA
| | - Matthew E Poynter
- Vermont Lung Center, Division of Pulmonary Disease and Critical Care, Department of Medicine, University of Vermont, Burlington, VT, 05405, USA.
| |
Collapse
|
15
|
Keeler SP, Agapov EV, Hinojosa ME, Letvin AN, Wu K, Holtzman MJ. Influenza A Virus Infection Causes Chronic Lung Disease Linked to Sites of Active Viral RNA Remnants. THE JOURNAL OF IMMUNOLOGY 2018; 201:2354-2368. [PMID: 30209189 DOI: 10.4049/jimmunol.1800671] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 08/14/2018] [Indexed: 12/18/2022]
Abstract
Clinical and experimental observations suggest that chronic lung disease is linked to respiratory viral infection. However, the long-term aspect of this relationship is not yet defined using a virus that replicates at properly high levels in humans and a corresponding animal model. In this study, we show that influenza A virus infection achieves 1 × 106-fold increases in viral load in the lung and dose-dependent severity of acute illness in mice. Moreover, these events are followed by persistence of negative- and positive-strand viral RNA remnants for 15 wk and chronic lung disease for at least 26 wk postinfection. The disease is manifested by focal areas of bronchiolization and mucus production that contain increased levels of viral RNA remnants along with mucin Muc5ac and Il13 mRNA compared with uninvolved areas of the lung. Excess mucus production and associated airway hyperreactivity (but not fibrosis or emphysema) are partially attenuated with loss of IL-13 production or signaling (using mice with IL-13 or STAT6 deficiency). These deficiencies cause reciprocal increases in l17a mRNA and neutrophils in the lung; however, none of these disease endpoints are changed with IL-13/IL-17a compared with IL-13 deficiency or STAT6/IL-17a compared with STAT6 deficiency. The results establish the capacity of a potent human respiratory virus to produce chronic lung disease focally at sites of active viral RNA remnants, likely reflecting locations of viral replication that reprogram the region. Viral dose dependency of disease also implicates high-level viral replication and severity of acute infection as determinants of chronic lung diseases such as asthma and COPD with IL-13-dependent and IL-13/IL-17-independent mechanisms.
Collapse
Affiliation(s)
- Shamus P Keeler
- Pulmonary and Critical Care Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Eugene V Agapov
- Pulmonary and Critical Care Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Michael E Hinojosa
- Pulmonary and Critical Care Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Adam N Letvin
- Pulmonary and Critical Care Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Kangyun Wu
- Pulmonary and Critical Care Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Michael J Holtzman
- Pulmonary and Critical Care Medicine, Washington University School of Medicine, St. Louis, MO 63110
| |
Collapse
|
16
|
Takamura S. Niches for the Long-Term Maintenance of Tissue-Resident Memory T Cells. Front Immunol 2018; 9:1214. [PMID: 29904388 PMCID: PMC5990602 DOI: 10.3389/fimmu.2018.01214] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 05/15/2018] [Indexed: 12/13/2022] Open
Abstract
Tissue-resident memory T cells (TRM cells) are a population of immune cells that reside in the lymphoid and non-lymphoid organs without recirculation through the blood. These important cells occupy and utilize unique anatomical and physiological niches that are distinct from those for other memory T cell populations, such as central memory T cells in the secondary lymphoid organs and effector memory T cells that circulate through the tissues. CD8+ TRM cells typically localize in the epithelial layers of barrier tissues where they are optimally positioned to act as sentinels to trigger antigen-specific protection against reinfection. CD4+ TRM cells typically localize below the epithelial layers, such as below the basement membrane, and cluster in lymphoid structures designed to optimize interactions with antigen-presenting cells upon reinfection. A key feature of TRM populations is their ability to be maintained in barrier tissues for prolonged periods of time. For example, skin CD8+ TRM cells displace epidermal niches originally occupied by γδ T cells, thereby enabling their stable persistence for years. It is also clear that the long-term maintenance of TRM cells in different microenvironments is dependent on multiple tissue-specific survival cues, although the specific details are poorly understood. However, not all TRM persist over the long term. Recently, we identified a new spatial niche for the maintenance of CD8+ TRM cells in the lung, which is created at the site of tissue regeneration after injury [termed repair-associated memory depots (RAMD)]. The short-lived nature of RAMD potentially explains the short lifespans of CD8+ TRM cells in this particular tissue. Clearly, a better understanding of the niche-dependent maintenance of TRM cells will be important for the development of vaccines designed to promote barrier immunity. In this review, we discuss recent advances in our understanding of the properties and nature of tissue-specific niches that maintain TRM cells in different tissues.
Collapse
Affiliation(s)
- Shiki Takamura
- Department of Immunology, Faculty of Medicine, Kindai University, Osaka, Japan
| |
Collapse
|
17
|
Hargrove MM, Snow SJ, Luebke RW, Wood CE, Krug JD, Krantz QT, King C, Copeland CB, McCullough SD, Gowdy KM, Kodavanti UP, Gilmour MI, Gavett SH. Effects of Simulated Smog Atmospheres in Rodent Models of Metabolic and Immunologic Dysfunction. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2018; 52:3062-3070. [PMID: 29384667 PMCID: PMC6233996 DOI: 10.1021/acs.est.7b06534] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
Air pollution is a diverse and dynamic mixture of gaseous and particulate matter, limiting our understanding of associated adverse health outcomes. The biological effects of two simulated smog atmospheres (SA) with different compositions but similar air quality health indexes were compared in a nonobese diabetic rat model (Goto-Kakizaki, GK) and three mouse immune models (house dust mite (HDM) allergy, antibody response to heat-killed pneumococcus, and resistance to influenza A infection). In GK rats, both SA-PM (high particulate matter) and SA-O3 (high ozone) decreased cholesterol levels immediately after a 4-h exposure, whereas only SA-O3 increased airflow limitation. Airway responsiveness to methacholine was increased in HDM-allergic mice compared with nonallergic mice, but exposure to SA-PM or SA-O3 did not significantly alter responsiveness. Exposure to SA-PM did not affect the IgM response to pneumococcus, and SA-O3 did not affect virus titers, although inflammatory cytokine levels were decreased in mice infected at the end of a 7-day exposure. Collectively, acute SA exposures produced limited health effects in animal models of metabolic and immune diseases. Effects of SA-O3 tended to be greater than those of SA-PM, suggesting that gas-phase components in photochemically derived multipollutant mixtures may be of greater concern than secondary organic aerosol PM.
Collapse
Affiliation(s)
- Marie McGee Hargrove
- Oak Ridge Institute for Science and Education, Research Triangle Park, NC 27709, USA
| | - Samantha J. Snow
- Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, NC 27711, USA
| | - Robert W. Luebke
- Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, NC 27711, USA
| | - Charles E. Wood
- Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, NC 27711, USA
| | - Jonathan D. Krug
- Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, NC 27711, USA
| | - Q. Todd Krantz
- Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, NC 27711, USA
| | - Charly King
- Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, NC 27711, USA
| | - Carey B. Copeland
- Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, NC 27711, USA
| | - Shaun D. McCullough
- Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, NC 27711, USA
| | - Kymberly M. Gowdy
- Department of Pharmacology and Toxicology, East Carolina University, Greenville, NC 27834, USA
| | - Urmila P. Kodavanti
- Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, NC 27711, USA
| | - M. Ian Gilmour
- Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, NC 27711, USA
| | - Stephen H. Gavett
- Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, NC 27711, USA
| |
Collapse
|
18
|
Bartley JM, Zhou X, Kuchel GA, Weinstock GM, Haynes L. Impact of Age, Caloric Restriction, and Influenza Infection on Mouse Gut Microbiome: An Exploratory Study of the Role of Age-Related Microbiome Changes on Influenza Responses. Front Immunol 2017; 8:1164. [PMID: 28979265 PMCID: PMC5611400 DOI: 10.3389/fimmu.2017.01164] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 09/01/2017] [Indexed: 12/17/2022] Open
Abstract
Immunosenescence refers to age-related declines in the capacity to respond to infections such as influenza (flu). Caloric restriction represents a known strategy to slow many aging processes, including those involving the immune system. More recently, some changes in the microbiome have been described with aging, while the gut microbiome appears to influence responses to flu vaccination and infection. With these considerations in mind, we used a well-established mouse model of flu infection to explore the impact of flu infection, aging, and caloric restriction on the gut microbiome. Young, middle-aged, and aged caloric restricted (CR) and ad lib fed (AL) mice were examined after a sublethal flu infection. All mice lost 10–20% body weight and, as expected for these early time points, losses were similar at different ages and between diet groups. Cytokine and chemokine levels were also similar with the notable exception of IL-1α, which rose more than fivefold in aged AL mouse serum, while it remained unchanged in aged CR serum. Fecal microbiome phyla abundance profiles were similar in young, middle-aged, and aged AL mice at baseline and at 4 days post flu infection, while increases in Proteobacteria were evident at 7 days post flu infection in all three age groups. CR mice, compared to AL mice in each age group, had increased abundance of Proteobacteria and Verrucomicrobia at all time points. Interestingly, principal coordinate analysis determined that diet exerts a greater effect on the microbiome than age or flu infection. Percentage body weight loss correlated with the relative abundance of Proteobacteria regardless of age, suggesting flu pathogenicity is related to Proteobacteria abundance. Further, several microbial Operational Taxonomic Units from the Bacteroidetes phyla correlated with serum chemokine/cytokines regardless of both diet and age suggesting an interplay between flu-induced systemic inflammation and gut microbiota. These exploratory studies highlight the impact of caloric restriction on fecal microbiome in both young and aged animals, as well as the many complex relationships between flu responses and gut microbiota. Thus, these preliminary studies provide the necessary groundwork to examine how gut microbiota alterations may be leveraged to influence declining immune responses with aging.
