1
|
Lam N, Lee Y, Farber DL. A guide to adaptive immune memory. Nat Rev Immunol 2024; 24:810-829. [PMID: 38831162 DOI: 10.1038/s41577-024-01040-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/26/2024] [Indexed: 06/05/2024]
Abstract
Immune memory - comprising T cells, B cells and plasma cells and their secreted antibodies - is crucial for human survival. It enables the rapid and effective clearance of a pathogen after re-exposure, to minimize damage to the host. When antigen-experienced, memory T cells become activated, they proliferate and produce effector molecules at faster rates and in greater magnitudes than antigen-inexperienced, naive cells. Similarly, memory B cells become activated and differentiate into antibody-secreting cells more rapidly than naive B cells, and they undergo processes that increase their affinity for antigen. The ability of T cells and B cells to form memory cells after antigen exposure is the rationale behind vaccination. Understanding immune memory not only is crucial for the design of more-efficacious vaccines but also has important implications for immunotherapies in infectious disease and cancer. This 'guide to' article provides an overview of the current understanding of the phenotype, function, location, and pathways for the generation, maintenance and protective capacity of memory T cells and memory B cells.
Collapse
Affiliation(s)
- Nora Lam
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - YoonSeung Lee
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY, USA
| | - Donna L Farber
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY, USA.
- Department of Surgery, Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|
2
|
Lucas ED, Huggins MA, Peng C, O'Connor C, Gress AR, Thefaine CE, Dehm EM, Kubota Y, Jameson SC, Hamilton SE. Circulating KLRG1 + long-lived effector memory T cells retain the flexibility to become tissue resident. Sci Immunol 2024; 9:eadj8356. [PMID: 38941479 DOI: 10.1126/sciimmunol.adj8356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 05/30/2024] [Indexed: 06/30/2024]
Abstract
KLRG1+ CD8 T cells persist for months after clearance of acute infections and maintain high levels of effector molecules, contributing protective immunity against systemic pathogens. Upon secondary infection, these long-lived effector cells (LLECs) are incapable of forming other circulating KLRG1- memory subsets such as central and effector memory T cells. Thus, KLRG1+ memory T cells are frequently referred to as a terminally differentiated population that is relatively short lived. Here, we show that after viral infection of mice, effector cells derived from LLECs rapidly enter nonlymphoid tissues and reduce pathogen burden but are largely dependent on receiving antigen cues from vascular endothelial cells. Single-cell RNA sequencing reveals that secondary memory cells in nonlymphoid tissues arising from either KLRG1+ or KLRG1- memory precursors develop a similar resident memory transcriptional signature. Thus, although LLECs cannot differentiate into other circulating memory populations, they still retain the flexibility to enter tissues and establish residency.
Collapse
Affiliation(s)
- Erin D Lucas
- Center for Immunology, Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Matthew A Huggins
- Center for Immunology, Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Changwei Peng
- Center for Immunology, Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Christine O'Connor
- Minnesota Supercomputing Institute, University of Minnesota, Saint Paul, MN 55108, USA
| | - Abigail R Gress
- Center for Immunology, Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Claire E Thefaine
- Center for Immunology, Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Emma M Dehm
- Center for Immunology, Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA
| | | | - Stephen C Jameson
- Center for Immunology, Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Sara E Hamilton
- Center for Immunology, Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
3
|
Wang S, Prieux M, de Bernard S, Dubois M, Laubreton D, Djebali S, Zala M, Arpin C, Genestier L, Leverrier Y, Gandrillon O, Crauste F, Jiang W, Marvel J. Exogenous IL-2 delays memory precursors generation and is essential for enhancing memory cells effector functions. iScience 2024; 27:109411. [PMID: 38510150 PMCID: PMC10952031 DOI: 10.1016/j.isci.2024.109411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 11/27/2023] [Accepted: 02/29/2024] [Indexed: 03/22/2024] Open
Abstract
To investigate the impact of paracrine IL-2 signals on memory precursor (MP) cell differentiation, we activated CD8 T cell in vitro in the presence or absence of exogenous IL-2 (ex-IL-2). We assessed memory differentiation by transferring these cells into virus-infected mice. Both conditions generated CD8 T cells that participate in the ongoing response and gave rise to similar memory cells. Nevertheless, when transferred into a naive host, T cells activated with ex-IL-2 generated a higher frequency of memory cells displaying increased functional memory traits. Single-cell RNA-seq analysis indicated that without ex-IL-2, cells rapidly acquire an MP signature, while in its presence they adopted an effector signature. This was confirmed at the protein level and in a functional assay. Overall, ex-IL-2 delays the transition into MP cells, allowing the acquisition of effector functions that become imprinted in their progeny. These findings may help to optimize the generation of therapeutic T cells.
