1
|
Hassert M, Pewe LL, He R, Heidarian M, Phruttiwanichakun P, van de Wall S, Mix MR, Salem AK, Badovinac VP, Harty JT. Regenerating murine CD8+ lung tissue resident memory T cells after targeted radiation exposure. J Exp Med 2024; 221:e20231144. [PMID: 38363548 PMCID: PMC10873130 DOI: 10.1084/jem.20231144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 11/06/2023] [Accepted: 01/31/2024] [Indexed: 02/17/2024] Open
Abstract
Radiation exposure occurs during medical procedures, nuclear accidents, or spaceflight, making effective medical countermeasures a public health priority. Naïve T cells are highly sensitive to radiation-induced depletion, although their numbers recover with time. Circulating memory CD8+ T cells are also depleted by radiation; however, their numbers do not recover. Critically, the impact of radiation exposure on tissue-resident memory T cells (TRM) remains unknown. Here, we found that sublethal thorax-targeted radiation resulted in the rapid and prolonged numerical decline of influenza A virus (IAV)-specific lung TRM in mice, but no decline in antigen-matched circulating memory T cells. Prolonged loss of lung TRM was associated with decreased heterosubtypic immunity. Importantly, boosting with IAV-epitope expressing pathogens that replicate in the lungs or peripheral tissues or with a peripherally administered mRNA vaccine regenerated lung TRM that was derived largely from circulating memory CD8+ T cells. Designing effective vaccination strategies to regenerate TRM will be important in combating the immunological effects of radiation exposure.
Collapse
Affiliation(s)
- Mariah Hassert
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Lecia L. Pewe
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Rui He
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA, USA
| | - Mohammad Heidarian
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Department of Pathology Graduate Programs, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Pornpoj Phruttiwanichakun
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA, USA
| | - Stephanie van de Wall
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Madison R. Mix
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Interdisciplinary Graduate Program in Immunology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Medical Scientist Training Program, University of Iowa, Iowa City, IA, USA
| | - Aliasger K. Salem
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA, USA
- Interdisciplinary Graduate Program in Immunology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Vladimir P. Badovinac
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Department of Pathology Graduate Programs, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Interdisciplinary Graduate Program in Immunology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - John T. Harty
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Department of Pathology Graduate Programs, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Interdisciplinary Graduate Program in Immunology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| |
Collapse
|
2
|
West EE, Merle NS, Kamiński MM, Palacios G, Kumar D, Wang L, Bibby JA, Overdahl K, Jarmusch AK, Freeley S, Lee DY, Thompson JW, Yu ZX, Taylor N, Sitbon M, Green DR, Bohrer A, Mayer-Barber KD, Afzali B, Kazemian M, Scholl-Buergi S, Karall D, Huemer M, Kemper C. Loss of CD4 + T cell-intrinsic arginase 1 accelerates Th1 response kinetics and reduces lung pathology during influenza infection. Immunity 2023; 56:2036-2053.e12. [PMID: 37572656 PMCID: PMC10576612 DOI: 10.1016/j.immuni.2023.07.014] [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: 10/14/2022] [Revised: 06/01/2023] [Accepted: 07/19/2023] [Indexed: 08/14/2023]
Abstract
Arginase 1 (Arg1), the enzyme catalyzing the conversion of arginine to ornithine, is a hallmark of IL-10-producing immunoregulatory M2 macrophages. However, its expression in T cells is disputed. Here, we demonstrate that induction of Arg1 expression is a key feature of lung CD4+ T cells during mouse in vivo influenza infection. Conditional ablation of Arg1 in CD4+ T cells accelerated both virus-specific T helper 1 (Th1) effector responses and its resolution, resulting in efficient viral clearance and reduced lung pathology. Using unbiased transcriptomics and metabolomics, we found that Arg1-deficiency was distinct from Arg2-deficiency and caused altered glutamine metabolism. Rebalancing this perturbed glutamine flux normalized the cellular Th1 response. CD4+ T cells from rare ARG1-deficient patients or CRISPR-Cas9-mediated ARG1-deletion in healthy donor cells phenocopied the murine cellular phenotype. Collectively, CD4+ T cell-intrinsic Arg1 functions as an unexpected rheostat regulating the kinetics of the mammalian Th1 lifecycle with implications for Th1-associated tissue pathologies.
Collapse
Affiliation(s)
- Erin E West
- Complement and Inflammation Research Section (CIRS), National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD, USA.
| | - Nicolas S Merle
- Complement and Inflammation Research Section (CIRS), National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Marcin M Kamiński
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Gustavo Palacios
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Dhaneshwar Kumar
- Immunoregulation Section, Kidney Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, MD, USA
| | - Luopin Wang
- Departments of Biochemistry and Computer Science, Purdue University, West Lafayette, IN, USA
| | - Jack A Bibby
- Complement and Inflammation Research Section (CIRS), National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Kirsten Overdahl
- Immunity, Inflammation, and Disease Laboratory, Division of Intramural Research, National Institute of Environmental Health Sciences (NIEHS), NIH, Research Triangle Park, NC, USA
| | - Alan K Jarmusch
- Immunity, Inflammation, and Disease Laboratory, Division of Intramural Research, National Institute of Environmental Health Sciences (NIEHS), NIH, Research Triangle Park, NC, USA
| | - Simon Freeley
- School of Immunology and Microbial Sciences, King's College London, Guy's Hospital, Great Maze Pond, London SE1 9RT, UK
| | | | - J Will Thompson
- Proteomics and Metabolomics Shared Resource, Center for Genomic and Computational Biology, Duke University, Durham, NC, USA
| | - Zu-Xi Yu
- Pathology Core, NHLBI, NIH, Bethesda, MD, USA
| | - Naomi Taylor
- Pediatric Oncology Branch, Rare Tumor Initiative, Center for Cancer Research, National Cancer Institute (NCI), NIH, Bethesda, MD, USA; Institut de Génétique Moléculaire de Montpellier (IGMM), Université de Montpellier, CNRS, Montpellier, France
| | - Marc Sitbon
- Pediatric Oncology Branch, Rare Tumor Initiative, Center for Cancer Research, National Cancer Institute (NCI), NIH, Bethesda, MD, USA; Institut de Génétique Moléculaire de Montpellier (IGMM), Université de Montpellier, CNRS, Montpellier, France
| | - Douglas R Green
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Andrea Bohrer
- Inflammation and Innate Immunity Unit, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, MD, USA
| | - Katrin D Mayer-Barber
- Inflammation and Innate Immunity Unit, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, MD, USA
| | - Behdad Afzali
- Immunoregulation Section, Kidney Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, MD, USA
| | - Majid Kazemian
- Departments of Biochemistry and Computer Science, Purdue University, West Lafayette, IN, USA
| | - Sabine Scholl-Buergi
- Clinic for Pediatrics I, Inherited Metabolic Disorders, Medical University of Innsbruck, Innsbruck, Austria
| | - Daniela Karall
- Clinic for Pediatrics I, Inherited Metabolic Disorders, Medical University of Innsbruck, Innsbruck, Austria
| | - Martina Huemer
- Division of Metabolism and Children's Research Center, University Children's Hospital Zurich, University of Zurich, Zurich, Switzerland; Department of Pediatric Endocrinology and Diabetology, University Children's Hospital Basel, Basel, Switzerland; Department of Pediatrics, Landeskrankenhaus (LKH) Bregenz, Bregenz, Austria
| | - Claudia Kemper
- Complement and Inflammation Research Section (CIRS), National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD, USA.
| |
Collapse
|
3
|
Evrard M, Becht E, Fonseca R, Obers A, Park SL, Ghabdan-Zanluqui N, Schroeder J, Christo SN, Schienstock D, Lai J, Burn TN, Clatch A, House IG, Beavis P, Kallies A, Ginhoux F, Mueller SN, Gottardo R, Newell EW, Mackay LK. Single-cell protein expression profiling resolves circulating and resident memory T cell diversity across tissues and infection contexts. Immunity 2023:S1074-7613(23)00262-5. [PMID: 37392736 DOI: 10.1016/j.immuni.2023.06.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 03/08/2023] [Accepted: 06/07/2023] [Indexed: 07/03/2023]
Abstract
Memory CD8+ T cells can be broadly divided into circulating (TCIRCM) and tissue-resident memory T (TRM) populations. Despite well-defined migratory and transcriptional differences, the phenotypic and functional delineation of TCIRCM and TRM cells, particularly across tissues, remains elusive. Here, we utilized an antibody screening platform and machine learning prediction pipeline (InfinityFlow) to profile >200 proteins in TCIRCM and TRM cells in solid organs and barrier locations. High-dimensional analyses revealed unappreciated heterogeneity within TCIRCM and TRM cell lineages across nine different organs after either local or systemic murine infection models. Additionally, we demonstrated the relative effectiveness of strategies allowing for the selective ablation of TCIRCM or TRM populations across organs and identified CD55, KLRG1, CXCR6, and CD38 as stable markers for characterizing memory T cell function during inflammation. Together, these data and analytical framework provide an in-depth resource for memory T cell classification in both steady-state and inflammatory conditions.