Collapse
Affiliation(s)
- Jenna M Bartley
- UConn Center on Aging, Farmington, CT, United States.,Department of Immunology, UConn Health, Farmington, CT, United States
| | - Xin Zhou
- Jackson Laboratory for Genomic Medicine, Farmington, CT, United States.,Department of Genetics and Genome Sciences, UConn Health, Farmington, CT, United States
| | - George A Kuchel
- UConn Center on Aging, Farmington, CT, United States.,Department of Genetics and Genome Sciences, UConn Health, Farmington, CT, United States
| | - George M Weinstock
- Jackson Laboratory for Genomic Medicine, Farmington, CT, United States.,Department of Genetics and Genome Sciences, UConn Health, Farmington, CT, United States
| | - Laura Haynes
- UConn Center on Aging, Farmington, CT, United States.,Department of Immunology, UConn Health, Farmington, CT, United States
| |
Collapse
|
19
|
Davis SM, Sweet LM, Oppenheimer KH, Suratt BT, Phillippe M. Estradiol and progesterone influence on influenza infection and immune response in a mouse model. Am J Reprod Immunol 2017; 78. [PMID: 28557314 DOI: 10.1111/aji.12695] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Accepted: 03/29/2017] [Indexed: 01/07/2023] Open
Abstract
PROBLEM Influenza infection severity may be mediated by estradiol and/or progesterone. METHOD OF STUDY An exploratory study was designed to evaluate 17-β-estradiol and progesterone on influenza infection and examine immune-mediated response in a mouse model. Inoculation with placebo or mouse-adapted H1N1 influenza virus occurred. Treatment groups included 17-β-estradiol, progesterone, ovariectomy, and pregnancy. Mice were assessed for morbidity and mortality. Toll-like receptor gene studies and airspace cell differentials were performed. RESULTS Onset of morbidity was earlier and morbidity duration greater for progesterone. Absence of morbidity/mortality and overall survival was greater for 17-β-estradiol. Airspace cell differentials suggest improved immune cell recruitment for 17-β-estradiol. Pregnant mouse data demonstrate significant mortality during the period of increased progesterone. Select immune cell markers demonstrate patterns of regulation that may promote proper immune response to influenza infection for 17-β-estradiol. CONCLUSION Estradiol may play a protective and progesterone a detrimental role in the pathophysiology of influenza infection.
Collapse
Affiliation(s)
- Sarah M Davis
- Department of Obstetrics and Gynecology, The Alpert Medical School of Brown University, Women & Infants Hospital of Rhode Island, Providence, RI, USA
| | - Leigh M Sweet
- University of Vermont College of Medicine, Burlington, VT, USA
| | | | - Benjamin T Suratt
- Department of Medicine, University of Vermont College of Medicine, Burlington, VT, USA
| | - Mark Phillippe
- Department of Obstetrics & Gynecology, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
20
|
Takamura S. Persistence in Temporary Lung Niches: A Survival Strategy of Lung-Resident Memory CD8 + T Cells. Viral Immunol 2017; 30:438-450. [PMID: 28418771 PMCID: PMC5512299 DOI: 10.1089/vim.2017.0016] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Respiratory virus infections, such as those mediated by influenza virus, parainfluenza virus, respiratory syncytial virus (RSV), severe acute respiratory syndrome coronavirus (SARS-CoV), rhinovirus, and adenovirus, are responsible for substantial morbidity and mortality, especially in children and older adults. Furthermore, the potential emergence of highly pathogenic strains of influenza virus poses a significant public health threat. Thus, the development of vaccines capable of eliciting long-lasting protective immunity to those pathogens is a major public health priority. CD8+ Tissue-resident memory T (TRM) cells are a newly defined population that resides permanently in the nonlymphoid tissues including the lung. These cells are capable of providing local protection immediately after infection, thereby promoting rapid host recovery. Recent studies have offered new insights into the anatomical niches that harbor lung CD8+ TRM cells, and also identified the requirement and limitations of TRM maintenance. However, it remains controversial whether lung CD8+ TRM cells are continuously replenished by new cells from the circulation or permanently lodged in this site. A better understanding of how lung CD8+ TRM cells are generated and maintained and the tissue-specific factors that drive local TRM formation is required for optimal vaccine development. This review focuses on recent advance in our understanding of CD8+ TRM cell establishment and maintenance in the lung, and describes how those processes are uniquely regulated in this tissue.
Collapse
Affiliation(s)
- Shiki Takamura
- Department of Immunology, Kindai University , Faculty of Medicine, Osaka, Japan
| |
Collapse
|
21
|
Genetic variation in chromosome Y regulates susceptibility to influenza A virus infection. Proc Natl Acad Sci U S A 2017; 114:3491-3496. [PMID: 28242695 DOI: 10.1073/pnas.1620889114] [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: 01/05/2023] Open
Abstract
Males of many species, ranging from humans to insects, are more susceptible than females to parasitic, fungal, bacterial, and viral infections. One mechanism that has been proposed to account for this difference is the immunocompetence handicap model, which posits that the greater infectious disease burden in males is due to testosterone, which drives the development of secondary male sex characteristics at the expense of suppressing immunity. However, emerging data suggest that cell-intrinsic (chromosome X and Y) sex-specific factors also may contribute to the sex differences in infectious disease burden. Using a murine model of influenza A virus (IAV) infection and a panel of chromosome Y (ChrY) consomic strains on the C57BL/6J background, we present data showing that genetic variation in ChrY influences IAV pathogenesis in males. Specific ChrY variants increase susceptibility to IAV in males and augment pathogenic immune responses in the lung, including activation of proinflammatory IL-17-producing γδ T cells, without affecting viral replication. In addition, susceptibility to IAV segregates independent of copy number variation in multicopy ChrY gene families that influence susceptibility to other immunopathological phenotypes, including survival after infection with coxsackievirus B3. These results demonstrate a critical role for genetic variation in ChrY in regulating susceptibility to infectious disease.
Collapse
|
22
|
Takamura S, Yagi H, Hakata Y, Motozono C, McMaster SR, Masumoto T, Fujisawa M, Chikaishi T, Komeda J, Itoh J, Umemura M, Kyusai A, Tomura M, Nakayama T, Woodland DL, Kohlmeier JE, Miyazawa M. Specific niches for lung-resident memory CD8+ T cells at the site of tissue regeneration enable CD69-independent maintenance. J Exp Med 2016; 213:3057-3073. [PMID: 27815325 PMCID: PMC5154946 DOI: 10.1084/jem.20160938] [Citation(s) in RCA: 168] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 08/29/2016] [Accepted: 10/11/2016] [Indexed: 11/05/2022] Open
Abstract
Takamura et al. show that most lung CD8+ TRM cells are not maintained in the inducible bronchus-associated lymphoid tissue (iBALT) but are maintained in specific niches created at the site of tissue regeneration, which are termed as repair-associated memory depots (RAMDs). CD8+ tissue-resident memory T cells (TRM cells) reside permanently in nonlymphoid tissues and provide a first line of protection against invading pathogens. However, the precise localization of CD8+ TRM cells in the lung, which physiologically consists of a markedly scant interstitium compared with other mucosa, remains unclear. In this study, we show that lung CD8+ TRM cells localize predominantly in specific niches created at the site of regeneration after tissue injury, whereas peripheral tissue-circulating CD8+ effector memory T cells (TEM cells) are widely but sparsely distributed in unaffected areas. Although CD69 inhibited sphingosine 1–phosphate receptor 1–mediated egress of CD8+ T cells immediately after their recruitment into lung tissues, such inhibition was not required for the retention of cells in the TRM niches. Furthermore, despite rigid segregation of TEM cells from the TRM niche, prime-pull strategy with cognate antigen enabled the conversion from TEM cells to TRM cells by creating de novo TRM niches. Such damage site–specific localization of CD8+ TRM cells may be important for efficient protection against secondary infections by respiratory pathogens.