Collapse
Affiliation(s)
- Shaoying Wang
- Centre International de Recherche en Infectiologie, INSERM, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, École Normale Supérieure de Lyon, Université de Lyon, Lyon, France
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Margaux Prieux
- Centre International de Recherche en Infectiologie, INSERM, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, École Normale Supérieure de Lyon, Université de Lyon, Lyon, France
- Laboratoire de Biologie et de Modélisation de la Cellule, Université de Lyon, ENS de Lyon, CNRS UMR 5239, INSERM U1210, Lyon, France
| | | | - Maxence Dubois
- Centre International de Recherche en Infectiologie, INSERM, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, École Normale Supérieure de Lyon, Université de Lyon, Lyon, France
| | - Daphne Laubreton
- Centre International de Recherche en Infectiologie, INSERM, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, École Normale Supérieure de Lyon, Université de Lyon, Lyon, France
| | - Sophia Djebali
- Centre International de Recherche en Infectiologie, INSERM, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, École Normale Supérieure de Lyon, Université de Lyon, Lyon, France
| | - Manon Zala
- Centre International de Recherche en Infectiologie, INSERM, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, École Normale Supérieure de Lyon, Université de Lyon, Lyon, France
- Faculté de Médecine Lyon-Sud, Université de Lyon, Oullins, France
| | - Christophe Arpin
- Centre International de Recherche en Infectiologie, INSERM, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, École Normale Supérieure de Lyon, Université de Lyon, Lyon, France
- Laboratoire de Biologie et de Modélisation de la Cellule, Université de Lyon, ENS de Lyon, CNRS UMR 5239, INSERM U1210, Lyon, France
| | - Laurent Genestier
- Centre International de Recherche en Infectiologie, INSERM, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, École Normale Supérieure de Lyon, Université de Lyon, Lyon, France
- Faculté de Médecine Lyon-Sud, Université de Lyon, Oullins, France
| | - Yann Leverrier
- Centre International de Recherche en Infectiologie, INSERM, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, École Normale Supérieure de Lyon, Université de Lyon, Lyon, France
| | - Olivier Gandrillon
- Inria, Villeurbanne, France
- Laboratoire de Biologie et de Modélisation de la Cellule, Université de Lyon, ENS de Lyon, CNRS UMR 5239, INSERM U1210, Lyon, France
| | - Fabien Crauste
- Laboratoire MAP5 (UMR CNRS 8145), Université Paris Cité, Paris, France
| | - Wenzheng Jiang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Jacqueline Marvel
- Centre International de Recherche en Infectiologie, INSERM, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, École Normale Supérieure de Lyon, Université de Lyon, Lyon, France
| |
Collapse
|
4
|
Abdullah L, Emiliani FE, Vaidya CM, Stuart H, Kolling FW, Ackerman ME, Song L, McKenna A, Huang YH. Hierarchal single-cell lineage tracing reveals differential fate commitment of CD8 T-cell clones in response to acute infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.21.586160. [PMID: 38585810 PMCID: PMC10996474 DOI: 10.1101/2024.03.21.586160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Generating balanced populations of CD8 effector and memory T cells is necessary for immediate and durable immunity to infections and cancer. Yet, a definitive understanding of CD8 differentiation remains unclear. We used CARLIN, a processive lineage recording mouse model with single-cell RNA-seq and TCR-seq to track endogenous antigen-specific CD8 T cells during acute viral infection. We identified a diverse repertoire of expanded T-cell clones represented by seven transcriptional states. TCR enrichment analysis revealed differential memory- or effector-fate biases within clonal populations. Shared Vb segments and amino acid motifs were found within biased categories despite high TCR diversity. Using single-cell CARLIN barcode-seq we tracked multi-generational clones and found that unlike unbiased or memory-biased clones, which stably retain their fate profiles, effector-biased clones could adopt memory- or effector-bias within subclones. Collectively, our study demonstrates that a heterogenous T-cell repertoire specific for a shared antigen is composed of clones with distinct TCR-intrinsic fate-biases.
Collapse
Affiliation(s)
- Leena Abdullah
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
| | - Francesco E. Emiliani
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
| | - Chinmay M. Vaidya
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, USA
| | - Hannah Stuart
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
| | | | - Margaret E. Ackerman
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, USA
- Dartmouth Cancer Center, Lebanon, NH 03756, USA
| | - Li Song
- Department of Biomedical Data Science, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
- Dartmouth Cancer Center, Lebanon, NH 03756, USA
| | - Aaron McKenna
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
- Dartmouth Cancer Center, Lebanon, NH 03756, USA
| | - Yina H. Huang
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
- Dartmouth Cancer Center, Lebanon, NH 03756, USA
- Department of Pathology and Laboratory Medicine, Dartmouth Health, Lebanon, NH 03756, USA
| |
Collapse
|
5
|
Jiang Y, Lai X, Liu Y, Yang C, Liu Z, Liu X, Yu T, Chen C, Khanniche A, Fan J, Lin Y, Zeng W. CD8 + T cells in fetal membranes display a unique phenotype, and their activation is involved in the pathophysiology of spontaneous preterm birth. J Pathol 2024; 262:240-253. [PMID: 38018407 DOI: 10.1002/path.6229] [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: 04/04/2023] [Revised: 08/16/2023] [Accepted: 10/11/2023] [Indexed: 11/30/2023]
Abstract
Preterm labor/birth is the leading cause of perinatal mortality and morbidity worldwide. Previous studies demonstrated that T cells were crucial for maintaining maternal-fetal immune tolerance during the first trimester of pregnancy; however, their phenotypes and functions in labor and delivery remain largely unknown. We recruited three cohorts of women at delivery for T-cell immunophenotyping in the placentas, fetal membranes, umbilical cord blood, and maternal peripheral blood. Our data showed a differential enrichment of T cells during the third trimester of human pregnancy, with CD4+ T cells being more observable within the umbilical cord blood, whereas CD8+ T cells became relatively more abundant in fetal membranes. CD4+ and CD8+ T cells derived from fetal membranes were dominated by effector memory T cells and exhibited extensive expression of activation markers but decreased expression of homing receptor. In comparison with term births, fetal membrane CD8+ T cells, especially the central memory subset, were significantly increased in frequency and showed more profound activation in spontaneous preterm birth patients. Finally, using an allogeneic mouse model, we found that T-cell-activation-induced preterm birth could be alleviated by the depletion of CD8+ T but not CD4+ T cells in vivo. Collectively, we showed that CD8+ T cells in fetal membranes displayed a unique phenotype, and their activation was involved in the pathophysiology of spontaneous preterm birth, which provides novel insights into the immune mechanisms of preterm birth and potential targets for the prevention of this syndrome. © 2023 The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Yinan Jiang
- Shanghai Key Laboratory of Embryo Original Diseases, The International Peace Maternity & Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Xintong Lai
- Shanghai Key Laboratory of Embryo Original Diseases, The International Peace Maternity & Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Yuxu Liu
- Shanghai Key Laboratory of Embryo Original Diseases, The International Peace Maternity & Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Cheng Yang
- Department of Infectious Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, PR China
| | - Zhicui Liu
- Department of Dermatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, PR China
| | - Xiaorui Liu
- Shanghai Key Laboratory of Embryo Original Diseases, The International Peace Maternity & Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Tiantian Yu
- Shanghai Key Laboratory of Embryo Original Diseases, The International Peace Maternity & Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Cailian Chen
- Department of Automation, Shanghai Jiao Tong University, Key Laboratory of System Control and Information Processing, Ministry of Education of China, Shanghai, PR China
| | - Asma Khanniche
- Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Jianxia Fan
- Shanghai Key Laboratory of Embryo Original Diseases, The International Peace Maternity & Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Yi Lin
- Reproductive Medicine Center, Shanghai Jiao Tong University School of Medicine Affiliated Sixth People's Hospital, Shanghai, PR China
| | - Weihong Zeng
- Shanghai Key Laboratory of Embryo Original Diseases, The International Peace Maternity & Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| |
Collapse
|
6
|
van der Heide V, Davenport B, Cubitt B, Roudko V, Choo D, Humblin E, Jhun K, Angeliadis K, Dawson T, Furtado G, Kamphorst A, Ahmed R, de la Torre JC, Homann D. Functional impairment of "helpless" CD8 + memory T cells is transient and driven by prolonged but finite cognate antigen presentation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.22.576725. [PMID: 38328184 PMCID: PMC10849538 DOI: 10.1101/2024.01.22.576725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
Generation of functional CD8 + T cell memory typically requires engagement of CD4 + T cells. However, in certain scenarios, such as acutely-resolving viral infections, effector (T E ) and subsequent memory (T M ) CD8 + T cell formation appear impervious to a lack of CD4 + T cell help during priming. Nonetheless, such "helpless" CD8 + T M respond poorly to pathogen rechallenge. At present, the origin and long-term evolution of helpless CD8 + T cell memory remain incompletely understood. Here, we demonstrate that helpless CD8 + T E differentiation is largely normal but a multiplicity of helpless CD8 T M defects, consistent with impaired memory maturation, emerge as a consequence of prolonged yet finite exposure to cognate antigen. Importantly, these defects resolve over time leading to full restoration of CD8 + T M potential and recall capacity. Our findings provide a unified explanation for helpless CD8 + T cell memory and emphasize an unexpected CD8 + T M plasticity with implications for vaccination strategies and beyond.