Collapse
Affiliation(s)
- Maximilien Evrard
- Department of Microbiology and Immunology, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Parkville, VIC 3010, Australia.
| | - Etienne Becht
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Raissa Fonseca
- Department of Microbiology and Immunology, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Parkville, VIC 3010, Australia
| | - Andreas Obers
- Department of Microbiology and Immunology, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Parkville, VIC 3010, Australia
| | - Simone L Park
- Department of Microbiology and Immunology, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Parkville, VIC 3010, Australia
| | - Nagela Ghabdan-Zanluqui
- Department of Microbiology and Immunology, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Parkville, VIC 3010, Australia
| | - Jan Schroeder
- Department of Microbiology and Immunology, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Parkville, VIC 3010, Australia
| | - Susan N Christo
- Department of Microbiology and Immunology, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Parkville, VIC 3010, Australia
| | - Dominik Schienstock
- Department of Microbiology and Immunology, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Parkville, VIC 3010, Australia
| | - Junyun Lai
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia; Cancer Immunology Program, Peter MacCallum Cancer Centre, Parkville, VIC 3010, Australia
| | - Thomas N Burn
- Department of Microbiology and Immunology, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Parkville, VIC 3010, Australia
| | - Allison Clatch
- Department of Microbiology and Immunology, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Parkville, VIC 3010, Australia
| | - Imran G House
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia; Cancer Immunology Program, Peter MacCallum Cancer Centre, Parkville, VIC 3010, Australia
| | - Paul Beavis
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia; Cancer Immunology Program, Peter MacCallum Cancer Centre, Parkville, VIC 3010, Australia
| | - Axel Kallies
- Department of Microbiology and Immunology, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Parkville, VIC 3010, Australia
| | - Florent Ginhoux
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A∗STAR), Singapore 138648, Singapore
| | - Scott N Mueller
- Department of Microbiology and Immunology, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Parkville, VIC 3010, Australia
| | - Raphael Gottardo
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; Centre Hospitalier Universitaire du Vaud and University of Lausanne, Lausanne 1011, Switzerland
| | - Evan W Newell
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Laura K Mackay
- Department of Microbiology and Immunology, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Parkville, VIC 3010, Australia.
| |
Collapse
|
4
|
Su FY, Zhao QH, Dahotre SN, Gamboa L, Bawage SS, Silva Trenkle AD, Zamat A, Phuengkham H, Ahmed R, Santangelo PJ, Kwong GA. In vivo mRNA delivery to virus-specific T cells by light-induced ligand exchange of MHC class I antigen-presenting nanoparticles. SCIENCE ADVANCES 2022; 8:eabm7950. [PMID: 35196075 PMCID: PMC8865765 DOI: 10.1126/sciadv.abm7950] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 01/25/2022] [Indexed: 05/06/2023]
Abstract
Simultaneous delivery of mRNA to multiple populations of antigen (Ag)-specific CD8+ T cells is challenging given the diversity of peptide epitopes and polymorphism of class I major histocompatibility complexes (MHCI). We developed Ag-presenting nanoparticles (APNs) for mRNA delivery using pMHCI molecules that were refolded with photocleavable peptides to allow rapid ligand exchange by UV light and site-specifically conjugated with a lipid tail for postinsertion into preformed mRNA lipid nanoparticles. Across different TCR transgenic mouse models (P14, OT-1, and Pmel), UV-exchanged APNs bound and transfected their cognate Ag-specific CD8+ T cells equivalent to APNs produced using conventionally refolded pMHCI molecules. In mice infected with PR8 influenza, multiplexed delivery of UV-exchanged APNs against three immunodominant epitopes led to ~50% transfection of a VHH mRNA reporter in cognate Ag-specific CD8+ T cells. Our data show that UV-mediated peptide exchange can be used to rapidly produce APNs for mRNA delivery to multiple populations of Ag-specific T cells in vivo.
Collapse
Affiliation(s)
- Fang-Yi Su
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
| | - Qingyang Henry Zhao
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
| | - Shreyas N. Dahotre
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
| | - Lena Gamboa
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
| | - Swapnil Subhash Bawage
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
| | - Aaron D. Silva Trenkle
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
| | - Ali Zamat
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
| | - Hathaichanok Phuengkham
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
| | - Rafi Ahmed
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA 30317, USA
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA 30322, USA
- Winship Cancer Institute of Emory University, Atlanta, GA 30322, USA
| | - Philip J. Santangelo
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
| | - Gabriel A. Kwong
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
- Winship Cancer Institute of Emory University, Atlanta, GA 30322, USA
- Institute for Electronics and Nanotechnology, Georgia Institute of Technology, Atlanta, GA 30332, USA
- Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA
- Integrated Cancer Research Center, Georgia Institute of Technology, Atlanta, GA 30332, USA
- Georgia ImmunoEngineering Consortium, Emory University and Georgia Institute of Technology, Atlanta, GA 30332, USA
| |
Collapse
|
5
|
Lauterbach H, Schmidt S, Katchar K, Qing X, Iacobucci C, Hwang A, Schlienger K, Berka U, Raguz J, Ahmadi-Erber S, Schippers T, Stemeseder F, Pinschewer DD, Matushansky I, Orlinger KK. Development and Characterization of a Novel Non-Lytic Cancer Immunotherapy Using a Recombinant Arenavirus Vector Platform. Front Oncol 2021; 11:732166. [PMID: 34722273 PMCID: PMC8551556 DOI: 10.3389/fonc.2021.732166] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 09/16/2021] [Indexed: 12/11/2022] Open
Abstract
Engineered viral vectors represent a promising strategy to trigger antigen-specific antitumor T cell responses. Arenaviruses have been widely studied because of their ability to elicit potent and protective T cell responses. Here, we provide an overview of a novel intravenously administered, replication-competent, non-lytic arenavirus-based vector technology that delivers tumor antigens to induce antigen-specific anti-cancer T cell responses. Preclinical studies in mice and cell culture experiments with human peripheral blood mononuclear cells demonstrate that arenavirus vectors preferentially infect antigen-presenting cells. This, in conjunction with a non-lytic functional activation of the infected antigen-presenting cells, leads to a robust antigen-specific CD8+ T cell response. T cell migration to, and infiltration of, the tumor microenvironment has been demonstrated in various preclinical tumor models with vectors encoding self- and non-self-antigens. The available data also suggest that arenavirus-based vector therapy can induce immunological memory protecting from tumor rechallenge. Based on promising preclinical data, a phase 1/2 clinical trial was initiated and is currently ongoing to test the activity and safety of arenavirus vectors, HB-201 and HB-202, created using lymphocytic choriomeningitis virus and Pichinde virus, respectively. Both vectors have been engineered to deliver non-oncogenic versions of the human papilloma virus 16 (HPV16) antigens E7 and E6 and will be injected intravenously with or without an initial intratumoral dose. This dose escalation/expansion study is being conducted in patients with recurrent or metastatic HPV16+ cancers. Promising preliminary data from this ongoing clinical study have been reported. Immunogenicity data from several patients demonstrate that a single injection of HB-201 or HB-202 monotherapy is highly immunogenic, as evidenced by an increase in inflammatory cytokines/chemokines and the expansion of antigen-specific CD8+ T cell responses. This response can be further enhanced by alternating injections of HB-202 and HB-201, which has resulted in frequencies of circulating HPV16 E7/E6-specific CD8+ T cells of up to 40% of the total CD8+ T cell compartment in peripheral blood in analyses to date. Treatment with intravenous administration also resulted in a disease control rate of 73% among 11 evaluable patients with head and neck cancer dosed every three weeks, including 2 patients with a partial response.
Collapse
Affiliation(s)
| | | | - Kia Katchar
- Hookipa Pharma Inc., New York, NY, United States
| | | | | | - Andy Hwang
- Hookipa Pharma Inc., New York, NY, United States
| | | | - Ursula Berka
- Hookipa Pharma Inc., New York, NY, United States
| | - Josipa Raguz
- Hookipa Pharma Inc., New York, NY, United States
| | | | | | | | - Daniel D Pinschewer
- Department of Biomedicine - Haus Petersplatz, Division of Experimental Virology, University of Basel, Basel, Switzerland
| | | | | |
Collapse
|
6
|
McGee HM, Marciscano AE, Campbell AM, Monjazeb AM, Kaech SM, Teijaro JR. Parallels Between the Antiviral State and the Irradiated State. J Natl Cancer Inst 2021; 113:969-979. [PMID: 33252657 PMCID: PMC8502484 DOI: 10.1093/jnci/djaa190] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 10/07/2020] [Accepted: 11/16/2020] [Indexed: 01/12/2023] Open
Abstract
Improved understanding of host antiviral defense and antitumor immunity have elucidated molecular pathways important to both processes. During viral infection, RNA or DNA in the host cell serves as a danger signal that initiates the antiviral response. Recent studies have elucidated similarities in the signaling pathways activated by viruses and the signaling pathways induced by tumor DNA that is released into the cytoplasm of irradiated tumor cells. Both the host defense to viral infection and the sterile inflammation provoked by radiotherapy induce a type I interferon response that is necessary for pathogen control and immune-mediated tumor control, respectively. These findings have led to the hypothesis that radiotherapy employs a form of viral mimicry.