Collapse
Affiliation(s)
- Shiki Takamura
- Department of Immunology, Faculty of Medicine, Kindai University, Osaka-Sayama, Osaka 589-8511, Japan
| | - Hideki Yagi
- Cell Biology Laboratory, Department of Pharmaceutical Sciences, Faculty of Pharmacy, Kindai University, Osaka-Sayama, Osaka 589-8511, Japan
| | - Yoshiyuki Hakata
- Department of Immunology, Faculty of Medicine, Kindai University, Osaka-Sayama, Osaka 589-8511, Japan
| | - Chihiro Motozono
- Department of Immunology, Faculty of Medicine, Kindai University, Osaka-Sayama, Osaka 589-8511, Japan
| | - Sean R McMaster
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA 30322
| | - Tomoko Masumoto
- Department of Immunology, Faculty of Medicine, Kindai University, Osaka-Sayama, Osaka 589-8511, Japan
| | - Makoto Fujisawa
- Department of Immunology, Faculty of Medicine, Kindai University, Osaka-Sayama, Osaka 589-8511, Japan
| | - Tomomi Chikaishi
- Department of Immunology, Faculty of Medicine, Kindai University, Osaka-Sayama, Osaka 589-8511, Japan
| | - Junko Komeda
- Cell Biology Laboratory, Department of Pharmaceutical Sciences, Faculty of Pharmacy, Kindai University, Osaka-Sayama, Osaka 589-8511, Japan
| | - Jun Itoh
- Cell Biology Laboratory, Department of Pharmaceutical Sciences, Faculty of Pharmacy, Kindai University, Osaka-Sayama, Osaka 589-8511, Japan
| | - Miki Umemura
- Cell Biology Laboratory, Department of Pharmaceutical Sciences, Faculty of Pharmacy, Kindai University, Osaka-Sayama, Osaka 589-8511, Japan
| | - Ami Kyusai
- Cell Biology Laboratory, Department of Pharmaceutical Sciences, Faculty of Pharmacy, Kindai University, Osaka-Sayama, Osaka 589-8511, Japan
| | - Michio Tomura
- Laboratory of Immunology, Faculty of Pharmacy, Osaka Otani University, Tondabayashi, Osaka 584-8540, Japan
| | - Toshinori Nakayama
- Department of Immunology, Graduate School of Medicine, Chiba University, Inage, Chiba 263-8522, Japan
| | - David L Woodland
- Keystone Symposia on Molecular and Cellular Biology, Silverthorne, CO 80498
| | - Jacob E Kohlmeier
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA 30322
| | - Masaaki Miyazawa
- Department of Immunology, Faculty of Medicine, Kindai University, Osaka-Sayama, Osaka 589-8511, Japan.,Anti-Aging Center, Kindai University, Osaka-Sayama, Osaka 589-8511, Japan
| |
Collapse
|
23
|
Lefebvre JS, Lorenzo EC, Masters AR, Hopkins JW, Eaton SM, Smiley ST, Haynes L. Vaccine efficacy and T helper cell differentiation change with aging. Oncotarget 2016; 7:33581-94. [PMID: 27177221 PMCID: PMC5085104 DOI: 10.18632/oncotarget.9254] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Accepted: 04/27/2016] [Indexed: 12/22/2022] Open
Abstract
Influenza and pneumonia are leading causes of death in elderly populations. With age, there is an increased inflammatory response and slower viral clearance during influenza infection which increases the risk of extended illness and mortality. Here we employ a preclinical murine model of influenza infection to examine the protective capacity of vaccination with influenza nucleoprotein (NP). While NP vaccination reduces influenza-induced lung inflammation in young mice, aged mice do not show this reduction, but are protected from influenza-induced mortality. Aged mice do make a significant amount of NP-specific IgG and adoptive transfer experiments show that NP antibody can protect from death but cannot reduce lung inflammation. Furthermore, young but not aged vaccinated mice generate significant numbers of NP-specific T cells following subsequent infection and few of these T cells are found in aged lungs early during infection. Importantly, aged CD4 T cells have a propensity to differentiate towards a T follicular helper (Tfh) phenotype rather than a T helper 1 (Th1) phenotype that predominates in the young. Since Th1 cells are important in viral clearance, reduced Th1 differentiation in the aged is critical and could account for some or all of the age-related differences in vaccine responses and infection resolution.
Collapse
Affiliation(s)
- Julie S. Lefebvre
- The Trudeau Institute, Saranac Lake, NY, United States of America
- Département de Pneumologie, Centre de Recherche de l'Institut Universitaire de Cardiologie et Pneumologie de Québec, Québec, QC, Canada
| | - Erica C. Lorenzo
- Department of Immunology and Center on Aging, University of Connecticut Health Center, Farmington, CT, United States of America
| | - April R. Masters
- Department of Immunology and Center on Aging, University of Connecticut Health Center, Farmington, CT, United States of America
| | - Jacob W. Hopkins
- Department of Immunology and Center on Aging, University of Connecticut Health Center, Farmington, CT, United States of America
| | - Sheri M. Eaton
- The Trudeau Institute, Saranac Lake, NY, United States of America
| | - Stephen T. Smiley
- The Trudeau Institute, Saranac Lake, NY, United States of America
- NIAID/NIH, Bethesda, MD, USA
| | - Laura Haynes
- Department of Immunology and Center on Aging, University of Connecticut Health Center, Farmington, CT, United States of America
| |
Collapse
|
24
|
Champagne DP, Hatle KM, Fortner KA, D'Alessandro A, Thornton TM, Yang R, Torralba D, Tomás-Cortázar J, Jun YW, Ahn KH, Hansen KC, Haynes L, Anguita J, Rincon M. Fine-Tuning of CD8(+) T Cell Mitochondrial Metabolism by the Respiratory Chain Repressor MCJ Dictates Protection to Influenza Virus. Immunity 2016; 44:1299-311. [PMID: 27234056 DOI: 10.1016/j.immuni.2016.02.018] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Revised: 02/18/2016] [Accepted: 02/18/2016] [Indexed: 12/27/2022]
Abstract
Mitochondrial respiration is regulated in CD8(+) T cells during the transition from naive to effector and memory cells, but mechanisms controlling this process have not been defined. Here we show that MCJ (methylation-controlled J protein) acted as an endogenous break for mitochondrial respiration in CD8(+) T cells by interfering with the formation of electron transport chain respiratory supercomplexes. Metabolic profiling revealed enhanced mitochondrial metabolism in MCJ-deficient CD8(+) T cells. Increased oxidative phosphorylation and subcellular ATP accumulation caused by MCJ deficiency selectively increased the secretion, but not expression, of interferon-γ. MCJ also adapted effector CD8(+) T cell metabolism during the contraction phase. Consequently, memory CD8(+) T cells lacking MCJ provided superior protection against influenza virus infection. Thus, MCJ offers a mechanism for fine-tuning CD8(+) T cell mitochondrial metabolism as an alternative to modulating mitochondrial mass, an energetically expensive process. MCJ could be a therapeutic target to enhance CD8(+) T cell responses.
Collapse
Affiliation(s)
- Devin P Champagne
- Program in Immunobiology, Department of Medicine, University of Vermont, Burlington, Vermont, 05405 USA
| | - Ketki M Hatle
- Program in Immunobiology, Department of Medicine, University of Vermont, Burlington, Vermont, 05405 USA
| | - Karen A Fortner
- Program in Immunobiology, Department of Medicine, University of Vermont, Burlington, Vermont, 05405 USA
| | - Angelo D'Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, CO 80045, USA
| | - Tina M Thornton
- Program in Immunobiology, Department of Medicine, University of Vermont, Burlington, Vermont, 05405 USA
| | - Rui Yang
- Program in Immunobiology, Department of Medicine, University of Vermont, Burlington, Vermont, 05405 USA
| | - Daniel Torralba
- Program in Immunobiology, Department of Medicine, University of Vermont, Burlington, Vermont, 05405 USA
| | - Julen Tomás-Cortázar
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Derio 48160 Bizkaia, Spain
| | - Yong Woong Jun
- Department of Chemistry, Center for Electro-Photo Behaviors in Advanced Molecular Systems, Pohang University of Science and Technology (POSTECH), Nam-Gu, Pohang, 790-784 Gyeongbuk, Republic of Korea
| | - Kyo Han Ahn
- Department of Chemistry, Center for Electro-Photo Behaviors in Advanced Molecular Systems, Pohang University of Science and Technology (POSTECH), Nam-Gu, Pohang, 790-784 Gyeongbuk, Republic of Korea
| | - Kirk C Hansen
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, CO 80045, USA
| | - Laura Haynes
- Center on Aging and Department of Immunology, University of Connecticut Health Center, Farmington, CT 06030 USA
| | - Juan Anguita
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Derio 48160 Bizkaia, Spain; Ikerbasque, Basque Foundation for Science, Bilbao, Bizkaia, Spain
| | - Mercedes Rincon
- Program in Immunobiology, Department of Medicine, University of Vermont, Burlington, Vermont, 05405 USA.