Collapse
|
7
|
DeGolier KR, Danis E, D'Antonio M, Cimons J, Yarnell M, Kedl RM, Kohler ME, Scott-Browne JP, Fry TJ. Antigen experience history directs distinct functional states of CD8+ CAR T cells during the anti-leukemia response. RESEARCH SQUARE 2023:rs.3.rs-3712137. [PMID: 38196657 PMCID: PMC10775394 DOI: 10.21203/rs.3.rs-3712137/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2024]
Abstract
Chimeric antigen receptor T cells are an effective therapy for B-lineage malignancies. However, many patients relapse and this therapeutic has yet to show strong efficacy in other hematologic or solid tumors. One opportunity for improvement lies in the ability to generate T cells with desirable functional characteristics. Here, we dissect the biology of CD8+ CAR T cells (CAR8) by controlling whether the T cell has encountered cognate TCR antigen prior to CAR generation. We find that prior antigen experience influences multiple aspects of in vitro and in vivo CAR8 functionality, resulting in superior effector function and leukemia clearance in the setting of limiting target antigen density compared to antigen-inexperienced T cells. However, this comes at the expense of inferior proliferative capacity, susceptibility to phenotypic exhaustion and dysfunction, and inability to clear wildtype leukemia in the setting of limiting CAR+ cell dose. Epigenomic and transcriptomic comparisons of these cell populations identified overexpression of the Runx2 transcription factor as a novel strategy to enhance CAR8 function, with a differential impact depending on prior cell state. Collectively, our data demonstrate that prior antigen experience determines functional attributes of a CAR T cell, as well as amenability to functional enhancement by transcription factor modulation.
Collapse
Affiliation(s)
- Kole R DeGolier
- Department of Immunology, University of Colorado Anschutz Medical Campus; Aurora, CO, USA
- Department of Pediatrics, University of Colorado Anschutz Medical Campus; Aurora, CO, USA
| | - Etienne Danis
- Biostatistics and Bioinformatics Shared Resource, University of Colorado Cancer Center, University of Colorado Anschutz Medical Campus; Aurora, CO, USA
| | - Marc D'Antonio
- Department of Immunology, University of Colorado Anschutz Medical Campus; Aurora, CO, USA
| | - Jennifer Cimons
- Department of Immunology, University of Colorado Anschutz Medical Campus; Aurora, CO, USA
- Department of Pediatrics, University of Colorado Anschutz Medical Campus; Aurora, CO, USA
| | - Michael Yarnell
- Department of Pediatrics, University of Colorado Anschutz Medical Campus; Aurora, CO, USA
- Center for Cancer and Blood Disorders, Children's Hospital Colorado; Aurora, CO, USA
| | - Ross M Kedl
- Department of Immunology, University of Colorado Anschutz Medical Campus; Aurora, CO, USA
| | - M Eric Kohler
- Department of Immunology, University of Colorado Anschutz Medical Campus; Aurora, CO, USA
- Department of Pediatrics, University of Colorado Anschutz Medical Campus; Aurora, CO, USA
- Center for Cancer and Blood Disorders, Children's Hospital Colorado; Aurora, CO, USA
| | - James P Scott-Browne
- Department of Immunology, University of Colorado Anschutz Medical Campus; Aurora, CO, USA
- Department of Immunology and Genomic Medicine, National Jewish Health, Denver, CO, USA
| | - Terry J Fry
- Department of Immunology, University of Colorado Anschutz Medical Campus; Aurora, CO, USA
- Department of Pediatrics, University of Colorado Anschutz Medical Campus; Aurora, CO, USA
- Center for Cancer and Blood Disorders, Children's Hospital Colorado; Aurora, CO, USA
| |
Collapse
|
8
|
Tremain AC, Wallace RP, Lorentz KM, Thornley TB, Antane JT, Raczy MR, Reda JW, Alpar AT, Slezak AJ, Watkins EA, Maulloo CD, Budina E, Solanki A, Nguyen M, Bischoff DJ, Harrington JL, Mishra R, Conley GP, Marlin R, Dereuddre-Bosquet N, Gallouët AS, LeGrand R, Wilson DS, Kontos S, Hubbell JA. Synthetically glycosylated antigens for the antigen-specific suppression of established immune responses. Nat Biomed Eng 2023; 7:1142-1155. [PMID: 37679570 DOI: 10.1038/s41551-023-01086-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 08/02/2023] [Indexed: 09/09/2023]
Abstract
Inducing antigen-specific tolerance during an established immune response typically requires non-specific immunosuppressive signalling molecules. Hence, standard treatments for autoimmunity trigger global immunosuppression. Here we show that established antigen-specific responses in effector T cells and memory T cells can be suppressed by a polymer glycosylated with N-acetylgalactosamine (pGal) and conjugated to the antigen via a self-immolative linker that allows for the dissociation of the antigen on endocytosis and its presentation in the immunoregulatory environment. We show that pGal-antigen therapy induces antigen-specific tolerance in a mouse model of experimental autoimmune encephalomyelitis (with programmed cell-death-1 and the co-inhibitory ligand CD276 driving the tolerogenic responses), as well as the suppression of antigen-specific responses to vaccination against a DNA-based simian immunodeficiency virus in non-human primates. Our findings show that pGal-antigen therapy invokes mechanisms of immune tolerance to resolve antigen-specific inflammatory T-cell responses and suggest that the therapy may be applicable across autoimmune diseases.