Collapse
Affiliation(s)
- Heather M McGee
- NOMIS Center for Immunobiology and Microbial Pathogenesis, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Ariel E Marciscano
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| | - Allison M Campbell
- Department of Therapeutic Radiology, Yale School of Medicine, New Haven, CT, USA
| | - Arta M Monjazeb
- Department of Radiation Oncology, UC Davis Comprehensive Cancer Center, Sacramento, CA, USA
| | - Susan M Kaech
- NOMIS Center for Immunobiology and Microbial Pathogenesis, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - John R Teijaro
- Department of Immunology and Microbiology, Scripps Research Institute, La Jolla, CA, USA
| |
Collapse
|
7
|
Anthony SM, Van Braeckel-Budimir N, Moioffer SJ, van de Wall S, Shan Q, Vijay R, Sompallae R, Hartwig SM, Jensen IJ, Varga SM, Butler NS, Xue HH, Badovinac VP, Harty JT. Protective function and durability of mouse lymph node-resident memory CD8 + T cells. eLife 2021; 10:68662. [PMID: 34143731 PMCID: PMC8213409 DOI: 10.7554/elife.68662] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 06/09/2021] [Indexed: 12/13/2022] Open
Abstract
Protective lung tissue-resident memory CD8+T cells (Trm) form after influenza A virus (IAV) infection. We show that IAV infection of mice generates CD69+CD103+and other memory CD8+T cell populations in lung-draining mediastinal lymph nodes (mLNs) from circulating naive or memory CD8+T cells. Repeated antigen exposure, mimicking seasonal IAV infections, generates quaternary memory (4M) CD8+T cells that protect mLN from viral infection better than 1M CD8+T cells. Better protection by 4M CD8+T cells associates with enhanced granzyme A/B expression and stable maintenance of mLN CD69+CD103+4M CD8+T cells, vs the steady decline of CD69+CD103+1M CD8+T cells, paralleling the durability of protective CD69+CD103+4M vs 1M in the lung after IAV infection. Coordinated upregulation in canonical Trm-associated genes occurs in circulating 4M vs 1M populations without the enrichment of canonical downregulated Trm genes. Thus, repeated antigen exposure arms circulating memory CD8+T cells with enhanced capacity to form long-lived populations of Trm that enhance control of viral infections of the mLN.
Collapse
Affiliation(s)
- Scott M Anthony
- Department of Pathology, The University of Iowa, Iowa City, United States
| | | | - Steven J Moioffer
- Department of Pathology, The University of Iowa, Iowa City, United States
| | | | - Qiang Shan
- Department of Microbiology and Immunology, The University of Iowa, Iowa City, United States.,Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, United States
| | - Rahul Vijay
- Department of Microbiology and Immunology, The University of Iowa, Iowa City, United States
| | | | - Stacey M Hartwig
- Department of Microbiology and Immunology, The University of Iowa, Iowa City, United States
| | - Isaac J Jensen
- Department of Pathology, The University of Iowa, Iowa City, United States.,Department of Microbiology and Immunology, The University of Iowa, Iowa City, United States.,Interdisciplinary Graduate Program in Immunology, The University of Iowa, Iowa City, United States
| | - Steven M Varga
- Department of Pathology, The University of Iowa, Iowa City, United States.,Department of Microbiology and Immunology, The University of Iowa, Iowa City, United States.,Interdisciplinary Graduate Program in Immunology, The University of Iowa, Iowa City, United States
| | - Noah S Butler
- Department of Microbiology and Immunology, The University of Iowa, Iowa City, United States.,Interdisciplinary Graduate Program in Immunology, The University of Iowa, Iowa City, United States
| | - Hai-Hui Xue
- Department of Microbiology and Immunology, The University of Iowa, Iowa City, United States.,Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, United States.,Interdisciplinary Graduate Program in Immunology, The University of Iowa, Iowa City, United States
| | - Vladimir P Badovinac
- Department of Pathology, The University of Iowa, Iowa City, United States.,Department of Microbiology and Immunology, The University of Iowa, Iowa City, United States.,Interdisciplinary Graduate Program in Immunology, The University of Iowa, Iowa City, United States
| | - John T Harty
- Department of Pathology, The University of Iowa, Iowa City, United States.,Interdisciplinary Graduate Program in Immunology, The University of Iowa, Iowa City, United States
| |
Collapse
|
8
|
Pauken KE, Godec J, Odorizzi PM, Brown KE, Yates KB, Ngiow SF, Burke KP, Maleri S, Grande SM, Francisco LM, Ali MA, Imam S, Freeman GJ, Haining WN, Wherry EJ, Sharpe AH. The PD-1 Pathway Regulates Development and Function of Memory CD8 + T Cells following Respiratory Viral Infection. Cell Rep 2021; 31:107827. [PMID: 32610128 DOI: 10.1016/j.celrep.2020.107827] [Citation(s) in RCA: 81] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 07/05/2019] [Accepted: 06/05/2020] [Indexed: 12/11/2022] Open
Abstract
The PD-1 pathway regulates dysfunctional T cells in chronic infection and cancer, but the role of this pathway during acute infection remains less clear. Here, we demonstrate that PD-1 signals are needed for optimal memory. Mice deficient in the PD-1 pathway exhibit impaired CD8+ T cell memory following acute influenza infection, including reduced virus-specific CD8+ T cell numbers and compromised recall responses. PD-1 blockade during priming leads to similar differences early post-infection but without the defect in memory formation, suggesting that timing and/or duration of PD-1 blockade could be tailored to modulate host responses. Our studies reveal a role for PD-1 as an integrator of CD8+ T cell signals that promotes CD8+ T cell memory formation and suggest PD-1 continues to fine-tune CD8+ T cells after they migrate into non-lymphoid tissues. These findings have important implications for PD-1-based immunotherapy, in which PD-1 inhibition may influence memory responses in patients.
Collapse
Affiliation(s)
- Kristen E Pauken
- Department of Immunology, Blavatnik Institute, Harvard Medical School, and Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Jernej Godec
- Department of Immunology, Blavatnik Institute, Harvard Medical School, and Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA; Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Pamela M Odorizzi
- Institute for Immunology and Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Keturah E Brown
- Department of Immunology, Blavatnik Institute, Harvard Medical School, and Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Kathleen B Yates
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Shin Foong Ngiow
- Institute for Immunology and Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Kelly P Burke
- Department of Immunology, Blavatnik Institute, Harvard Medical School, and Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA; Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA 02115, USA
| | - Seth Maleri
- Department of Immunology, Blavatnik Institute, Harvard Medical School, and Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Shannon M Grande
- Department of Immunology, Blavatnik Institute, Harvard Medical School, and Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Loise M Francisco
- Department of Immunology, Blavatnik Institute, Harvard Medical School, and Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Mohammed-Alkhatim Ali
- Institute for Immunology and Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Sabrina Imam
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Gordon J Freeman
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA 02115, USA
| | - W Nicholas Haining
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Division of Hematology/Oncology, Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - E John Wherry
- Institute for Immunology and Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA.
| | - Arlene H Sharpe
- Department of Immunology, Blavatnik Institute, Harvard Medical School, and Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115, USA.
| |
Collapse
|
9
|
Ebina-Shibuya R, West EE, Spolski R, Li P, Oh J, Kazemian M, Gromer D, Swanson P, Du N, McGavern DB, Leonard WJ. Thymic stromal lymphopoietin limits primary and recall CD8 + T-cell anti-viral responses. eLife 2021; 10:e61912. [PMID: 33439121 PMCID: PMC7806261 DOI: 10.7554/elife.61912] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Accepted: 12/06/2020] [Indexed: 11/16/2022] Open
Abstract
Thymic stromal lymphopoietin (TSLP) is a cytokine that acts directly on CD4+ T cells and dendritic cells to promote progression of asthma, atopic dermatitis, and allergic inflammation. However, a direct role for TSLP in CD8+ T-cell primary responses remains controversial and its role in memory CD8+ T cell responses to secondary viral infection is unknown. Here, we investigate the role of TSLP in both primary and recall responses in mice using two different viral systems. Interestingly, TSLP limited the primary CD8+ T-cell response to influenza but did not affect T cell function nor significantly alter the number of memory CD8+ T cells generated after influenza infection. However, TSLP inhibited memory CD8+ T-cell responses to secondary viral infection with influenza or acute systemic LCMV infection. These data reveal a previously unappreciated role for TSLP on recall CD8+ T-cell responses in response to viral infection, findings with potential translational implications.
Collapse
Affiliation(s)
- Risa Ebina-Shibuya
- Laboratory of Molecular Immunology, Immunology Center, National Heart, Lung, and Blood Institute National Institutes of HealthBethesdaUnited States
| | - Erin E West
- Laboratory of Molecular Immunology, Immunology Center, National Heart, Lung, and Blood Institute National Institutes of HealthBethesdaUnited States
| | - Rosanne Spolski
- Laboratory of Molecular Immunology, Immunology Center, National Heart, Lung, and Blood Institute National Institutes of HealthBethesdaUnited States
| | - Peng Li
- Laboratory of Molecular Immunology, Immunology Center, National Heart, Lung, and Blood Institute National Institutes of HealthBethesdaUnited States
| | - Jangsuk Oh
- Laboratory of Molecular Immunology, Immunology Center, National Heart, Lung, and Blood Institute National Institutes of HealthBethesdaUnited States
| | - Majid Kazemian
- Laboratory of Molecular Immunology, Immunology Center, National Heart, Lung, and Blood Institute National Institutes of HealthBethesdaUnited States
| | - Daniel Gromer
- Laboratory of Molecular Immunology, Immunology Center, National Heart, Lung, and Blood Institute National Institutes of HealthBethesdaUnited States
| | - Phillip Swanson
- Viral Immunology & Intravital Imaging Section, National Institute of Neurological Disorders and Stroke, National Institutes of HealthBethesdaUnited States
| | - Ning Du
- Laboratory of Molecular Immunology, Immunology Center, National Heart, Lung, and Blood Institute National Institutes of HealthBethesdaUnited States
| | - Dorian B McGavern
- Viral Immunology & Intravital Imaging Section, National Institute of Neurological Disorders and Stroke, National Institutes of HealthBethesdaUnited States
| | - Warren J Leonard
- Laboratory of Molecular Immunology, Immunology Center, National Heart, Lung, and Blood Institute National Institutes of HealthBethesdaUnited States
| |
Collapse
|
10
|
He Q, Jiang L, Cao K, Zhang L, Xie X, Zhang S, Ding X, He Y, Zhang M, Qiu T, Jin X, Zhao C, Zhang X, Xu J. A Systemic Prime-Intrarectal Pull Strategy Raises Rectum-Resident CD8+ T Cells for Effective Protection in a Murine Model of LM-OVA Infection. Front Immunol 2020; 11:571248. [PMID: 33072113 PMCID: PMC7541937 DOI: 10.3389/fimmu.2020.571248] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 08/18/2020] [Indexed: 01/01/2023] Open
Abstract
As the entry sites of many pathogens such as human immunodeficiency virus (HIV), mucosal sites are defended by rapidly reacting resident memory T cells (TRM). TRMs represent a special subpopulation of memory T cells that persist long term in non-lymphoid sites without entering the circulation and provide the “sensing and alarming” role in the first-line defense against infection. The rectum and vagina are the two primary mucosal portals for HIV entry. However, compared to vaginal TRM, rectal TRM is poorly understood. Herein, we investigated the optimal vaccination strategy to induce rectal TRM. We identified an intranasal prime–intrarectal boost (pull) strategy that is effective in engaging rectal TRM alongside circulating memory T cells and demonstrated its protective efficacy in mice against infection of Listeria monocytogenes. On the contrary, the same vaccine delivered via either intranasal or intrarectal route failed to raise rectal TRM, setting it apart from vaginal TRM, which can be induced by both intranasal and intrarectal immunizations. Moreover, intramuscular prime was also effective in inducing rectal TRM in combination with intrarectal pull, highlighting the need of a primed systemic T cell response. A comparison of different pull modalities led to the identification that raising rectal TRM is mainly driven by local antigen presence. We further demonstrated the interval between prime and boost steps to be critical for the induction of rectal TRM, revealing circulating recently activated CD8+ T cells as the likely primary pullable precursor of rectal TRM. Altogether, our studies lay a new framework for harnessing rectal TRM in vaccine development.