| |
Collapse
|
25
|
Duan S, Thomas PG. Balancing Immune Protection and Immune Pathology by CD8(+) T-Cell Responses to Influenza Infection. Front Immunol 2016; 7:25. [PMID: 26904022 PMCID: PMC4742794 DOI: 10.3389/fimmu.2016.00025] [Citation(s) in RCA: 108] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 01/18/2016] [Indexed: 02/05/2023] Open
Abstract
Influenza A virus (IAV) is a significant human pathogen causing annual epidemics and periodic pandemics. CD8+ cytotoxic T lymphocyte (CTL)-mediated immunity contributes to the clearance of virus-infected cells, and CTL immunity targeting the conserved internal proteins of IAVs is a key protection mechanism when neutralizing antibodies are absent during heterosubtypic IAV infection. However, CTL infiltration into the airways, its cytotoxicity, and the effects of produced proinflammatory cytokines can cause severe lung tissue injury, thereby contributing to immunopathology. Studies have discovered complicated and exquisite stimulatory and inhibitory mechanisms that regulate CTL magnitude and effector activities during IAV infection. Here, we review the state of knowledge on the roles of IAV-specific CTLs in immune protection and immunopathology during IAV infection in animal models, highlighting the key findings of various requirements and constraints regulating the balance of immune protection and pathology involved in CTL immunity. We also discuss the evidence of cross-reactive CTL immunity as a positive correlate of cross-subtype protection during secondary IAV infection in both animal and human studies. We argue that the effects of CTL immunity on protection and immunopathology depend on multiple layers of host and viral factors, including complex host mechanisms to regulate CTL magnitude and effector activity, the pathogenic nature of the IAV, the innate response milieu, and the host historical immune context of influenza infection. Future efforts are needed to further understand these key host and viral factors, especially to differentiate those that constrain optimally effective CTL antiviral immunity from those necessary to restrain CTL-mediated non-specific immunopathology in the various contexts of IAV infection, in order to develop better vaccination and therapeutic strategies for modifying protective CTL immunity.
Collapse
Affiliation(s)
- Susu Duan
- Department of Immunology, St. Jude Children's Research Hospital , Memphis, TN , USA
| | - Paul G Thomas
- Department of Immunology, St. Jude Children's Research Hospital , Memphis, TN , USA
| |
Collapse
|
26
|
Kedl RM, Tamburini BA. Antigen archiving by lymph node stroma: A novel function for the lymphatic endothelium. Eur J Immunol 2015; 45:2721-9. [PMID: 26278423 DOI: 10.1002/eji.201545739] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Revised: 06/01/2015] [Accepted: 08/12/2015] [Indexed: 12/22/2022]
Abstract
Secondary lymphoid stroma performs far more functions than simple structural support for lymphoid tissues, providing a host of soluble and membrane-bound cues to trafficking leukocytes during inflammation and homeostasis. More recently it has become clear that stromal cells can manipulate T-cell responses, either through direct antigen-mediated stimulation of T cells or more indirectly through the retention and management of antigen after viral infection or vaccination. In light of recent data, this review provides an overview of stromal cell subsets and functions during the progression of an adaptive immune response with particular emphasis on antigen capture and retention by follicular dendritic cells as well as the recently described "antigen archiving" function of lymphatic endothelial cells (LECs). Given its impact on the maintenance of protective immune memory, we conclude by discussing the most pressing questions pertaining to LEC antigen capture, archiving and exchange with hematopoetically derived antigen-presenting cells.
Collapse
Affiliation(s)
- Ross M Kedl
- Department of Immunology and Microbiology, University of Colorado Denver, School of Medicine, Aurora, CO, USA
| | - Beth A Tamburini
- Department of Immunology and Microbiology, University of Colorado Denver, School of Medicine, Aurora, CO, USA
| |
Collapse
|
27
|
Antigen capture and archiving by lymphatic endothelial cells following vaccination or viral infection. Nat Commun 2014; 5:3989. [PMID: 24905362 PMCID: PMC4073648 DOI: 10.1038/ncomms4989] [Citation(s) in RCA: 107] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Accepted: 04/29/2014] [Indexed: 01/13/2023] Open
Abstract
Antigen derived from viral infections with influenza and Vesicular Stomatitis Virus (VSV) can persist after resolution of infection. Here we show that antigen can similarly persist for weeks following viral challenge and vaccination. Antigen is captured by Lymphatic Endothelial Cells (LECs), under conditions that induce LEC proliferation. Consistent with published data showing that viral antigen persistence impacts on the function of circulating memory T cells, we find that vaccine elicited antigen persistence, found on LECs, positively influences the degree of protective immunity elicited by circulating memory CD8+ T cells. The coupling of LEC proliferation and antigen capture identifies a mechanism by which the LECs store, or “archive”, antigens for extended periods of time after antigen challenge, thereby increasing IFNγ/IL-2 production and enhancing protection against infection. These findings therefore have the potential to impact future vaccination strategies and our understanding of the role for persisting antigen in both vaccine and infectious settings.
Collapse
|
28
|
Lv J, Wang D, Hua YH, Pei SJ, Wang J, Hu WW, Wang XL, Jia N, Jiang QS. Pulmonary immune responses to 2009 pandemic influenza A (H1N1) virus in mice. BMC Infect Dis 2014; 14:197. [PMID: 24725777 PMCID: PMC4002205 DOI: 10.1186/1471-2334-14-197] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Accepted: 04/07/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Well-characterized mice models will afford a cheaper, easy-handling opportunity for a more comprehensive understanding of 2009 influenza A (H1N1) virus's pathogenesis potential. We aimed to provide a robust description of pulmonary immune responses in the mice infected by the virus. METHODS BALB/c mice were inoculated intranasally with A/Beijing/501/2009(H1N1) (BJ501) and A/PR/8/34(H1N1) (PR8) viruses and compared for survival rate, viral replication, and kinetics of pulmonary immune responses. RESULTS BJ501 virus replicated less efficiently in the lungs than PR8, and both caused lethal illness in the mice. The transient increases of pulmonary TNF-α 2 days post infection for BJ501 and of INF-γ and IL-10 at 6 days post infection for PR8 were observed. IL-2+ and IL-4+ secreting cells showed significant increase 12 days post infection, while IFN-γ+, IgG+ and IgA+ secreting cells increased 6 days post infection. The different patterns of pulmonary immunological parameters between two viruses were at most seen in IL-6, IL-17 secretion and IgG1/IgG2a ratio. CONCLUSIONS The BALB/c mouse is evaluated as a good pathogenic model for studying BJ501 2009 H1N1 virus. The work provided some basic and detailed data, which might be referred when further evaluating innate and adapted pulmonary immune responses and local viral load in mice.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Xi-Liang Wang
- The Second Artillery General Hospital, PLA, Beijing 100088, China.
| | | | | |
Collapse
|
29
|
Lv J, Hua Y, Wang D, Liu A, An J, Li A, Wang Y, Wang X, Jia N, Jiang Q. Kinetics of pulmonary immune cells, antibody responses and their correlations with the viral clearance of influenza A fatal infection in mice. Virol J 2014; 11:57. [PMID: 24666970 PMCID: PMC3986938 DOI: 10.1186/1743-422x-11-57] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2013] [Accepted: 03/13/2014] [Indexed: 12/03/2022] Open
Abstract
Fatal influenza A virus infection is a major threat to public health throughout the world. Lung macrophages and neutrophils have critical roles for both the pathogenesis and viral clearance of fatal viral infections. These are complicated by the interaction of innate immunity and adaptive immunity against viral infection. In this study, we investigated the overall kinetics of lung macrophages, neutrophils, CD4+T cells, CD8+T cells, CD38+ cells, and CD138+ cells, the levels of antibody and cytokine responses, both in the early and late phases of fatal infection with A/PR/8/34 (H1N1) virus in mice. The changes in lung viral load were also evaluated. We found that pulmonary macrophages and neutrophils both accumulated in the early and late phases of fatal infections and they positively correlated with the lung and serum antibody titers, and negatively correlated with the viral load locally. The secretion of IL-6 might relate to high numbers of macrophages and neutrophils in the early infection. The work implies that pulmonary macrophages, neutrophils and the antibody response all have an essential role in virus elimination of fatal influenza A viral infection. These findings may have implications for the development of prophylactic and therapeutic strategies in fatal influenza A viral infection. Further evaluation of the cooperation among macrophages, neutrophils and antibody responses in eliminating the virus with fatal infection is needed.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Xiliang Wang
- The Second Artillery General Hospital, PLA, 16 Xinjiekouwai Street, Xicheng District Beijing 100088, China.