Collapse
Affiliation(s)
- Andrew C Tremain
- Committee on Immunology, University of Chicago, Chicago, IL, USA
| | - Rachel P Wallace
- Pritzker School for Molecular Engineering, University of Chicago, Chicago, IL, USA
| | | | | | - Jennifer T Antane
- Pritzker School for Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Michal R Raczy
- Pritzker School for Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Joseph W Reda
- Pritzker School for Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Aaron T Alpar
- Pritzker School for Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Anna J Slezak
- Pritzker School for Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Elyse A Watkins
- Pritzker School for Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Chitavi D Maulloo
- Pritzker School for Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Erica Budina
- Pritzker School for Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Ani Solanki
- Animal Resources Center, University of Chicago, Chicago, IL, USA
| | - Mindy Nguyen
- Animal Resources Center, University of Chicago, Chicago, IL, USA
| | | | | | | | | | - Romain Marlin
- Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Université Paris-Saclay, INSERM, CEA, Fontenay-aux-Roses, France
| | - Nathalie Dereuddre-Bosquet
- Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Université Paris-Saclay, INSERM, CEA, Fontenay-aux-Roses, France
| | - Anne-Sophie Gallouët
- Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Université Paris-Saclay, INSERM, CEA, Fontenay-aux-Roses, France
| | - Roger LeGrand
- Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Université Paris-Saclay, INSERM, CEA, Fontenay-aux-Roses, France
| | - D Scott Wilson
- Pritzker School for Molecular Engineering, University of Chicago, Chicago, IL, USA.
- Biomedical Engineering Department, Johns Hopkins University, Baltimore, MD, USA.
| | | | - Jeffrey A Hubbell
- Committee on Immunology, University of Chicago, Chicago, IL, USA.
- Pritzker School for Molecular Engineering, University of Chicago, Chicago, IL, USA.
- Committee on Cancer Biology, University of Chicago, Chicago, IL, USA.
| |
Collapse
|
9
|
Louis TL, Wong WH, Yao P, Kurd NS, Tysl T, Indralingam CS, Ma S, Huang WJM, Chang JT. Regulation of CD8 T Cell Differentiation by the RNA-Binding Protein DDX5. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:241-251. [PMID: 37265401 PMCID: PMC10373580 DOI: 10.4049/jimmunol.2200778] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 05/05/2023] [Indexed: 06/03/2023]
Abstract
The RNA-binding protein DEAD-box protein 5 (DDX5) is a polyfunctional regulator of gene expression, but its role in CD8+ T cell biology has not been extensively investigated. In this study, we demonstrate that deletion of DDX5 in murine CD8+ T cells reduced the differentiation of terminal effector, effector memory T, and terminal effector memory cells while increasing the generation of central memory T cells, whereas forced expression of DDX5 elicited the opposite phenotype. DDX5-deficient CD8+ T cells exhibited increased expression of genes that promote central memory T cell differentiation, including Tcf7 and Eomes. Taken together, these findings reveal a role for DDX5 in regulating the differentiation of effector and memory CD8+ T cell subsets in response to microbial infection.
Collapse
Affiliation(s)
- Tiani L. Louis
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - William H. Wong
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Priscilla Yao
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Nadia S. Kurd
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Tiffani Tysl
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | | | - Shengyun Ma
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
| | - Wendy Jia Men Huang
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
| | - John T. Chang
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
- Department of Medicine, Jennifer Moreno Department of Veteran Affairs Medical Center, San Diego, CA, USA
| |
Collapse
|
10
|
Wu JE, Manne S, Ngiow SF, Baxter AE, Huang H, Freilich E, Clark ML, Lee JH, Chen Z, Khan O, Staupe RP, Huang YJ, Shi J, Giles JR, Wherry EJ. In Vitro Modeling of CD8 T Cell Exhaustion Enables CRISPR Screening to Reveal a Role for BHLHE40. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.17.537229. [PMID: 37131713 PMCID: PMC10153201 DOI: 10.1101/2023.04.17.537229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Identifying novel molecular mechanisms of exhausted CD8 T cells (T ex ) is a key goal of improving immunotherapy of cancer and other diseases. However, high-throughput interrogation of in vivo T ex can be costly and inefficient. In vitro models of T ex are easily customizable and quickly generate high cellular yield, offering an opportunity to perform CRISPR screening and other high-throughput assays. We established an in vitro model of chronic stimulation and benchmarked key phenotypic, functional, transcriptional, and epigenetic features against bona fide in vivo T ex . We leveraged this model of in vitro chronic stimulation in combination with pooled CRISPR screening to uncover transcriptional regulators of T cell exhaustion. This approach identified several transcription factors, including BHLHE40. In vitro and in vivo validation defined a role for BHLHE40 in regulating a key differentiation checkpoint between progenitor and intermediate subsets of T ex . By developing and benchmarking an in vitro model of T ex , we demonstrate the utility of mechanistically annotated in vitro models of T ex , in combination with high-throughput approaches, as a discovery pipeline to uncover novel T ex biology.