Collapse
Affiliation(s)
- Qian He
- Shanghai Public Health Clinical Center and Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Lang Jiang
- Shanghai Public Health Clinical Center and Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Kangli Cao
- Shanghai Public Health Clinical Center and Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Linxia Zhang
- Shanghai Public Health Clinical Center and Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xinci Xie
- Shanghai Public Health Clinical Center and Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Shuye Zhang
- Shanghai Public Health Clinical Center and Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xiangqing Ding
- Shanghai Public Health Clinical Center and Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yongquan He
- Shanghai Public Health Clinical Center and Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Miaomiao Zhang
- Shanghai Public Health Clinical Center and Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Tianyi Qiu
- Shanghai Public Health Clinical Center and Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xuanxuan Jin
- Shanghai Public Health Clinical Center and Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Chen Zhao
- Shanghai Public Health Clinical Center and Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xiaoyan Zhang
- Shanghai Public Health Clinical Center and Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jianqing Xu
- Shanghai Public Health Clinical Center and Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
11
|
Urban SL, Jensen IJ, Shan Q, Pewe LL, Xue HH, Badovinac VP, Harty JT. Peripherally induced brain tissue-resident memory CD8 + T cells mediate protection against CNS infection. Nat Immunol 2020; 21:938-949. [PMID: 32572242 PMCID: PMC7381383 DOI: 10.1038/s41590-020-0711-8] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 05/13/2020] [Indexed: 12/27/2022]
Abstract
The central nervous system (CNS) is classically viewed as immune-privileged; however, recent advances highlight interactions between the peripheral immune system and CNS in controlling infections and tissue homeostasis. Tissue-resident memory (TRM) CD8+ T cells in the CNS are generated after brain infections, but it is unknown whether CNS infection is required to generate brain TRM cells. We show that peripheral infections generate antigen-specific CD8+ memory T cells in the brain that adopt a unique TRM signature. Upon depletion of circulating and perivascular memory T cells, this brain signature was enriched and the surveilling properties of brain TRM cells was revealed by intravital imaging. Notably, peripherally induced brain TRM cells showed evidence of rapid activation and enhanced cytokine production and mediated protection after brain infections. These data reveal that peripheral immunizations can generate brain TRM cells and will guide potential use of T cells as therapeutic strategies against CNS infections and neurological diseases.
Collapse
Affiliation(s)
- Stina L Urban
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Isaac J Jensen
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Interdisciplinary Graduate Program in Immunology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Qiang Shan
- Center for Discovery and Innovation, Hackensack University Medical Center, Hackensack, NJ, USA
| | - Lecia L Pewe
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Hai-Hui Xue
- Center for Discovery and Innovation, Hackensack University Medical Center, Hackensack, NJ, USA
- VA New Jersey Health Care System, East Orange, NJ, USA
| | - Vladimir P Badovinac
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Interdisciplinary Graduate Program in Immunology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, USA
| | - John T Harty
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA.
- Interdisciplinary Graduate Program in Immunology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
12
|
T Cell Factor 1 Suppresses CD103+ Lung Tissue-Resident Memory T Cell Development. Cell Rep 2020; 31:107484. [DOI: 10.1016/j.celrep.2020.03.048] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 01/07/2020] [Accepted: 03/13/2020] [Indexed: 01/31/2023] Open
|
13
|
Van Braeckel-Budimir N, Varga SM, Badovinac VP, Harty JT. Repeated Antigen Exposure Extends the Durability of Influenza-Specific Lung-Resident Memory CD8 + T Cells and Heterosubtypic Immunity. Cell Rep 2019; 24:3374-3382.e3. [PMID: 30257199 DOI: 10.1016/j.celrep.2018.08.073] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 08/01/2018] [Accepted: 08/24/2018] [Indexed: 01/29/2023] Open
Abstract
Lung-resident primary memory CD8+ T cell populations (Trm) induced by a single influenza infection decline within months, rendering the host susceptible to new heterosubtypic influenza infections. Here, we demonstrate that, relative to single virus exposure, repeated antigen exposure dramatically alters the dynamics of influenza-specific lung Trm populations. Lung Trm derived from repeatedly stimulated circulating memory CD8+ T cells exhibit extended durability and protective heterosubtypic immunity relative to primary lung Trm. Parabiosis studies reveal that the enhanced durability of lung Trm after multiple antigen encounters resulted from the generation of long-lasting circulating effector memory (Tem) populations, which maintained the ability to be recruited to the lung parenchyma and converted to Trm, in combination with enhanced survival of these cells in the lung. Thus, generating a long-lasting Trm precursor pool through repeated intranasal immunizations might be a promising strategy to establish long-lasting lung Trm-mediated heterosubtypic immunity against influenza.
Collapse
Affiliation(s)
| | - Steven M Varga
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA 52242, USA; Department of Pathology, University of Iowa, Iowa City, IA 52242, USA; Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA 52242, USA
| | - Vladimir P Badovinac
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA 52242, USA; Department of Pathology, University of Iowa, Iowa City, IA 52242, USA; Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA 52242, USA
| | - John T Harty
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA 52242, USA; Department of Pathology, University of Iowa, Iowa City, IA 52242, USA; Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA 52242, USA.
| |
Collapse
|
14
|
Brown FD, Sen DR, LaFleur MW, Godec J, Lukacs-Kornek V, Schildberg FA, Kim HJ, Yates KB, Ricoult SJH, Bi K, Trombley JD, Kapoor VN, Stanley IA, Cremasco V, Danial NN, Manning BD, Sharpe AH, Haining WN, Turley SJ. Fibroblastic reticular cells enhance T cell metabolism and survival via epigenetic remodeling. Nat Immunol 2019; 20:1668-1680. [PMID: 31636464 DOI: 10.1038/s41590-019-0515-x] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 09/11/2019] [Indexed: 12/16/2022]
Abstract
Lymph node fibroblastic reticular cells (FRCs) respond to signals from activated T cells by releasing nitric oxide, which inhibits T cell proliferation and restricts the size of the expanding T cell pool. Whether interactions with FRCs also support the function or differentiation of activated CD8+ T cells is not known. Here we report that encounters with FRCs enhanced cytokine production and remodeled chromatin accessibility in newly activated CD8+ T cells via interleukin-6. These epigenetic changes facilitated metabolic reprogramming and amplified the activity of pro-survival pathways through differential transcription factor activity. Accordingly, FRC conditioning significantly enhanced the persistence of virus-specific CD8+ T cells in vivo and augmented their differentiation into tissue-resident memory T cells. Our study demonstrates that FRCs play a role beyond restricting T cell expansion-they can also shape the fate and function of CD8+ T cells.