| | | | | |
Collapse
|
30
|
Workman AM, Jacobs AK, Vogel AJ, Condon S, Brown DM. Inflammation enhances IL-2 driven differentiation of cytolytic CD4 T cells. PLoS One 2014; 9:e89010. [PMID: 24586481 PMCID: PMC3930678 DOI: 10.1371/journal.pone.0089010] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Accepted: 01/14/2014] [Indexed: 12/24/2022] Open
Abstract
Cytolytic CD4 T cells (CD4 CTL) have been identified in vivo in response to viral infections; however, the factors necessary for driving the cytolytic phenotype have not been fully elucidated. Our previously published work suggests IL-2 may be the master regulator of perforin-mediated cytotoxicity in CD4 effectors. To further dissect the role of IL-2 in CD4 CTL generation, T cell receptor transgenic mice deficient in the ability to produce IL-2 or the high affinity IL-2 receptor (IL-2Rα, CD25) were used. Increasing concentrations of IL-2 were necessary to drive perforin (Prf) expression and maximal cytotoxicity. Granzyme B (GrB) expression and killing correlated with STAT5 activation and CD25 expression in vitro, suggesting that signaling through the high affinity IL-2R is critical for full cytotoxicity. IL-2 signaling was also necessary in vivo for inducing the Th1 phenotype and IFN-γ expression in CD4 T cells during influenza A (IAV) infection. In addition, GrB expression, as measured by mean fluorescent intensity, was decreased in CD25 deficient cells; however, the frequency of CD4 cells expressing GrB was unchanged. Similarly, analysis of cytolytic markers such as CD107a/b and Eomesodermin indicate high IL-2Rα expression is not necessary to drive the CD4 CTL phenotype during IAV infection. Thus, inflammatory signals induced by viral infection may overcome the need for strong IL-2 signals in driving cytotoxicity in CD4 cells.
Collapse
Affiliation(s)
- Aspen M Workman
- School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, Nebraska, United States of America ; Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, Nebraska, United States of America
| | - Ashley K Jacobs
- Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, Nebraska, United States of America
| | - Alexander J Vogel
- School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, Nebraska, United States of America
| | - Shirley Condon
- School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, Nebraska, United States of America ; Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, Nebraska, United States of America
| | - Deborah M Brown
- School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, Nebraska, United States of America ; Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, Nebraska, United States of America
| |
Collapse
|
31
|
Olson MR, McDermott DS, Varga SM. The initial draining lymph node primes the bulk of the CD8 T cell response and influences memory T cell trafficking after a systemic viral infection. PLoS Pathog 2012; 8:e1003054. [PMID: 23236277 PMCID: PMC3516554 DOI: 10.1371/journal.ppat.1003054] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2011] [Accepted: 10/10/2012] [Indexed: 11/18/2022] Open
Abstract
Lymphocytic choriomeningitis virus (LCMV) causes a systemic infection in mice with virus replication occurring in both peripheral tissues and secondary lymphoid organs. Because of the rapid systemic dissemination of the virus, the secondary lymphoid organs responsible for the induction of the LCMV-specific CD8 T cell response are poorly defined. We show that the mediastinal lymph node (MedLN) serves as the primary draining lymph node following LCMV infection. In addition, we demonstrate that the MedLN is responsible for priming the majority of the virus-specific CD8 T cell response. Following resolution of the acute infection, the draining MedLN exhibits characteristics of a reactive lymph node including an increased presence of germinal center B cells and increased cellularity for up to 60 days post-infection. Furthermore, the reactive MedLN harbors an increased frequency of CD62L(-) effector memory CD8 T cells as compared to the non-draining lymph nodes. The accumulation of LCMV-specific CD62L(-) memory CD8 T cells in the MedLN is independent of residual antigen and is not a unique feature of the MedLN as footpad infection with LCMV leads to a similar increase of virus-specific CD62L(-) effector memory CD8 T cells in the draining popliteal lymph node. Our results indicate that CD62L(-) effector memory CD8 T cells are granted preferential access into the draining lymph nodes for an extended time following resolution of an infection.
Collapse
Affiliation(s)
- Matthew R. Olson
- Department of Microbiology, University of Iowa, Iowa City, Iowa, United States of America
| | - Daniel S. McDermott
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, Iowa, United States of America
| | - Steven M. Varga
- Department of Microbiology, University of Iowa, Iowa City, Iowa, United States of America
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, Iowa, United States of America
- Department of Pathology, University of Iowa, Iowa City, Iowa, United States of America
- * E-mail:
| |
Collapse
|
32
|
Haynes L, Szaba FM, Eaton SM, Kummer LW, Lanthier PA, Petell AH, Duso DK, Luo D, Lin JS, Lefebvre JS, Randall TD, Johnson LL, Kohlmeier JE, Woodland DL, Smiley ST. Immunity to the conserved influenza nucleoprotein reduces susceptibility to secondary bacterial infections. THE JOURNAL OF IMMUNOLOGY 2012; 189:4921-9. [PMID: 23028058 DOI: 10.4049/jimmunol.1201916] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Influenza causes >250,000 deaths annually in the industrialized world, and bacterial infections frequently cause secondary illnesses during influenza outbreaks, including pneumonia, bronchitis, sinusitis, and otitis media. In this study, we demonstrate that cross-reactive immunity to mismatched influenza strains can reduce susceptibility to secondary bacterial infections, even though this fails to prevent influenza infection. Specifically, infecting mice with H3N2 influenza before challenging with mismatched H1N1 influenza reduces susceptibility to either Gram-positive Streptococcus pneumoniae or Gram-negative Klebsiella pneumoniae. Vaccinating mice with the highly conserved nucleoprotein of influenza also reduces H1N1-induced susceptibility to lethal bacterial infections. Both T cells and Abs contribute to defense against influenza-induced bacterial diseases; influenza cross-reactive T cells reduce viral titers, whereas Abs to nucleoprotein suppress induction of inflammation in the lung. These findings suggest that nonneutralizing influenza vaccines that fail to prevent influenza infection may nevertheless protect the public from secondary bacterial diseases when neutralizing vaccines are not available.
Collapse
|
33
|
Essential role of IL-6 in protection against H1N1 influenza virus by promoting neutrophil survival in the lung. Mucosal Immunol 2012; 5:258-66. [PMID: 22294047 PMCID: PMC3328598 DOI: 10.1038/mi.2012.2] [Citation(s) in RCA: 214] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Influenza virus infection is considered a major worldwide public health problem. Seasonal infections with the most common influenza virus strains (e.g., H1N1) can usually be resolved, but they still cause a high rate of mortality. The factors that influence the outcome of the infection remain unclear. Here, we show that deficiency of interleukin (IL)-6 or IL-6 receptor is sufficient for normally sublethal doses of H1N1 influenza A virus to cause death in mice. IL-6 is necessary for resolution of influenza infection by protecting neutrophils from virus-induced death in the lung and by promoting neutrophil-mediated viral clearance. Loss of IL-6 results in persistence of the influenza virus in the lung leading to pronounced lung damage and, ultimately, death. Thus, we demonstrate that IL-6 is a vital innate immune cytokine in providing protection against influenza A infection. Genetic or environmental factors that impair IL-6 production or signaling could increase mortality to influenza virus infection.
Collapse
|
34
|
Bassett JD, Swift SL, Bramson JL. Optimizing vaccine-induced CD8(+) T-cell immunity: focus on recombinant adenovirus vectors. Expert Rev Vaccines 2012; 10:1307-19. [PMID: 21919620 DOI: 10.1586/erv.11.88] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Recombinant adenoviruses have emerged as promising viral vectors for CD8(+) T-cell vaccines. Our studies have indicated that unlike most acute infections, the CD8(+) T-cell memory population elicited by recombinant human adenovirus serotype 5 (rHuAd5) displays a dominant effector memory phenotype. Persistent, low-level transgene expression from the rHuAd5 vector sustains the CD8(+) T-cell memory population and a nonhematopoietic cell compartment appears to be involved in long-term presentation of adenoviral antigens. Although we are beginning to learn more about the factors that control the maintenance and functionality of memory CD8(+) T cells, we do not yet fully understand what comprises a protective CD8(+) T-cell response. Results from upcoming Phase II clinical trials will be important for determining whether rHuAd5 T-cell vaccines are effective in humans and should help identify correlates of CD8(+) T-cell protection.