Collapse
Affiliation(s)
- Jennifer E. Wu
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania; Philadelphia, PA, USA
- Institute for Immunology and Immune Health, Perelman School of Medicine, University of Pennsylvania; Philadelphia, PA, USA
- Parker Institute for Cancer Immunotherapy at University of Pennsylvania; Philadelphia, PA, USA
| | - Sasikanth Manne
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania; Philadelphia, PA, USA
- Institute for Immunology and Immune Health, Perelman School of Medicine, University of Pennsylvania; Philadelphia, PA, USA
| | - Shin Foong Ngiow
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania; Philadelphia, PA, USA
- Institute for Immunology and Immune Health, Perelman School of Medicine, University of Pennsylvania; Philadelphia, PA, USA
- Parker Institute for Cancer Immunotherapy at University of Pennsylvania; Philadelphia, PA, USA
| | - Amy E. Baxter
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania; Philadelphia, PA, USA
- Institute for Immunology and Immune Health, Perelman School of Medicine, University of Pennsylvania; Philadelphia, PA, USA
| | - Hua Huang
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania; Philadelphia, PA, USA
- Institute for Immunology and Immune Health, Perelman School of Medicine, University of Pennsylvania; Philadelphia, PA, USA
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania; Philadelphia, PA, USA
| | - Elizabeth Freilich
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania; Philadelphia, PA, USA
- Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania; Philadelphia, PA, USA
| | - Megan L. Clark
- Institute for Immunology and Immune Health, Perelman School of Medicine, University of Pennsylvania; Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Joanna H. Lee
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania; Philadelphia, PA, USA
- Institute for Immunology and Immune Health, Perelman School of Medicine, University of Pennsylvania; Philadelphia, PA, USA
| | - Zeyu Chen
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania; Philadelphia, PA, USA
- Institute for Immunology and Immune Health, Perelman School of Medicine, University of Pennsylvania; Philadelphia, PA, USA
- Present Address: Department of Cancer Biology, Dana-Farber Cancer Institute, Harvard Medical School; Boston, MA, USA
| | - Omar Khan
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania; Philadelphia, PA, USA
- Institute for Immunology and Immune Health, Perelman School of Medicine, University of Pennsylvania; Philadelphia, PA, USA
- Present Address: Department of Laboratory Medicine, University of California, San Francisco; San Francisco, CA, USA
| | - Ryan P. Staupe
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania; Philadelphia, PA, USA
- Institute for Immunology and Immune Health, Perelman School of Medicine, University of Pennsylvania; Philadelphia, PA, USA
- Present Address: Infectious Diseases and Vaccines, MRL, Merck & Co., Inc, West Point, PA, USA
| | - Yinghui J. Huang
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania; Philadelphia, PA, USA
- Institute for Immunology and Immune Health, Perelman School of Medicine, University of Pennsylvania; Philadelphia, PA, USA
| | - Junwei Shi
- Institute for Immunology and Immune Health, Perelman School of Medicine, University of Pennsylvania; Philadelphia, PA, USA
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania; Philadelphia, PA, USA
- Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania; Philadelphia, PA, USA
| | - Josephine R. Giles
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania; Philadelphia, PA, USA
- Institute for Immunology and Immune Health, Perelman School of Medicine, University of Pennsylvania; Philadelphia, PA, USA
- Parker Institute for Cancer Immunotherapy at University of Pennsylvania; Philadelphia, PA, USA
| | - E. John Wherry
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania; Philadelphia, PA, USA
- Institute for Immunology and Immune Health, Perelman School of Medicine, University of Pennsylvania; Philadelphia, PA, USA
- Parker Institute for Cancer Immunotherapy at University of Pennsylvania; Philadelphia, PA, USA
| |
Collapse
|
11
|
Osum KC, Jenkins MK. Toward a general model of CD4 + T cell subset specification and memory cell formation. Immunity 2023; 56:475-484. [PMID: 36921574 PMCID: PMC10084496 DOI: 10.1016/j.immuni.2023.02.010] [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: 12/27/2022] [Accepted: 02/16/2023] [Indexed: 03/17/2023]
Abstract
In the past few decades, a number of transformative discoveries have been made regarding memory CD8+ T cell biology; meanwhile, the CD4+ T cell field has lagged behind this progress. This perspective focuses on CD4+ helper T (Th) cell subset specification and memory cell formation. Here, we argue that the sheer number of Th effector and memory cell subsets and a focus on their differences have been a barrier to a general model of CD4+ memory T cell formation that applies to all immune responses. We highlight a bifurcation model that relies on an IL-2 signal-dependent switch as an explanation for the balanced production of diverse Th memory cells that participate in cell-mediated or humoral immunity in most contexts.
Collapse
Affiliation(s)
- Kevin C Osum
- Department of Microbiology and Immunology, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Marc K Jenkins
- Department of Microbiology and Immunology, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455, USA.
| |
Collapse
|
12
|
Heidarian M, Griffith TS, Badovinac VP. Sepsis-induced changes in differentiation, maintenance, and function of memory CD8 T cell subsets. Front Immunol 2023; 14:1130009. [PMID: 36756117 PMCID: PMC9899844 DOI: 10.3389/fimmu.2023.1130009] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 01/09/2023] [Indexed: 01/24/2023] Open
Abstract
Formation of long-lasting memory lymphocytes is one of the foundational characteristics of adaptive immunity and the basis of many vaccination strategies. Following the rapid expansion and contraction of effector CD8 T cells, the surviving antigen (Ag)-specific cells give rise to the memory CD8 T cells that persist for a long time and are phenotypically and functionally distinct from their naïve counterparts. Significant heterogeneity exists within the memory CD8 T cell pool, as different subsets display distinct tissue localization preferences, cytotoxic ability, and proliferative capacity, but all memory CD8 T cells are equipped to mount an enhanced immune response upon Ag re-encounter. Memory CD8 T cells demonstrate numerical stability under homeostatic conditions, but sepsis causes a significant decline in the number of memory CD8 T cells and diminishes their Ag-dependent and -independent functions. Sepsis also rewires the transcriptional profile of memory CD8 T cells, which profoundly impacts memory CD8 T cell differentiation and, ultimately, the protective capacity of memory CD8 T cells upon subsequent stimulation. This review delves into different aspects of memory CD8 T cell subsets as well as the immediate and long-term impact of sepsis on memory CD8 T cell biology.