Collapse
Affiliation(s)
- Flavian D Brown
- Division of Medical Sciences, Harvard Medical School, Boston, MA, USA.,Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.,Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA, USA.,Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA.,Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, USA.,Neon Therapeutics Inc., Cambridge, MA, USA
| | - Debattama R Sen
- Division of Medical Sciences, Harvard Medical School, Boston, MA, USA.,Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Martin W LaFleur
- Division of Medical Sciences, Harvard Medical School, Boston, MA, USA.,Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.,Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA, USA.,Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA
| | - Jernej Godec
- Division of Medical Sciences, Harvard Medical School, Boston, MA, USA.,Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.,Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA, USA.,Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA
| | - Veronika Lukacs-Kornek
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, USA.,Institute of Experimental Immunology, University Hospital of the Rheinische Friedrich-Wilhelms-University, Bonn, Germany
| | - Frank A Schildberg
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA, USA.,Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA.,Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, USA.,Clinic for Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | - Hye-Jung Kim
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA, USA.,Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Kathleen B Yates
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.,Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Stéphane J H Ricoult
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Kevin Bi
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.,Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Justin D Trombley
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA, USA.,Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA
| | - Varun N Kapoor
- Department of Cancer Immunology, Genentech, South San Francisco, CA, USA
| | - Illana A Stanley
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA.,Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Viviana Cremasco
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, USA.,Immuno-Oncology, Novartis Institutes for BioMedical Research, Cambridge, MA, USA
| | - Nika N Danial
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA.,Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Brendan D Manning
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Arlene H Sharpe
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA, USA. .,Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA. .,Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA, USA. .,Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA.
| | - W Nicholas Haining
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA. .,Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA, USA. .,Division of Pediatric Hematology and Oncology, Children's Hospital, Boston, MA, USA. .,Merck Research Laboratories, Boston, MA, USA.
| | - Shannon J Turley
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, USA. .,Department of Cancer Immunology, Genentech, South San Francisco, CA, USA.
| |
Collapse
|
15
|
Westerhof LM, McGuire K, MacLellan L, Flynn A, Gray JI, Thomas M, Goodyear CS, MacLeod MK. Multifunctional cytokine production reveals functional superiority of memory CD4 T cells. Eur J Immunol 2019; 49:2019-2029. [PMID: 31177549 PMCID: PMC6900100 DOI: 10.1002/eji.201848026] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 04/29/2019] [Accepted: 06/06/2019] [Indexed: 11/20/2022]
Abstract
T cell protective immunity is associated with multifunctional memory cells that produce several different cytokines. Currently, our understanding of when and how these cells are generated is limited. We have used an influenza virus mouse infection model to investigate whether the cytokine profile of memory T cells is reflective of primary responding cells or skewed toward a distinct profile. We found that, in comparison to primary cells, memory T cells tended to make multiple cytokines simultaneously. Analysis of the timings of release of cytokine by influenza virus‐specific T cells, demonstrated that primary responding CD4 T cells from lymphoid organs were unable to produce a sustained cytokine response. In contrast CD8 T cells, memory CD4 T cells, and primary responding CD4 T cells from the lung produced a sustained cytokine response throughout the restimulation period. Moreover, memory CD4 T cells were more resistant than primary responding CD4 T cells to inhibitors that suppress T cell receptor signaling. Together, these data suggest that memory CD4 T cells display superior cytokine responses compared to primary responding cells. These data are key to our ability to identify the cues that drive the generation of protective memory CD4 T cells following infection.
Collapse
Affiliation(s)
- Lotus M Westerhof
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, 120 University Place, University of Glasgow, Glasgow, UK.,GLAZgo Discovery Centre, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Kris McGuire
- GLAZgo Discovery Centre, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Lindsay MacLellan
- GLAZgo Discovery Centre, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Ashley Flynn
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, 120 University Place, University of Glasgow, Glasgow, UK
| | - Joshua I Gray
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, 120 University Place, University of Glasgow, Glasgow, UK
| | - Matthew Thomas
- Respiratory, Inflammation and Autoimmunity IMED, AstraZeneca, Gothenburg, Sweden
| | - Carl S Goodyear
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, 120 University Place, University of Glasgow, Glasgow, UK.,GLAZgo Discovery Centre, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Megan Kl MacLeod
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, 120 University Place, University of Glasgow, Glasgow, UK
| |
Collapse
|
16
|
Shane HL, Reagin KL, Klonowski KD. The Respiratory Environment Diverts the Development of Antiviral Memory CD8 T Cells. THE JOURNAL OF IMMUNOLOGY 2018; 200:3752-3761. [PMID: 29669782 DOI: 10.4049/jimmunol.1701268] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 03/24/2018] [Indexed: 12/21/2022]
Abstract
Our understanding of memory CD8+ T cells has been largely derived from acute, systemic infection models. However, memory CD8+ T cells generated from mucosal infection exhibit unique properties and, following respiratory infection, are not maintained in the lung long term. To better understand how infection route modifies memory differentiation, we compared murine CD8+ T cell responses to a vesicular stomatitis virus (VSV) challenge generated intranasally (i.n.) or i.v. The i.n. infection resulted in greater peak expansion of VSV-specific CD8+ T cells. However, this numerical advantage was rapidly lost during the contraction phase of the immune response, resulting in memory CD8+ T cell numerical deficiencies when compared with i.v. infection. Interestingly, the antiviral CD8+ T cells generated in response to i.n. VSV exhibited a biased and sustained proportion of early effector cells (CD127loKLRG1lo) akin to the developmental program favored after i.n. influenza infection, suggesting that respiratory infection broadly favors an incomplete memory differentiation program. Correspondingly, i.n. VSV infection resulted in lower CD122 expression and eomesodermin levels by VSV-specific CD8+ T cells, further indicative of an inferior transition to bona fide memory. These results may be due to distinct (CD103+CD11b+) dendritic cell subsets in the i.n. versus i.v. T cell priming environments, which express molecules that regulate T cell signaling and the balance between tolerance and immunity. Therefore, we propose that distinct immunization routes modulate both the quality and quantity of antiviral effector and memory CD8+ T cells in response to an identical pathogen and should be considered in CD8+ T cell-based vaccine design.
Collapse
Affiliation(s)
- Hillary L Shane
- Department of Cellular Biology, University of Georgia, Athens, GA 30602-2607
| | - Katie L Reagin
- Department of Cellular Biology, University of Georgia, Athens, GA 30602-2607
| | | |
Collapse
|
17
|
Martin MD, Danahy DB, Hartwig SM, Harty JT, Badovinac VP. Revealing the Complexity in CD8 T Cell Responses to Infection in Inbred C57B/6 versus Outbred Swiss Mice. Front Immunol 2017; 8:1527. [PMID: 29213267 PMCID: PMC5702636 DOI: 10.3389/fimmu.2017.01527] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 10/27/2017] [Indexed: 12/30/2022] Open
Abstract
Recent work has suggested that current mouse models may underrepresent the complexity of human immune responses. While most mouse immunology studies utilize inbred mouse strains, it is unclear if conclusions drawn from inbred mice can be extended to all mouse strains or generalized to humans. We recently described a “surrogate activation marker” approach that could be used to track polyclonal CD8 T cell responses in inbred and outbred mice and noted substantial discord in the magnitude and kinetics of CD8 T cell responses in individual outbred mice following infection. However, how the memory CD8 T cell response develops following infection and the correlates of memory CD8 T cell-mediated protection against re-infection in outbred mice remains unknown. In this study, we investigated development of pathogen-specific memory CD8 T cell responses in inbred C57B/6 and outbred National Institutes of Health Swiss mice following lymphocytic choriomeningitis virus or L. monocytogenes infection. Interestingly, the size of the memory CD8 T cell pool generated and rate of phenotypic progression was considerably more variable in individual outbred compared to inbred mice. Importantly, while prior infection provided both inbred and outbred cohorts of mice with protection against re-infection that was dependent on the dose of primary infection, levels of memory CD8 T cells generated and degree of protection against re-infection did not correlate with primary infection dose in all outbred mice. While variation in CD8 T cell responses to infection is not entirely surprising due to the genetic diversity present, analysis of infection-induced immunity in outbred hosts may reveal hidden complexity in CD8 T cell responses in genetically diverse populations and might help us further bridge the gap between mouse and human studies.
Collapse
Affiliation(s)
- Matthew D Martin
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Derek B Danahy
- Interdisciplinary Graduate Program in Immunology, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Stacey M Hartwig
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA, United States.,Department of Microbiology, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - John T Harty
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA, United States.,Interdisciplinary Graduate Program in Immunology, Carver College of Medicine, University of Iowa, Iowa City, IA, United States.,Department of Microbiology, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Vladimir P Badovinac
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA, United States.,Interdisciplinary Graduate Program in Immunology, Carver College of Medicine, University of Iowa, Iowa City, IA, United States.,Department of Microbiology, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| |
Collapse
|
18
|
Moffett HF, Cartwright ANR, Kim HJ, Godec J, Pyrdol J, Äijö T, Martinez GJ, Rao A, Lu J, Golub TR, Cantor H, Sharpe AH, Novina CD, Wucherpfennig KW. The microRNA miR-31 inhibits CD8 + T cell function in chronic viral infection. Nat Immunol 2017; 18:791-799. [PMID: 28530712 PMCID: PMC5753758 DOI: 10.1038/ni.3755] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Accepted: 04/27/2017] [Indexed: 12/13/2022]
Abstract
During infection, antigen-specific T cells undergo tightly regulated developmental transitions controlled by transcriptional and post-transcriptional regulation of gene expression. We found that the microRNA miR-31 was strongly induced by activation of the T cell antigen receptor (TCR) in a pathway involving calcium and activation of the transcription factor NFAT. During chronic infection with lymphocytic choriomeningitis virus (LCMV) clone 13, miR-31-deficent mice recovered from clinical disease, while wild-type mice continued to show signs of disease. This disease phenotype was explained by the presence of larger numbers of cytokine-secreting LCMV-specific CD8+ T cells in miR-31-deficent mice than in wild-type mice. Mechanistically, miR-31 increased the sensitivity of T cells to type I interferons, which interfered with effector T cell function and increased the expression of several proteins related to T cell dysfunction during chronic infection. These studies identify miR-31 as an important regulator of T cell exhaustion in chronic infection.