Collapse
Affiliation(s)
- Jennifer D Bassett
- Centre for Gene Therapeutics, Department of Pathology and Molecular Medicine, McMaster University, Room MDCL-5071, 1200 Main Street West, Hamilton, Ontario, L8N 3Z5, Canada
| | | | | |
Collapse
|
35
|
McHeyzer-Williams M, Okitsu S, Wang N, McHeyzer-Williams L. Molecular programming of B cell memory. Nat Rev Immunol 2011; 12:24-34. [PMID: 22158414 DOI: 10.1038/nri3128] [Citation(s) in RCA: 326] [Impact Index Per Article: 25.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The development of high-affinity B cell memory is regulated through three separable phases, each involving antigen recognition by specific B cells and cognate T helper cells. Initially, antigen-primed B cells require cognate T cell help to gain entry into the germinal centre pathway to memory. Once in the germinal centre, B cells with variant B cell receptors must access antigens and present them to germinal centre T helper cells to enter long-lived memory B cell compartments. Following antigen recall, memory B cells require T cell help to proliferate and differentiate into plasma cells. A recent surge of information - resulting from dynamic B cell imaging in vivo and the elucidation of T follicular helper cell programmes - has reshaped the conceptual landscape surrounding the generation of memory B cells. In this Review, we integrate this new information about each phase of antigen-specific B cell development to describe the newly unravelled molecular dynamics of memory B cell programming.
Collapse
Affiliation(s)
- Michael McHeyzer-Williams
- Department of Immunology and Microbial Sciences, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, USA.
| | | | | | | |
Collapse
|
36
|
Live attenuated influenza vaccine (LAIV) impacts innate and adaptive immune responses. Vaccine 2011; 29:7849-56. [PMID: 21816194 DOI: 10.1016/j.vaccine.2011.07.093] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2011] [Revised: 07/19/2011] [Accepted: 07/20/2011] [Indexed: 11/22/2022]
Abstract
Influenza A infection induces a massive inflammatory response in the lungs that leads to significant illness and increases the susceptibility to secondary bacterial pneumonia. The most efficient way to prevent influenza infection is through vaccination. While inactivated vaccines induce protective levels of serum antibodies to influenza hemaglutinin (HA) and neuraminidase (NA) surface proteins, these are strain specific and offer little protection against heterosubtypic influenza viruses. In contrast, live attenuated influenza vaccines (LAIVs) induce a T cell response in addition to antibody responses against HA and NA surface proteins. Importantly, LAIV vaccination induces a response in a mouse model that protects against illness due to heterosubtypic influenza strains. While it is not completely clear what is the mechanism of action of LAIV heterosubtypic protection in humans, it has been shown that LAIV induces heterosubtypic protection in mice that is dependent upon a Type 1 immune response and requires CD8 T cells. In this study, we show that LAIV-induced immunity leads to significantly reduced viral titers and inflammatory responses in the lungs of mice following heterosubtypic infection. Not only are viral titers reduced in LAIV vaccinated mice, the amounts of inflammatory cytokines and chemokines in lung tissue are significantly lower. Additionally, we show that LAIV vaccination of healthy adults also induces a robust Type 1 memory response including the production of chemokines and cytokines involved in T cell activation and recruitment. Thus, our results indicate that LAIV vaccination functions by inducing immune memory which can act to modulate the immune response to subsequent heterosubtypic challenge by influencing both innate and adaptive responses.
Collapse
|
37
|
Ortiz-Stern A, Kanda A, Mionnet C, Cazareth J, Lazzari A, Fleury S, Dombrowicz D, Glaichenhaus N, Julia V. Langerin+ dendritic cells are responsible for LPS-induced reactivation of allergen-specific Th2 responses in postasthmatic mice. Mucosal Immunol 2011; 4:343-53. [PMID: 21048704 DOI: 10.1038/mi.2010.73] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Allergic asthma is a T cell-dependent inflammatory lung disease that results from complex interactions between genetic predisposition and environmental factors, including exposure to lipopolysaccharide (LPS). In this study, we have shown that airway LPS exposure was sufficient to induce airway hyperreactivity (AHR) and eosinophil recruitment in mice that had previously experienced an acute episode of allergic asthma. LPS-induced disease reactivation depended on the activation of allergen-specific CD4(+) T cells by a subset of lung langerin(+) dendritic cells (DCs) that retained the allergen. Upon LPS exposure, migration of langerin(+) DCs from lungs to draining lymph nodes increased and LPS-exposed langerin(+) DCs instructed CD4(+) T cells toward a T helper (Th) 2 response. Selective depletion of langerin(+) DCs prevented LPS-induced eosinophil recruitment and T-cell activation, further demonstrating a critical role for langerin(+) DCs in disease reactivation. This finding provides a possible explanation for the subclinical worsening of asthmatics following exposure to low-dose LPS.
Collapse
Affiliation(s)
- A Ortiz-Stern
- Université de Nice-Sophia Antipolis, Valbonne, France
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Fousteri G, Dave A, Juedes A, Juntti T, Morin B, Togher L, Farber DL, von Herrath M. Increased memory conversion of naïve CD8 T cells activated during late phases of acute virus infection due to decreased cumulative antigen exposure. PLoS One 2011; 6:e14502. [PMID: 21253594 PMCID: PMC3017078 DOI: 10.1371/journal.pone.0014502] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2010] [Accepted: 12/16/2010] [Indexed: 11/28/2022] Open
Abstract
Background Memory CD8 T cells form an essential part of protective immunity against viral infections. Antigenic load, costimulation, CD4-help, cytokines and chemokines fluctuate during the course of an antiviral immune response thus affecting CD8 T cell activation and memory conversion. Methodology/Principal Findings In the present study, naïve TCR transgenic LCMV-specific P14 CD8 T cells engaged at a late stage during the acute antiviral LCMV response showed reduced expansion kinetics but greater memory conversion in the spleen. Such late activated cells displayed a memory precursor effector phenotype already at the peak of the systemic antiviral response, suggesting that the environment determined their fate during antigen encounter. In the spleen, the majority of late transferred cells exhibited a central memory phenotype compared to the effector memory displayed by the early transferred cells. Increasing the inflammatory response by exogenous administration of IFNγ, PolyI:C or CpG did not affect memory conversion in the late transferred group, suggesting that the diverging antigen load early versus later during acute infection had determined their fate. In agreement, reduction in the LCMV antigenic load after ribavirin treatment enhanced the contribution of early transferred cells to the long lasting memory pool. Conclusions/Significance Our results show that naïve CD8 cells, exposed to reduced duration or concentration of antigen during viral infection convert into memory more efficiently, an observation that could have significant implications for vaccine design.
Collapse
Affiliation(s)
- Georgia Fousteri
- Diabetes Center, La Jolla Institute for Allergy and Immunology, La Jolla, California, United States of America
| | - Amy Dave
- Diabetes Center, La Jolla Institute for Allergy and Immunology, La Jolla, California, United States of America
| | - Amy Juedes
- Diabetes Center, La Jolla Institute for Allergy and Immunology, La Jolla, California, United States of America
| | - Therese Juntti
- Diabetes Center, La Jolla Institute for Allergy and Immunology, La Jolla, California, United States of America
| | - Bret Morin
- Diabetes Center, La Jolla Institute for Allergy and Immunology, La Jolla, California, United States of America
| | - Lisa Togher
- Diabetes Center, La Jolla Institute for Allergy and Immunology, La Jolla, California, United States of America
| | - Donna L. Farber
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, New York, United States of America
| | - Matthias von Herrath
- Diabetes Center, La Jolla Institute for Allergy and Immunology, La Jolla, California, United States of America
- * E-mail:
| |
Collapse
|
39
|
McCloskey ML, Curotto de Lafaille MA, Carroll MC, Erlebacher A. Acquisition and presentation of follicular dendritic cell-bound antigen by lymph node-resident dendritic cells. ACTA ACUST UNITED AC 2010; 208:135-48. [PMID: 21173103 PMCID: PMC3023135 DOI: 10.1084/jem.20100354] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Follicular dendritic cells (DCs [FDCs]) are prominent stromal cell constituents of B cell follicles with the remarkable ability to retain complement-fixed antigens on their cell surface for extended periods of time. These retained immune complexes have long been known to provide the antigenic stimulus that drives antibody affinity maturation, but their role in cellular immunity has remained unclear. In this study, we show that FDC-retained antigens are continually sampled by lymph node-resident DCs for presentation to CD8 T cells. This novel pathway of antigen acquisition was detectable when FDCs were loaded with purified antigens bound into classical antigen-antibody immune complexes, as well as after pregnancy, when they are loaded physiologically with antigens associated with the complement-fixed microparticles released from the placenta into maternal blood. In both cases, ensuing antigen presentation was profoundly tolerogenic, as it induced T cell deletion even under inflammatory conditions. These results significantly broaden the scope of FDC function and suggest new ways that the complement system and persistent antigen presentation might influence T cell activation and the maintenance of peripheral immune tolerance.