Collapse
Affiliation(s)
| | - Thomas S. Griffith
- Department of Urology, University of Minnesota, Minneapolis, MN, United States,Minneapolis Veterans Affairs Health Care System, Minneapolis, MN, United States
| | - Vladimir P. Badovinac
- Department of Pathology, University of Iowa, Iowa, IA, United States,Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa, IA, United States,*Correspondence: Vladimir P. Badovinac,
| |
Collapse
|
13
|
Collard MK, Tourneur-Marsille J, Uzzan M, Albuquerque M, Roy M, Dumay A, Freund JN, Hugot JP, Guedj N, Treton X, Panis Y, Ogier-Denis E. The Appendix Orchestrates T-Cell Mediated Immunosurveillance in Colitis-Associated Cancer. Cell Mol Gastroenterol Hepatol 2023; 15:665-687. [PMID: 36332814 PMCID: PMC9871441 DOI: 10.1016/j.jcmgh.2022.10.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 10/11/2022] [Accepted: 10/20/2022] [Indexed: 11/06/2022]
Abstract
BACKGROUND & AIMS Although appendectomy may reduce colorectal inflammation in patients with ulcerative colitis (UC), this surgical procedure has been suggested to be associated with an increased risk of colitis-associated cancer (CAC). Our aim was to explore the mechanism underlying the appendectomy-associated increased risk of CAC. METHODS Five-week-old male BALB/c mice underwent appendectomy, appendicitis induction, or sham laparotomy. They were then exposed to azoxymethane/dextran sodium sulfate (AOM/DSS) to induce CAC. Mice were killed 12 weeks later, and colons were taken for pathological analysis and immunohistochemistry (CD3 and CD8 staining). Human colonic tumors from 21 patients with UC who underwent surgical resection for CAC were immunophenotyped and stratified according to appendectomy status. RESULTS Whereas appendectomy significantly reduced colitis severity and increased CAC number, appendicitis induction without appendectomy led to opposite results. Intratumor CD3+ and CD8+ T-cell densities were lower after appendectomy and higher after appendicitis induction compared with the sham laparotomy group. Blocking lymphocyte trafficking to the colon with the anti-α4β7 integrin antibody or a sphingosine-1-phosphate receptor agonist suppressed the inducing effect of the appendectomy on tumors' number and on CD3+/CD8+ intratumoral density. CD8+ or CD3+ T cells isolated from inflammatory neo-appendix and intravenously injected into AOM/DSS-treated recipient mice increased CD3+/CD8+ T-cell tumor infiltration and decreased tumor number. In UC patients with a history of appendectomy, intratumor CD3+ and CD8+ T-cell densities were decreased compared with UC patients without history of appendectomy. CONCLUSIONS In UC, appendectomy could suppress a major site of T-cell priming, resulting in a less efficient CAC immunosurveillance.
Collapse
Affiliation(s)
- Maxime K Collard
- Assistance Publique Hôpitaux de Paris, Service de Chirurgie Colorectale, Hôpital Beaujon, Clichy, France; Université de Paris, Centre de Recherche sur l'Inflammation, INSERM, U1149, CNRS, ERL8252, "Gut Inflammation", Paris, France
| | - Julien Tourneur-Marsille
- Université de Paris, Centre de Recherche sur l'Inflammation, INSERM, U1149, CNRS, ERL8252, "Gut Inflammation", Paris, France
| | - Mathieu Uzzan
- Université de Paris, Centre de Recherche sur l'Inflammation, INSERM, U1149, CNRS, ERL8252, "Gut Inflammation", Paris, France; Assistance Publique Hôpitaux de Paris, Service de Gastroentérologie, Hôpital Beaujon, Clichy, France
| | - Miguel Albuquerque
- Assistance Publique Hôpitaux de Paris, Service d'Anatomopathologie, Hôpital Beaujon, Clichy, France
| | - Maryline Roy
- Université de Paris, Centre de Recherche sur l'Inflammation, INSERM, U1149, CNRS, ERL8252, "Gut Inflammation", Paris, France
| | - Anne Dumay
- Université de Paris, Centre de Recherche sur l'Inflammation, INSERM, U1149, CNRS, ERL8252, "Gut Inflammation", Paris, France
| | - Jean-Noël Freund
- Université de Strasbourg, Inserm, IRFAC / UMR-S1113, FHU ARRIMAGE, ITI InnoVec, FMTS, Strasbourg, France
| | - Jean-Pierre Hugot
- Université de Paris, Centre de Recherche sur l'Inflammation, INSERM, U1149, CNRS, ERL8252, "Gut Inflammation", Paris, France
| | - Nathalie Guedj
- Université de Paris, Centre de Recherche sur l'Inflammation, INSERM, U1149, CNRS, ERL8252, "Gut Inflammation", Paris, France; Assistance Publique Hôpitaux de Paris, Service d'Anatomopathologie, Hôpital Beaujon, Clichy, France
| | - Xavier Treton
- Université de Paris, Centre de Recherche sur l'Inflammation, INSERM, U1149, CNRS, ERL8252, "Gut Inflammation", Paris, France; Assistance Publique Hôpitaux de Paris, Service de Gastroentérologie, Hôpital Beaujon, Clichy, France
| | - Yves Panis
- Assistance Publique Hôpitaux de Paris, Service de Chirurgie Colorectale, Hôpital Beaujon, Clichy, France; Université de Paris, Centre de Recherche sur l'Inflammation, INSERM, U1149, CNRS, ERL8252, "Gut Inflammation", Paris, France
| | - Eric Ogier-Denis
- Université de Paris, Centre de Recherche sur l'Inflammation, INSERM, U1149, CNRS, ERL8252, "Gut Inflammation", Paris, France; INSERM, Université Rennes, CLCC Eugène Marquis, «Chemistry, Oncogenesis, Stress Signaling» UMR_S 1242, Rennes, France.
| |
Collapse
|
14
|
Respiratory mucosal vaccination of peptide-poloxamine-DNA nanoparticles provides complete protection against lethal SARS-CoV-2 challenge. Biomaterials 2023; 292:121907. [PMID: 36436305 PMCID: PMC9673044 DOI: 10.1016/j.biomaterials.2022.121907] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 10/28/2022] [Accepted: 11/06/2022] [Indexed: 11/21/2022]
Abstract
The ongoing SARS-CoV-2 pandemic represents a brutal reminder of the continual threat of mucosal infectious diseases. Mucosal immunity may provide robust protection at the predominant sites of SARS-CoV-2 infection. However, it remains unclear whether respiratory mucosal administration of DNA vaccines could confer protective immune responses against SARS-CoV-2 challenge due to insurmountable barriers posed by the airway. Here, we applied self-assembled peptide-poloxamine nanoparticles with mucus-penetrating properties for pulmonary inoculation of a COVID-19 DNA vaccine (pSpike/PP-sNp). The pSpike/PP-sNp not only displays superior gene transfection and favorable biocompatibility in the mouse airway, but also promotes a tripartite immunity consisting of systemic, cellular, and mucosal immune responses that are characterized by mucosal IgA secretion, high levels of neutralizing antibodies, and resident memory phenotype T-cell responses in the lungs of mice. Most importantly, immunization with pSpike/PP-sNp completely eliminates SARS-CoV-2 infection in both upper and lower respiratory tracts and enables 100% survival rate of mice following lethal SARS-CoV-2 challenge. Our findings indicate PP-sNp is a promising platform in mediating DNA vaccines to elicit all-around mucosal immunity against SARS-CoV-2.