Collapse
Affiliation(s)
- Howell F Moffett
- Department of Cancer Immunology &Virology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Adam N R Cartwright
- Department of Cancer Immunology &Virology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Hye-Jung Kim
- Department of Cancer Immunology &Virology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Jernej Godec
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts, USA
| | - Jason Pyrdol
- Department of Cancer Immunology &Virology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Tarmo Äijö
- Department of Information and Computer Science, Aalto University School of Science, Aalto, Finland
| | - Gustavo J Martinez
- La Jolla Institute for Allergy and Immunology, La Jolla, California, USA
| | - Anjana Rao
- La Jolla Institute for Allergy and Immunology, La Jolla, California, USA
| | - Jun Lu
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Todd R Golub
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Harvey Cantor
- Department of Cancer Immunology &Virology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Arlene H Sharpe
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts, USA
| | - Carl D Novina
- Department of Cancer Immunology &Virology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Kai W Wucherpfennig
- Department of Cancer Immunology &Virology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
19
|
He R, Yang X, Liu C, Chen X, Wang L, Xiao M, Ye J, Wu Y, Ye L. Efficient control of chronic LCMV infection by a CD4 T cell epitope-based heterologous prime-boost vaccination in a murine model. Cell Mol Immunol 2017; 15:815-826. [PMID: 28287115 DOI: 10.1038/cmi.2017.3] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 12/08/2016] [Accepted: 12/08/2016] [Indexed: 02/07/2023] Open
Abstract
CD4+ T cells are essential for sustaining CD8+ T cell responses during a chronic infection. The adoptive transfer of virus-specific CD4+ T cells has been shown to efficiently rescue exhausted CD8+ T cells. However, the question of whether endogenous virus-specific CD4+ T cell responses can be enhanced by certain vaccination strategies and subsequently reinvigorate exhausted CD8+ T cells remains unexplored. In this study, we developed a CD4+ T cell epitope-based heterologous prime-boost immunization strategy and examined the efficacy of this strategy using a mouse model of chronic lymphocytic choriomeningitis virus (LCMV) infection. We primed chronically LCMV-infected mice with a Listeria monocytogenes vector that expressed the LCMV glycoprotein-specific I-Ab-restricted CD4+ T cell epitope GP61-80 (LM-GP61) and subsequently boosted the primed mice with an influenza virus A (PR8 strain) vector that expressed the same CD4+ T cell epitope (IAV-GP61). This heterologous prime-boost vaccination strategy elicited strong anti-viral CD4+ T cell responses, which further improved both the quantity and quality of the virus-specific CD8+ T cells and led to better control of the viral loads. The combination of this strategy and the blockade of the programmed cell death-1 (PD-1) inhibitory pathway further enhanced the anti-viral CD8+ T cell responses and viral clearance. Thus, a heterologous prime-boost immunization that selectively induces virus-specific CD4+ T cell responses in conjunction with blockade of the inhibitory pathway may represent a promising therapeutic approach to treating patients with chronic viral infections.
Collapse
Affiliation(s)
- Ran He
- Center for Clinical Laboratory, Zhujiang Hospital, Southern Medical University, 510515, Guangzhou, China.,Institute of Immunology, Medical School, Third Military Medical University, 400038, Chongqing, China
| | - Xinxin Yang
- Institute of Immunology, Medical School, Third Military Medical University, 400038, Chongqing, China
| | - Cheng Liu
- Institute of Immunology, Medical School, Third Military Medical University, 400038, Chongqing, China
| | - Xiangyu Chen
- Institute of Immunology, Medical School, Third Military Medical University, 400038, Chongqing, China
| | - Lin Wang
- Institute of Immunology, Medical School, Third Military Medical University, 400038, Chongqing, China
| | - Minglu Xiao
- Institute of Immunology, Medical School, Third Military Medical University, 400038, Chongqing, China
| | - Jianqiang Ye
- Ministry of Education Key Laboratory for Avian Preventive Medicine, College of Veterinary Medicine, Yangzhou University, 225009, Yangzhou, China, Jiangsu.,Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, 225009, Yangzhou, China, Jiangsu
| | - Yuzhang Wu
- Center for Clinical Laboratory, Zhujiang Hospital, Southern Medical University, 510515, Guangzhou, China.,Institute of Immunology, Medical School, Third Military Medical University, 400038, Chongqing, China
| | - Lilin Ye
- Institute of Immunology, Medical School, Third Military Medical University, 400038, Chongqing, China.
| |
Collapse
|
20
|
Slütter B, Van Braeckel-Budimir N, Abboud G, Varga SM, Salek-Ardakani S, Harty JT. Dynamics of influenza-induced lung-resident memory T cells underlie waning heterosubtypic immunity. Sci Immunol 2017; 2:2/7/eaag2031. [PMID: 28783666 DOI: 10.1126/sciimmunol.aag2031] [Citation(s) in RCA: 220] [Impact Index Per Article: 31.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 12/06/2016] [Indexed: 12/14/2022]
Abstract
Lung-resident memory CD8 T cells (TRM) induced by influenza A virus (IAV) that are pivotal for providing subtype-transcending protection against IAV infection (heterosubtypic immunity) are not maintained long term, causing gradual loss of protection. The short-lived nature of lung TRM contrasts sharply with long-term maintenance of TRM induced by localized infections in the skin and in other tissues. We show that the decline in lung TRM is determined by an imbalance between apoptosis and lung recruitment and conversion to TRM of circulating memory cells. We show that circulating effector memory cells (TEM) rather than central memory cells (TCM) are the precursors for conversion to lung TRM Time-dependent changes in expression of genes critical for lymphocyte trafficking and TRM differentiation, in concert with enrichment of TCM, diminish the capacity of circulating memory CD8 T cells to form TRM with time, explaining why IAV-induced TRM are not stably maintained. Systemic booster immunization, through increasing the number of circulating TEM, increases lung TRM, providing a potential new avenue to enhance IAV vaccines.
Collapse
Affiliation(s)
- Bram Slütter
- Department of Microbiology, University of Iowa, Iowa City, IA 52242, USA.,Cluster of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, 2333 CC Leiden, Netherlands
| | | | - Georges Abboud
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL 32611, USA
| | - Steven M Varga
- Department of Microbiology, University of Iowa, Iowa City, IA 52242, USA.,Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA 52242, USA.,Department of Pathology, University of Iowa, Iowa City, IA 52242, USA
| | - Shahram Salek-Ardakani
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL 32611, USA
| | - John T Harty
- Department of Microbiology, University of Iowa, Iowa City, IA 52242, USA. .,Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA 52242, USA.,Department of Pathology, University of Iowa, Iowa City, IA 52242, USA
| |
Collapse
|
21
|
Bally APR, Austin JW, Boss JM. Genetic and Epigenetic Regulation of PD-1 Expression. THE JOURNAL OF IMMUNOLOGY 2016; 196:2431-7. [PMID: 26945088 DOI: 10.4049/jimmunol.1502643] [Citation(s) in RCA: 169] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The inhibitory immune receptor programmed cell death-1 (PD-1) is intricately regulated. In T cells, PD-1 is expressed in response to most immune challenges, but it is rapidly downregulated in acute settings, allowing for normal immune responses. On chronically stimulated Ag-specific T cells, PD-1 expression remains high, leading to an impaired response to stimuli. Ab blockade of PD-1 interactions during chronic Ag settings partially restores immune function and is now used clinically to treat a variety of devastating cancers. Understanding the regulation of PD-1 expression may be useful for developing novel immune-based therapies. In this review, the molecular mechanisms that drive dynamic PD-1 expression during acute and chronic antigenic stimuli are discussed. An array of cis-DNA elements, transcription factors, and epigenetic components, including DNA methylation and histone modifications, control PD-1 expression. The interplay between these regulators fine-tunes PD-1 expression in different inflammatory environments and across numerous cell types to modulate immune responses.
Collapse
Affiliation(s)
- Alexander P R Bally
- Department of Microbiology and Immunology and Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA 30322
| | - James W Austin
- Department of Microbiology and Immunology and Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA 30322
| | - Jeremy M Boss
- Department of Microbiology and Immunology and Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA 30322
| |
Collapse
|
22
|
Martin MD, Kim MT, Shan Q, Sompallae R, Xue HH, Harty JT, Badovinac VP. Phenotypic and Functional Alterations in Circulating Memory CD8 T Cells with Time after Primary Infection. PLoS Pathog 2015; 11:e1005219. [PMID: 26485703 PMCID: PMC4618693 DOI: 10.1371/journal.ppat.1005219] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Accepted: 09/21/2015] [Indexed: 12/22/2022] Open
Abstract
Memory CD8 T cells confer increased protection to immune hosts upon secondary viral, bacterial, and parasitic infections. The level of protection provided depends on the numbers, quality (functional ability), and location of memory CD8 T cells present at the time of infection. While primary memory CD8 T cells can be maintained for the life of the host, the full extent of phenotypic and functional changes that occur over time after initial antigen encounter remains poorly characterized. Here we show that critical properties of circulating primary memory CD8 T cells, including location, phenotype, cytokine production, maintenance, secondary proliferation, secondary memory generation potential, and mitochondrial function change with time after infection. Interestingly, phenotypic and functional alterations in the memory population are not due solely to shifts in the ratio of effector (CD62Llo) and central memory (CD62Lhi) cells, but also occur within defined CD62Lhi memory CD8 T cell subsets. CD62Lhi memory cells retain the ability to efficiently produce cytokines with time after infection. However, while it is was not formally tested whether changes in CD62Lhi memory CD8 T cells over time occur in a cell intrinsic manner or are due to selective death and/or survival, the gene expression profiles of CD62Lhi memory CD8 T cells change, phenotypic heterogeneity decreases, and mitochondrial function and proliferative capacity in either a lymphopenic environment or in response to antigen re-encounter increase with time. Importantly, and in accordance with their enhanced proliferative and metabolic capabilities, protection provided against chronic LCMV clone-13 infection increases over time for both circulating memory CD8 T cell populations and for CD62Lhi memory cells. Taken together, the data in this study reveal that memory CD8 T cells continue to change with time after infection and suggest that the outcome of vaccination strategies designed to elicit protective memory CD8 T cells using single or prime-boost immunizations depends upon the timing between antigen encounters.