Collapse
Affiliation(s)
- Megan L McCloskey
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA
| | | | | | | |
Collapse
|
40
|
CD8+ T-cell expansion and maintenance after recombinant adenovirus immunization rely upon cooperation between hematopoietic and nonhematopoietic antigen-presenting cells. Blood 2010; 117:1146-55. [PMID: 21088134 DOI: 10.1182/blood-2010-03-272336] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
We have recently reported that CD8(+) T-cell memory maintenance after immunization with recombinant human adenovirus type 5 (rHuAd5) is dependent upon persistent transgene expression beyond the peak of the response. In this report, we have further investigated the location and nature of the cell populations responsible for this sustained response. The draining lymph nodes were found to be important for primary expansion but not for memory maintenance, suggesting that antigen presentation through a nonlymphoid source was required. Using bone marrow chimeric mice, we determined that antigen presentation by nonhematopoietic antigen-presenting cells (APCs) was sufficient for maintenance of CD8(+) T-cell numbers. However, antigen presentation by this mechanism alone yielded a memory population that displayed alterations in phenotype, cytokine production and protective capacity, indicating that antigen presentation through both hematopoietic and nonhematopoietic APCs ultimately defines the memory CD8(+) T-cell response produced by rHuAd5. These results shed new light on the immunobiology of rHuAd5 vectors and provide evidence for a mechanism of CD8(+) T-cell expansion and memory maintenance that relies upon both hematopoietic and nonhematopoietic APCs.
Collapse
|
41
|
Wakim LM, Woodward-Davis A, Bevan MJ. Memory T cells persisting within the brain after local infection show functional adaptations to their tissue of residence. Proc Natl Acad Sci U S A 2010; 107:17872-9. [PMID: 20923878 PMCID: PMC2964240 DOI: 10.1073/pnas.1010201107] [Citation(s) in RCA: 425] [Impact Index Per Article: 30.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The brain is not routinely surveyed by lymphocytes and is defined as an immuno-privileged site. However, viral infection of the brain results in the infiltration and long-term persistence of pathogen-specific CD8(+) T cells. These cells survive without replenishment from the circulation and are referred to as resident memory T cells (Trm). Brain Trm selectively express the integrin CD103, the expression of which is dependent on antigen recognition within the tissue. After clearance of virus, CD8(+) T cells persist in tight clusters, presumably at prior infection hot spots. Antigen persistence is not a prerequisite for T-cell retention, as suggested by the failure to detect viral genomes in the T-cell clusters. Furthermore, we show that an intracranial dendritic cell immunization regimen, which allows the transient introduction of antigen, also results in the generation of memory T cells that persist long term in the brain. Brain Trm die rapidly on isolation from the tissue and fail to undergo recall expansion after adoptive transfer into the bloodstream of antigen-challenged recipients. These ex vivo defects imply a dependency on the local milieu for function and survival. Cumulatively, this work shows that Trm are a specialized population of memory T cells that can be deposited in tissues previously thought to be beyond routine immune surveillance.
Collapse
Affiliation(s)
- Linda M. Wakim
- Department of Immunology, Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195
| | - Amanda Woodward-Davis
- Department of Immunology, Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195
| | - Michael J. Bevan
- Department of Immunology, Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195
| |
Collapse
|
42
|
Price GE, Soboleski MR, Lo CY, Misplon JA, Quirion MR, Houser KV, Pearce MB, Pappas C, Tumpey TM, Epstein SL. Single-dose mucosal immunization with a candidate universal influenza vaccine provides rapid protection from virulent H5N1, H3N2 and H1N1 viruses. PLoS One 2010; 5:e13162. [PMID: 20976273 PMCID: PMC2953831 DOI: 10.1371/journal.pone.0013162] [Citation(s) in RCA: 105] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2010] [Accepted: 09/01/2010] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND The sudden emergence of novel influenza viruses is a global public health concern. Conventional influenza vaccines targeting the highly variable surface glycoproteins hemagglutinin and neuraminidase must antigenically match the emerging strain to be effective. In contrast, "universal" vaccines targeting conserved viral components could be used regardless of viral strain or subtype. Previous approaches to universal vaccination have required protracted multi-dose immunizations. Here we evaluate a single dose universal vaccine strategy using recombinant adenoviruses (rAd) expressing the conserved influenza virus antigens matrix 2 and nucleoprotein. METHODOLOGY/PRINCIPAL FINDINGS In BALB/c mice, administration of rAd via the intranasal route was superior to intramuscular immunization for induction of mucosal responses and for protection against highly virulent H1N1, H3N2, or H5N1 influenza virus challenge. Mucosally vaccinated mice not only survived, but had little morbidity and reduced lung virus titers. Protection was observed as early as 2 weeks post-immunization, and lasted at least 10 months, as did antibodies and lung T cells with activated phenotypes. Virus-specific IgA correlated with but was not essential for protection, as demonstrated in studies with IgA-deficient animals. CONCLUSION/SIGNIFICANCE Mucosal administration of NP and M2-expressing rAd vectors provided rapid and lasting protection from influenza viruses in a subtype-independent manner. Such vaccines could be used in the interval between emergence of a new virus strain and availability of strain-matched vaccines against it. This strikingly effective single-dose vaccination thus represents a candidate off-the-shelf vaccine for emergency use during an influenza pandemic.
Collapse
MESH Headings
- Administration, Intranasal
- Antigens, Viral/immunology
- Humans
- Immunity, Mucosal
- Influenza A Virus, H1N1 Subtype/immunology
- Influenza A Virus, H1N1 Subtype/isolation & purification
- Influenza A Virus, H1N1 Subtype/pathogenicity
- Influenza A Virus, H3N2 Subtype/immunology
- Influenza A Virus, H3N2 Subtype/isolation & purification
- Influenza A Virus, H3N2 Subtype/pathogenicity
- Influenza A Virus, H5N1 Subtype/immunology
- Influenza A Virus, H5N1 Subtype/isolation & purification
- Influenza A Virus, H5N1 Subtype/pathogenicity
- Influenza Vaccines/administration & dosage
- Influenza Vaccines/immunology
- Influenza, Human/prevention & control
- Influenza, Human/virology
- Virulence
Collapse
Affiliation(s)
- Graeme E. Price
- Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Rockville, Maryland, United States of America
| | - Mark R. Soboleski
- Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Rockville, Maryland, United States of America
| | - Chia-Yun Lo
- Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Rockville, Maryland, United States of America
| | - Julia A. Misplon
- Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Rockville, Maryland, United States of America
| | - Mary R. Quirion
- Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Rockville, Maryland, United States of America
| | - Katherine V. Houser
- National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Melissa B. Pearce
- National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Claudia Pappas
- National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Terrence M. Tumpey
- National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Suzanne L. Epstein
- Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Rockville, Maryland, United States of America
- * E-mail:
| |
Collapse
|
43
|
Abstract
Signals orchestrating productive CD4+ T-cell responses are well documented; however, the regulation of contraction of CD4+ T-cell effector populations following the resolution of primary immune responses is not well understood. While distinct mechanisms of T-cell death have been defined, the relative importance of discrete death pathways during the termination of immune responses in vivo remains unclear. Here, we review the current understanding of cell-intrinsic and -extrinsic variables that regulate contraction of CD4+ T-cell effector populations through multiple pathways that operate both initially during T-cell priming and later during the effector phase. We discuss the relative importance of antigen-dependent and -independent mechanisms of CD4+ T-cell contraction during in vivo responses, with a special emphasis on influenza virus infection. In this model, we highlight the roles of greater differentiation and presence in the lung of CD4+ effector T cells, as well as their polarization to particular T-helper subsets, in maximizing contraction. We also discuss the role of autocrine interleukin-2 in limiting the extent of contraction, and we point out that these same factors regulate contraction during secondary CD4+ T-cell responses.