Collapse
|
15
|
Sjaastad FV, Huggins MA, Lucas ED, Skon-Hegg C, Swanson W, Martin MD, Salgado OC, Xu J, Pierson M, Dileepan T, Kucaba TA, Hamilton SE, Griffith TS. Reduced T Cell Priming in Microbially Experienced "Dirty" Mice Results from Limited IL-27 Production by XCR1+ Dendritic Cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 209:2149-2159. [PMID: 36426978 PMCID: PMC10065988 DOI: 10.4049/jimmunol.2200324] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 09/28/2022] [Indexed: 01/04/2023]
Abstract
Successful vaccination strategies offer the potential for lifelong immunity against infectious diseases and cancer. There has been increased attention regarding the limited translation of some preclinical findings generated using specific pathogen-free (SPF) laboratory mice to humans. One potential reason for the difference between preclinical and clinical findings lies in maturation status of the immune system at the time of challenge. In this study, we used a "dirty" mouse model, where SPF laboratory mice were cohoused (CoH) with pet store mice to permit microbe transfer and immune system maturation, to investigate the priming of a naive T cell response after vaccination with a peptide subunit mixed with polyinosinic-polycytidylic acid and agonistic anti-CD40 mAb. Although this vaccination platform induced robust antitumor immunity in SPF mice, it failed to do so in microbially experienced CoH mice. Subsequent investigation revealed that despite similar numbers of Ag-specific naive CD4 and CD8 T cell precursors, the expansion, differentiation, and recall responses of these CD4 and CD8 T cell populations in CoH mice were significantly reduced compared with SPF mice after vaccination. Evaluation of the dendritic cell compartment revealed reduced IL-27p28 expression by XCR1+ dendritic cells from CoH mice after vaccination, correlating with reduced T cell expansion. Importantly, administration of recombinant IL-27:EBI3 complex to CoH mice shortly after vaccination significantly boosted Ag-specific CD8 and CD4 T cell expansion, further implicating the defect to be T cell extrinsic. Collectively, our data show the potential limitation of exclusive use of SPF mice when testing vaccine efficacy.
Collapse
Affiliation(s)
- Frances V Sjaastad
- Department of Urology, University of Minnesota, Minneapolis, MN
- Microbiology, Immunology, and Cancer Biology Ph.D. Program, University of Minnesota, Minneapolis, MN
| | - Matthew A Huggins
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN
| | - Erin D Lucas
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN
| | - Cara Skon-Hegg
- Department of Urology, University of Minnesota, Minneapolis, MN
| | - Whitney Swanson
- Department of Urology, University of Minnesota, Minneapolis, MN
| | | | - Oscar C Salgado
- Microbiology, Immunology, and Cancer Biology Ph.D. Program, University of Minnesota, Minneapolis, MN
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN
| | - Julie Xu
- Department of Urology, University of Minnesota, Minneapolis, MN
| | - Mark Pierson
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN
| | - Thamotharampillai Dileepan
- Center for Immunology, University of Minnesota, Minneapolis, MN
- Department of Microbiology & Immunology, University of Minnesota, Minneapolis, MN
| | - Tamara A Kucaba
- Department of Urology, University of Minnesota, Minneapolis, MN
| | - Sara E Hamilton
- Microbiology, Immunology, and Cancer Biology Ph.D. Program, University of Minnesota, Minneapolis, MN
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN
- Center for Immunology, University of Minnesota, Minneapolis, MN
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN; and
| | - Thomas S Griffith
- Department of Urology, University of Minnesota, Minneapolis, MN
- Microbiology, Immunology, and Cancer Biology Ph.D. Program, University of Minnesota, Minneapolis, MN
- Center for Immunology, University of Minnesota, Minneapolis, MN
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN; and
- Minneapolis VA Health Care System, Minneapolis, MN
| |
Collapse
|
16
|
CD8 + T cell metabolic rewiring defined by scRNA-seq identifies a critical role of ASNS expression dynamics in T cell differentiation. Cell Rep 2022; 41:111639. [PMID: 36384124 DOI: 10.1016/j.celrep.2022.111639] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 08/05/2022] [Accepted: 10/19/2022] [Indexed: 11/18/2022] Open
Abstract
T cells dynamically rewire their metabolism during an immune response. We applied single-cell RNA sequencing to CD8+ T cells activated and differentiated in vitro in physiological medium to resolve these metabolic dynamics. We identify a differential time-dependent reliance of activating T cells on the synthesis versus uptake of various non-essential amino acids, which we corroborate with functional assays. We also identify metabolic genes that potentially dictate the outcome of T cell differentiation, by ranking them based on their expression dynamics. Among them, we find asparagine synthetase (Asns), whose expression peaks for effector T cells and decays toward memory formation. Disrupting these expression dynamics by ASNS overexpression promotes an effector phenotype, enhancing the anti-tumor response of adoptively transferred CD8+ T cells in a mouse melanoma model. We thus provide a resource of dynamic expression changes during CD8+ T cell activation and differentiation, and identify ASNS expression dynamics as a modulator of CD8+ T cell differentiation.
Collapse
|
17
|
Naoun AA, Raphael I, Forsthuber TG. Immunoregulation via Cell Density and Quorum Sensing-like Mechanisms: An Underexplored Emerging Field with Potential Translational Implications. Cells 2022; 11:cells11152442. [PMID: 35954285 PMCID: PMC9368058 DOI: 10.3390/cells11152442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 07/27/2022] [Accepted: 08/01/2022] [Indexed: 11/16/2022] Open
Abstract
Quorum sensing (QS) was historically described as a mechanism by which bacteria detect and optimize their population density via gene regulation based on dynamic environmental cues. Recently, it was proposed that QS or similar mechanisms may have broader applications across different species and cell types. Indeed, emerging evidence shows that the mammalian immune system can also elicit coordinated responses on a population level to regulate cell density and function, thus suggesting that QS-like mechanisms may also be a beneficial trait of the immune system. In this review, we explore and discuss potential QS-like mechanisms deployed by the immune system to coordinate cellular-level responses, such as T cell responses mediated via the common gamma chain (γc) receptor cytokines and the aryl hydrocarbon receptors (AhRs). We present evidence regarding a novel role of QS as a multifunctional mechanism coordinating CD4+ and CD8+ T cell behavior during steady state and in response to infection, inflammatory diseases, and cancer. Successful clinical therapies such as adoptive cell transfer for cancer treatment may be re-evaluated to harness the effects of the QS mechanism(s) and enhance treatment responsiveness. Moreover, we discuss how signaling threshold perturbations through QS-like mediators may result in disturbances of the complex crosstalk between immune cell populations, undesired T cell responses, and induction of autoimmune pathology. Finally, we discuss the potential therapeutic role of modulating immune-system-related QS as a promising avenue to treat human diseases.