Collapse
Affiliation(s)
- Matthew D. Martin
- 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
| | - Marie T. Kim
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, Iowa, United States of America
| | - Qiang Shan
- Department of Microbiology, University of Iowa, Iowa City, Iowa, United States of America
| | - Ramakrishna Sompallae
- Iowa Institute of Human Genetics Bioinformatics Division, University of Iowa, Iowa City, Iowa, United States of America
| | - Hai-Hui Xue
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, Iowa, United States of America
- Department of Microbiology, University of Iowa, Iowa City, Iowa, United States of America
| | - John T. Harty
- 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
- Department of Microbiology, University of Iowa, Iowa City, Iowa, United States of America
| | - Vladimir P. Badovinac
- 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
|
23
|
Evaluation of non-reciprocal heterologous immunity between unrelated viruses. Virology 2015; 482:89-97. [PMID: 25838115 DOI: 10.1016/j.virol.2015.03.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Revised: 12/17/2014] [Accepted: 03/03/2015] [Indexed: 11/22/2022]
Abstract
Heterologous immunity refers to the phenomenon whereby a history of an immune response against one pathogen can provide a level of immunity to a second unrelated pathogen. Previous investigations have shown that heterologous immunity is not necessarily reciprocal, such as in the case of vaccinia virus (VACV). Replication of VACV is reduced in mice immune to a variety of pathogens, while VACV fails to induce immunity to several of the same pathogens, including lymphocytic choriomeningitis virus (LCMV). Here we examine the lack of reciprocity of heterologous immunity between VACV and LCMV and find that they induce qualitatively different memory CD8 T cells. However, depending on the repertoire of an individual host, VACV can provide protection against LCMV simply by experimentally amplifying the quantity of T cells cross-reactive with the two viruses. Thus, one cause for lack of reciprocity is differences in the frequencies of cross-reactive T cells in immune hosts.
Collapse
|
24
|
Inducible RNAi in vivo reveals that the transcription factor BATF is required to initiate but not maintain CD8+ T-cell effector differentiation. Proc Natl Acad Sci U S A 2014; 112:512-7. [PMID: 25548173 DOI: 10.1073/pnas.1413291112] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The differentiation of effector CD8(+) T cells is critical for the development of protective responses to pathogens and for effective vaccines. In the first few hours after activation, naive CD8(+) T cells initiate a transcriptional program that leads to the formation of effector and memory T cells, but the regulation of this process is poorly understood. Investigating the role of specific transcription factors (TFs) in determining CD8(+) effector T-cell fate by gene knockdown with RNAi is challenging because naive T cells are refractory to transduction with viral vectors without extensive ex vivo stimulation, which obscures the earliest events in effector differentiation. To overcome this obstacle, we developed a novel strategy to test the function of genes in naive CD8(+) T cells in vivo by creating bone marrow chimera from hematopoietic progenitors transduced with an inducible shRNA construct. Following hematopoietic reconstitution, this approach allowed inducible in vivo gene knockdown in any cell type that developed from this transduced progenitor pool. We demonstrated that lentivirus-transduced progenitor cells could reconstitute normal hematopoiesis and develop into naive CD8(+) T cells that were indistinguishable from wild-type naive T cells. This experimental system enabled induction of efficient gene knockdown in vivo without subsequent manipulation. We applied this strategy to show that the TF BATF is essential for initial commitment of naive CD8(+) T cells to effector development but becomes dispensable by 72h. This approach makes possible the study of gene function in vivo in unperturbed cells of hematopoietic origin that are refractory to viral transduction.
Collapse
|
25
|
Zickovich JM, Meyer SI, Yagita H, Obar JJ. Agonistic anti-CD40 enhances the CD8+ T cell response during vesicular stomatitis virus infection. PLoS One 2014; 9:e106060. [PMID: 25166494 PMCID: PMC4148391 DOI: 10.1371/journal.pone.0106060] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Accepted: 07/31/2014] [Indexed: 02/07/2023] Open
Abstract
Intracellular pathogens are capable of inducing vigorous CD8+ T cell responses. However, we do not entirely understand the factors driving the generation of large pools of highly protective memory CD8+ T cells. Here, we studied the generation of endogenous ovalbumin-specific memory CD8+ T cells following infection with recombinant vesicular stomatitis virus (VSV) and Listeria monocytogenes (LM). VSV infection resulted in the generation of a large ovalbumin-specific memory CD8+ T cell population, which provided minimal protective immunity that waned with time. In contrast, the CD8+ T cell population of LM-ova provided protective immunity and remained stable with time. Agonistic CD40 stimulation during CD8+ T cell priming in response to VSV infection enabled the resultant memory CD8+ T cell population to provide strong protective immunity against secondary infection. Enhanced protective immunity by agonistic anti-CD40 was dependent on CD70. Agonistic anti-CD40 not only enhanced the size of the resultant memory CD8+ T cell population, but enhanced their polyfunctionality and sensitivity to antigen. Our data suggest that immunomodulation of CD40 signaling may be a key adjuvant to enhance CD8+ T cell response during development of VSV vaccine strategies.
Collapse
Affiliation(s)
- Julianne M. Zickovich
- Department of Microbiology & Immunology, Montana State University, Bozeman, Montana, United States of America
| | - Susan I. Meyer
- Department of Microbiology & Immunology, Montana State University, Bozeman, Montana, United States of America
| | - Hideo Yagita
- Department of Immunology, Juntendo University, School of Medicine, Toyko, Japan
| | - Joshua J. Obar
- Department of Microbiology & Immunology, Montana State University, Bozeman, Montana, United States of America
- * E-mail:
| |
Collapse
|
26
|
Qualitative and quantitative analysis of adenovirus type 5 vector-induced memory CD8 T cells: not as bad as their reputation. J Virol 2013; 87:6283-95. [PMID: 23536658 DOI: 10.1128/jvi.00465-13] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
It has been reported that adenovirus (Ad)-primed CD8 T cells may display a distinct and partially exhausted phenotype. Given the practical implications of this claim, we decided to analyze in detail the quality of Ad-primed CD8 T cells by directly comparing these cells to CD8 T cells induced through infection with lymphocytic choriomeningitis virus (LCMV). We found that localized immunization with intermediate doses of Ad vector induces a moderate number of functional CD8 T cells which qualitatively match those found in LCMV-infected mice. The numbers of these cells may be efficiently increased by additional adenoviral boosting, and, importantly, the generated secondary memory cells cannot be qualitatively differentiated from those induced by primary infection with replicating virus. Quantitatively, DNA priming prior to Ad vaccination led to even higher numbers of memory cells. In this case, the vaccination led to the generation of a population of memory cells characterized by relatively low CD27 expression and high CD127 and killer cell lectin-like receptor subfamily G member 1 (KLRG1) expression. These memory CD8 T cells were capable of proliferating in response to viral challenge and protecting against infection with live virus. Furthermore, viral challenge was followed by sustained expansion of the memory CD8 T-cell population, and the generated memory cells did not appear to have been driven toward exhaustive differentiation. Based on these findings, we suggest that adenovirus-based prime-boost regimens (including Ad serotype 5 [Ad5] and Ad5-like vectors) represent an effective means to induce a substantially expanded, long-lived population of high-quality transgene-specific memory CD8 T cells.
Collapse
|
27
|
Laidlaw BJ, Decman V, Ali MAA, Abt MC, Wolf AI, Monticelli LA, Mozdzanowska K, Angelosanto JM, Artis D, Erikson J, Wherry EJ. Cooperativity between CD8+ T cells, non-neutralizing antibodies, and alveolar macrophages is important for heterosubtypic influenza virus immunity. PLoS Pathog 2013; 9:e1003207. [PMID: 23516357 PMCID: PMC3597515 DOI: 10.1371/journal.ppat.1003207] [Citation(s) in RCA: 124] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2011] [Accepted: 01/10/2013] [Indexed: 11/29/2022] Open
Abstract
Seasonal epidemics of influenza virus result in ∼36,000 deaths annually in the United States. Current vaccines against influenza virus elicit an antibody response specific for the envelope glycoproteins. However, high mutation rates result in the emergence of new viral serotypes, which elude neutralization by preexisting antibodies. T lymphocytes have been reported to be capable of mediating heterosubtypic protection through recognition of internal, more conserved, influenza virus proteins. Here, we demonstrate using a recombinant influenza virus expressing the LCMV GP33-41 epitope that influenza virus-specific CD8+ T cells and virus-specific non-neutralizing antibodies each are relatively ineffective at conferring heterosubtypic protective immunity alone. However, when combined virus-specific CD8 T cells and non-neutralizing antibodies cooperatively elicit robust protective immunity. This synergistic improvement in protective immunity is dependent, at least in part, on alveolar macrophages and/or other lung phagocytes. Overall, our studies suggest that an influenza vaccine capable of eliciting both CD8+ T cells and antibodies specific for highly conserved influenza proteins may be able to provide heterosubtypic protection in humans, and act as the basis for a potential “universal” vaccine. Influenza virus continues to pose a significant risk to global health and is responsible for thousands of deaths each year in the United States. This threat is largely due to the ability of the influenza virus to undergo rapid changes, allowing it to escape from immune responses elicited by previous infections or vaccinations. Certain internal determinants of the influenza virus are largely conserved across different viral strains and represent attractive targets for potential “universal” influenza vaccines. Here, we demonstrated that cross-subtype protection against the influenza virus could be obtained through simultaneous priming of multiple arms of the immune response against conserved elements of the influenza virus. These results suggest a novel strategy that could potentially form a primary component of a universal influenza vaccine capable of providing long-lasting protection.