Collapse
Affiliation(s)
- K Kai McKinstry
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | | | | |
Collapse
|
44
|
Kim TS, Hufford MM, Sun J, Fu YX, Braciale TJ. Antigen persistence and the control of local T cell memory by migrant respiratory dendritic cells after acute virus infection. ACTA ACUST UNITED AC 2010; 207:1161-72. [PMID: 20513748 PMCID: PMC2882836 DOI: 10.1084/jem.20092017] [Citation(s) in RCA: 146] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Acute viral infections induce robust adaptive immune responses resulting in virus clearance. Recent evidence suggests that there may be depots of viral antigen that persist in draining lymph nodes (DLNs) after virus clearance and could, therefore, affect the adaptive immune response and memory T cell formation. The nature of these residual antigen depots, the mechanism of antigen persistence, and the impact of the persistent antigen on memory T cells remain ill defined. Using a mouse model of influenza virus infection of the respiratory tract, we identified respiratory dendritic cells (RDCs) as essential for both sampling and presenting residual viral antigen. RDCs in the previously infected lung capture residual viral antigen deposited in an irradiation-resistant cell type. RDCs then transport the viral antigen to the LNs draining the site of infection, where they present the antigen to T cells. Lastly, we document preferential localization of memory T cells to the DLNs after virus clearance as a consequence of presentation of residual viral antigen by the migrant RDC.
Collapse
Affiliation(s)
- Taeg S Kim
- Beirne B. Carter Center for Immunology Research, Department of Microbiology, University of Virginia, Charlottesville, VA 22908, USA
| | | | | | | | | |
Collapse
|
45
|
Cockburn IA, Chen YC, Overstreet MG, Lees JR, van Rooijen N, Farber DL, Zavala F. Prolonged antigen presentation is required for optimal CD8+ T cell responses against malaria liver stage parasites. PLoS Pathog 2010; 6:e1000877. [PMID: 20463809 PMCID: PMC2865532 DOI: 10.1371/journal.ppat.1000877] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2010] [Accepted: 03/25/2010] [Indexed: 11/18/2022] Open
Abstract
Immunization with irradiated sporozoites is currently the most effective vaccination strategy against liver stages of malaria parasites, yet the mechanisms underpinning the success of this approach are unknown. Here we show that the complete development of protective CD8+ T cell responses requires prolonged antigen presentation. Using TCR transgenic cells specific for the malaria circumsporozoite protein, a leading vaccine candidate, we found that sporozoite antigen persists for over 8 weeks after immunization--a remarkable finding since irradiated sporozoites are incapable of replication and do not differentiate beyond early liver stages. Persisting antigen was detected in lymphoid organs and depends on the presence of CD11c+ cells. Prolonged antigen presentation enhanced the magnitude of the CD8+ T cell response in a number of ways. Firstly, reducing the time primed CD8+ T cells were exposed to antigen in vivo severely reduced the final size of the developing memory population. Secondly, fully developed memory cells expanded in previously immunized mice but not when transferred to naïve animals. Finally, persisting antigen was able to prime naïve cells, including recent thymic emigrants, to become functional effector cells capable of eliminating parasites in the liver. Together these data show that the optimal development of protective CD8+ T cell immunity against malaria liver stages is dependent upon the prolonged presentation of sporozoite-derived antigen.
Collapse
Affiliation(s)
- Ian A. Cockburn
- Johns Hopkins Malaria Research Institute and Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Yun-Chi Chen
- Johns Hopkins Malaria Research Institute and Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Michael G. Overstreet
- Johns Hopkins Malaria Research Institute and Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Jason R. Lees
- Department of Surgery, University of Maryland at Baltimore, Baltimore, Maryland, United States of America
| | - Nico van Rooijen
- Vrije Universiteit, VUMC, Department of Molecular Cell Biology, Faculty of Medicine, Amsterdam, The Netherlands
| | - Donna L. Farber
- Department of Surgery, University of Maryland at Baltimore, Baltimore, Maryland, United States of America
| | - Fidel Zavala
- Johns Hopkins Malaria Research Institute and Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
46
|
Abstract
While many aspects of memory T-cell immunobiology have been characterized, we suggest that we know only a fraction of the effector functions that CD4 T cells can bring to bear during secondary challenges. Exploring the full impact of memory CD4 T-cell responses is key to the development of improved vaccines against many prominent pathogens, including influenza viruses, and also to a better understanding of the mechanisms of autoimmunity. Here we discuss factors regulating the generation of memory CD4 T cells during the activation of naïve cells and how the nature of the transition from highly activated effector to resting memory upon the resolution of primary responses might impact memory CD4 T-cell heterogeneity in vivo. We stress that memory CD4 T cells have unique functional attributes beyond the secretion of T helper (Th) subset-associated cytokines that can shape highly effective secondary responses through novel mechanisms. These include the recruitment of innate inflammatory responses at early phases of secondary responses as well as the action of enhanced direct effector functions at later phases, in addition to well-established helper roles for CD8 T-cell and B-cell responses.
Collapse
|
47
|
Brown DM. Cytolytic CD4 cells: Direct mediators in infectious disease and malignancy. Cell Immunol 2010; 262:89-95. [PMID: 20236628 DOI: 10.1016/j.cellimm.2010.02.008] [Citation(s) in RCA: 118] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2010] [Revised: 02/16/2010] [Accepted: 02/19/2010] [Indexed: 12/22/2022]
Abstract
CD4 T cells have traditionally been regarded as helpers and regulators of adaptive immune responses; however, a novel role for CD4 T cells as direct mediators of protection against viral infections has emerged. CD4 T cells with cytolytic potential have been described for almost 40 years, but their role in host protection against infectious disease is only beginning to be realized. In this review, we describe the current literature identifying these cells in patients with various infections, mouse models of viral infection and our own work investigating the development of cytolytic CD4 cells in vivo and in vitro. CD4 CTL are no longer considered an artefact of cell culture and may play a physiological role in viral infections such as EBV, CMV, HIV and influenza. Therefore, vaccine strategies aimed at targeting CD4 CTL should be developed in conjunction with vaccines incorporating B cell and CD8 CTL epitopes.
Collapse
Affiliation(s)
- Deborah M Brown
- University of Nebraska-Lincoln, School of Biological Sciences and Nebraska Center for Virology, Lincoln, NE 68583-0900, USA.
| |
Collapse
|
48
|
Thomas PG, Brown SA, Morris MY, Yue W, So J, Reynolds C, Webby RJ, Doherty PC. Physiological numbers of CD4+ T cells generate weak recall responses following influenza virus challenge. THE JOURNAL OF IMMUNOLOGY 2010; 184:1721-7. [PMID: 20061406 DOI: 10.4049/jimmunol.0901427] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Naive and recall CD4(+) T cell responses were probed with recombinant influenza A viruses incorporating the OVA OT-II peptide. The extent of OT-II-specific CD4(+) T cell expansion was greater following primary exposure, with secondary challenge achieving no significant increase in numbers, despite higher precursor frequencies. Adoptive transfer experiments with OT-II TCR-transgenic T cells established that the predominant memory set is CD62L(hi), whereas the CD62L(lo) precursors make little contribution to the recall response. Unlike the situation described by other investigators, in which the transfer of very large numbers of in vitro-activated CD4 effectors can modify the disease process, providing CD62L(hi) or CD62L(lo) OT-II-specific T cells at physiological levels neither enhanced virus clearance nor altered clinical progression. Some confounding effects of the transgenic model were observed, with decreasing primary expansion efficiency correlating with greater numbers of transferred cells. This was associated with increased levels of mRNA for the proapoptotic molecule Bim in cells recovered following high-dose transfer. However, even with very low numbers of transferred cells, memory T cells did not expand significantly following secondary challenge. A similar result was recorded in mice primed and boosted to respond to an endogenous IA(b)-restricted epitope derived from the influenza virus hemagglutinin glycoprotein. Depletion of CD8(+) T cells during secondary challenge generated an increased accumulation of OT-II-specific T cells but only at the site of infection. Taken together, significant expansion was not a feature of these secondary influenza-specific CD4 T cell responses and the recall of memory did not enhance recovery.
Collapse
Affiliation(s)
- Paul G Thomas
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Kalia V, Sarkar S, Ahmed R. CD8 T-Cell Memory Differentiation during Acute and Chronic Viral Infections. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2010; 684:79-95. [DOI: 10.1007/978-1-4419-6451-9_7] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
|
50
|
McGill J, Heusel JW, Legge KL. Innate immune control and regulation of influenza virus infections. J Leukoc Biol 2009; 86:803-12. [PMID: 19643736 DOI: 10.1189/jlb.0509368] [Citation(s) in RCA: 127] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Adaptive immune responses are critical for the control and clearance of influenza A virus (IAV) infection. However, in recent years, it has become increasingly apparent that innate immune cells, including natural killer cells, alveolar macrophages (aMphi), and dendritic cells (DC) are essential following IAV infection in the direct control of viral replication or in the induction and regulation of virus-specific adaptive immune responses. This review will discuss the role of these innate immune cells following IAV infection, with a particular focus on DC and their ability to induce and regulate the adaptive IAV-specific immune response.
Collapse
Affiliation(s)
- Jodi McGill
- Department of Pathology and Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, Iowa 52242, USA
| | | | | |
Collapse
|