Collapse
Affiliation(s)
- Brian J. Laidlaw
- Department of Microbiology and Institute for Immunology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Vilma Decman
- The Wistar Institute, Philadelphia, Pennsylvania, United States of America
| | - Mohammed-Alkhatim A. Ali
- Department of Microbiology and Institute for Immunology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Michael C. Abt
- Department of Microbiology and Institute for Immunology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Amaya I. Wolf
- The Wistar Institute, Philadelphia, Pennsylvania, United States of America
| | - Laurel A. Monticelli
- Department of Microbiology and Institute for Immunology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
| | | | - Jill M. Angelosanto
- Department of Microbiology and Institute for Immunology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - David Artis
- Department of Microbiology and Institute for Immunology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Jan Erikson
- The Wistar Institute, Philadelphia, Pennsylvania, United States of America
| | - E. John Wherry
- Department of Microbiology and Institute for Immunology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
28
|
Pang IK, Ichinohe T, Iwasaki A. IL-1R signaling in dendritic cells replaces pattern-recognition receptors in promoting CD8⁺ T cell responses to influenza A virus. Nat Immunol 2013; 14:246-53. [PMID: 23314004 PMCID: PMC3577947 DOI: 10.1038/ni.2514] [Citation(s) in RCA: 112] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2012] [Accepted: 12/07/2012] [Indexed: 12/25/2022]
Abstract
Immune responses to vaccines require direct recognition of pathogen-associated molecular patterns (PAMPs) through pattern recognition receptors (PRRs) on dendritic cells (DCs). Unlike vaccines, infection by a live pathogen often impairs DC function and inflicts additional damage to the host. Here, we found that following live influenza A infection, signaling through the interleukin-1 receptor (IL-1R), but not the PRRs, TLR7 and RIG-I, is required for productive CD8+ T cell priming. DCs activated by IL-1 in trans were both required and sufficient for the generation of virus-specific CD8+ T cell immunity. Our data reveal a critical role of a bystander cytokine in CD8+ T cell priming during a live viral infection.
Collapse
Affiliation(s)
- Iris K Pang
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA
| | | | | |
Collapse
|
29
|
Patel A, Kobinger GP. Evaluation of mismatched immunity against influenza viruses. Future Virol 2012. [DOI: 10.2217/fvl.12.105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Prior immunity against influenza A viruses generates sterilizing immunity against matched (homologous) viruses and varying levels of protection against mismatched (heterologous) viruses of the same or different subtypes. Natural immunity carries the risk of high morbidity and mortality, therefore immunization offers the best preventative measure. Antibody responses against the viral hemagglutinin protein correlate with protection in humans and evidence increasingly supports a role for robust cellular immune responses. By exploiting mismatched immunity, current conventional and experimental vaccine candidates can improve the generation of cross-protective immune responses against heterologous viruses. Experimental vaccines such as virus-like particles, DNA vectors, viral vectors and broadly neutralizing antibodies are able to expand cross-protection through mismatched B- and T-cell responses. However, the generation of mismatched immune responses can also have the opposite effect and impair protective immunity. This review discusses mismatched immunity in the context of natural infection and immunization. Additionally, we discuss strategies to exploit mismatched immunity in order to improve current conventional and experimental influenza A virus vaccines.
Collapse
Affiliation(s)
- Ami Patel
- San Raffaele-Telethon Institute of Gene Therapy (hSR-TIGET), Milan, Italy Division of Gene Therapy & Regenerative Medicine, via Olgettina 58, Milan, Italy, 20132
| | - Gary P Kobinger
- Department of Medical Microbiology, University of Manitoba, Winnipeg, Manitoba, Canada
- Department of Immunology, University of Manitoba, Winnipeg, Manitoba, Canada
- Special Pathogens Programme, National Microbiology Laboratory, Public Health Agency of Canada, Canadian Science Centre for Human & Animal Health, 1015 Arlington Street, Winnipeg, Manitoba, Canada, R3E 3R2
| |
Collapse
|
30
|
Jegaskanda S, Reece JC, De Rose R, Stambas J, Sullivan L, Brooks AG, Kent SJ, Sexton A. Comparison of influenza and SIV specific CD8 T cell responses in macaques. PLoS One 2012; 7:e32431. [PMID: 22403659 PMCID: PMC3293803 DOI: 10.1371/journal.pone.0032431] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2011] [Accepted: 01/30/2012] [Indexed: 12/12/2022] Open
Abstract
Macaques are a potentially useful non-human primate model to compare memory T-cell immunity to acute virus pathogens such as influenza virus and effector T-cell responses to chronic viral pathogens such as SIV. However, immunological reagents to study influenza CD8+ T-cell responses in the macaque model are limited. We recently developed an influenza-SIV vaccination model of pigtail macaques (Macaca nemestrina) and used this to study both influenza-specific and SIV-specific CD8+ T-cells in 39 pigtail macaques expressing the common Mane-A*10+ (Mane-A01*084) MHC-I allele. To perform comparative studies between influenza and SIV responses a common influenza nucleoprotein-specific CD8+ T-cell response was mapped to a minimal epitope (termed RA9), MHC-restricted to Mane-A*10 and an MHC tetramer developed to study this response. Influenza-specific memory CD8+ T-cell response maintained a highly functional profile in terms of multitude of effector molecule expression (CD107a, IFN-γ, TNF-α, MIP-1β and IL-2) and showed high avidity even in the setting of SIV infection. In contrast, within weeks following active SIV infection, SIV-specific CD8+ effector T-cells expressed fewer cytokines/degranulation markers and had a lower avidity compared to influenza specific CD8+ T-cells. Further, the influenza specific memory CD8 T-cell response retained stable expression of the exhaustion marker programmed death-marker-1 (PD-1) and co-stimulatory molecule CD28 following infection with SIV. This contrasted with the effector SIV-specific CD8+ T-cells following SIV infection which expressed significantly higher amounts of PD-1 and lower amounts of CD28. Our results suggest that strategies to maintain a more functional CD8+ T-cell response, profile may assist in controlling HIV disease.
Collapse
Affiliation(s)
- Sinthujan Jegaskanda
- Department of Microbiology and Immunology, University of Melbourne, Melbourne, Victoria, Australia
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Prlic M, Sacks JA, Bevan MJ. Dissociating markers of senescence and protective ability in memory T cells. PLoS One 2012; 7:e32576. [PMID: 22396780 PMCID: PMC3292574 DOI: 10.1371/journal.pone.0032576] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2011] [Accepted: 01/27/2012] [Indexed: 01/30/2023] Open
Abstract
No unique transcription factor or biomarker has been identified to reliably distinguish effector from memory T cells. Instead a set of surface markers including IL-7Rα and KLRG1 is commonly used to predict the potential of CD8 effector T cells to differentiate into memory cells. Similarly, these surface markers together with the tumor necrosis factor family member CD27 are frequently used to predict a memory T cell's ability to mount a recall response. Expression of these markers changes every time a memory cell is stimulated and repeated stimulation can lead to T cell senescence and loss of memory T cell responsiveness. This is a concern for prime–boost vaccine strategies which repeatedly stimulate T cells with the aim of increasing memory T cell frequency. The molecular cues that cause senescence are still unknown, but cell division history is likely to play a major role. We sought to dissect the roles of inflammation and cell division history in developing T cell senescence and their impact on the expression pattern of commonly used markers of senescence. We developed a system that allows priming of CD8 T cells with minimal inflammation and without acquisition of maximal effector function, such as granzyme expression, but a cell division history similar to priming with systemic inflammation. Memory cells derived from minimal effector T cells are fully functional upon rechallenge, have full access to non-lymphoid tissue and appear to be less senescent by phenotype upon rechallenge. However, we report here that these currently used biomarkers to measure senescence do not predict proliferative potential or protective ability, but merely reflect initial priming conditions.
Collapse
Affiliation(s)
- Martin Prlic
- Department of Immunology and Howard Hughes Medical Institute, University of Washington, Seattle, Washington, United States of America
- * E-mail: (MP); (MJB)
| | | | - Michael J. Bevan
- Department of Immunology and Howard Hughes Medical Institute, University of Washington, Seattle, Washington, United States of America
- * E-mail: (MP); (MJB)
| |
Collapse
|
32
|
Verbist KC, Field MB, Klonowski KD. Cutting edge: IL-15-independent maintenance of mucosally generated memory CD8 T cells. THE JOURNAL OF IMMUNOLOGY 2011; 186:6667-71. [PMID: 21572025 DOI: 10.4049/jimmunol.1004022] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Effective vaccines against intracellular pathogens rely on the generation and maintenance of memory CD8 T cells (T(mem)). Hitherto, evidence has indicated that CD8 T(mem) use the common γ-chain cytokine IL-15 for their steady-state maintenance in the absence of Ag. This evidence, however, has been amassed predominantly from models of acute, systemic infections. Given that the route of infection can have significant impact on the quantity and quality of the resultant T(mem), reliance on limited models of infection may restrict our understanding of long-term CD8 T(mem) survival. In this article, we show IL-15-independent generation, maintenance, and function of CD8 T(mem) after respiratory infection with influenza virus. Importantly, we demonstrate that alternating between mucosal and systemic deliveries of the identical virus prompts this change in IL-15 dependence, necessitating a re-evaluation of the current model of CD8 T(mem) maintenance.
Collapse
Affiliation(s)
- Katherine C Verbist
- Department of Cellular Biology, University of Georgia, Athens, GA 30602, USA
| | | | | |
Collapse
|