Collapse
Affiliation(s)
- Adrian A. Naoun
- Department of Molecular Microbiology and Immunology, University of Texas at San Antonio, San Antonio, TX 78249, USA
| | - Itay Raphael
- Department of Neurological Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15217, USA
- Correspondence: (I.R.); (T.G.F.)
| | - Thomas G. Forsthuber
- Department of Molecular Microbiology and Immunology, University of Texas at San Antonio, San Antonio, TX 78249, USA
- Correspondence: (I.R.); (T.G.F.)
| |
Collapse
|
18
|
Immune tolerance of food is mediated by layers of CD4 + T cell dysfunction. Nature 2022; 607:762-768. [PMID: 35794484 DOI: 10.1038/s41586-022-04916-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 05/27/2022] [Indexed: 11/08/2022]
Abstract
Gastrointestinal health depends on the adaptive immune system tolerating the foreign proteins in food1,2. This tolerance is paradoxical because the immune system normally attacks foreign substances by generating inflammation. Here we addressed this conundrum by using a sensitive cell enrichment method to show that polyclonal CD4+ T cells responded to food peptides, including a natural one from gliadin, by proliferating weakly in secondary lymphoid organs of the gut-liver axis owing to the action of regulatory T cells. A few food-specific T cells then differentiated into T follicular helper cells that promoted a weak antibody response. Most cells in the expanded population, however, lacked canonical T helper lineage markers and fell into five subsets dominated by naive-like or T follicular helper-like anergic cells with limited capacity to form inflammatory T helper 1 cells. Eventually, many of the T helper lineage-negative cells became regulatory T cells themselves through an interleukin-2-dependent mechanism. Our results indicate that exposure to food antigens causes cognate CD4+ naive T cells to form a complex set of noncanonical hyporesponsive T helper cell subsets that lack the inflammatory functions needed to cause gut pathology and yet have the potential to produce regulatory T cells that may suppress it.
Collapse
|
19
|
Breed ER, Vobořil M, Ashby KM, Martinez RJ, Qian L, Wang H, Salgado OC, O'Connor CH, Hogquist KA. Type 2 cytokines in the thymus activate Sirpα + dendritic cells to promote clonal deletion. Nat Immunol 2022; 23:1042-1051. [PMID: 35637352 PMCID: PMC10037932 DOI: 10.1038/s41590-022-01218-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 04/18/2022] [Indexed: 12/30/2022]
Abstract
The thymus contains a diversity of dendritic cells (DCs) that exist in defined locations and have different antigen-processing and -presenting features. This suggests that they play nonredundant roles in mediating thymocyte selection. In an effort to eliminate SIRPα+ classic DC2 subsets, we discovered that a substantial proportion expresses the surface lectin, CD301b, in the thymus. These cells resemble the CD301b+ type 2 immune response promoting DCs that are present in the skin-draining lymph nodes. Transcriptional and phenotypic comparison to other DC subsets in the thymus revealed that thymic CD301b+ cDCs represent an activated state that exhibits enhanced antigen processing and presentation. Furthermore, a CD301b+ cDC2 subset demonstrated a type 2 cytokine signature and required steady-state interleukin-4 receptor signaling. Selective ablation of CD301b+ cDC2 subsets impaired clonal deletion without affecting regulatory T cells (Treg cells). The T cell receptor α repertoire sequencing confirmed that a cDC2 subset promotes deletion of conventional T cells with minimal effect on Treg cell selection. Together, these findings suggest that cytokine-induced activation of DCs in the thymus substantially enforces central tolerance.
Collapse
Affiliation(s)
- Elise R Breed
- Department of Laboratory Medicine and Pathology, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Matouš Vobořil
- Department of Laboratory Medicine and Pathology, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Katherine M Ashby
- Department of Laboratory Medicine and Pathology, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Ryan J Martinez
- Department of Laboratory Medicine and Pathology, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Lily Qian
- Department of Laboratory Medicine and Pathology, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Haiguang Wang
- Department of Laboratory Medicine and Pathology, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Oscar C Salgado
- Department of Laboratory Medicine and Pathology, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Christine H O'Connor
- Research Informatics Solutions, Laboratory Medicine and Pathology Group, Minnesota Supercomputing Institute, Minneapolis, MN, USA
| | - Kristin A Hogquist
- Department of Laboratory Medicine and Pathology, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN, USA.
| |
Collapse
|
20
|
Rehm A, Wirges A, Hoser D, Fischer C, Herda S, Gerlach K, Sauer S, Willimsky G, Höpken UE. EBAG9 controls CD8+ T cell memory formation responding to tumor challenge in mice. JCI Insight 2022; 7:155534. [PMID: 35482418 PMCID: PMC9220939 DOI: 10.1172/jci.insight.155534] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 04/22/2022] [Indexed: 11/17/2022] Open
Abstract
Insight into processes that determine CD8+ T cell memory formation has been obtained from infection models. These models are biased toward an inflammatory milieu and often use high-avidity CD8+ T cells in adoptive-transfer procedures. It is unclear whether these conditions mimic the differentiation processes of an endogenous repertoire that proceed upon noninflammatory conditions prevailing in premalignant tumor lesions. We examined the role of cytolytic capacity on CD8+ T cell fate decisions when primed by tumor cells or by minor histocompatibility antigen–mismatched leukocytes. CD8+ memory commitment was analyzed in Ebag9-deficient mice that exhibited enhanced tumor cell lysis. This property endowed Ebag9–/– mice with extended control of Tcl-1 oncogene–induced chronic lymphocytic leukemia progression. In Ebag9–/– mice, an expanded memory population was obtained for anti-HY and anti–SV-40 T antigen–specific T cells, despite unchanged effector frequencies in the primary response. By comparing the single-cell transcriptomes of CD8+ T cells responding to tumor cell vaccination, we found differential distribution of subpopulations between Ebag9+/+ and Ebag9–/– T cells. In Ebag9–/– cells, these larger clusters contained genes encoding transcription factors regulating memory cell differentiation and anti-apoptotic gene functions. Our findings link EBAG9-controlled cytolytic activity and the commitment to the CD8+ memory lineage.
Collapse
Affiliation(s)
- Armin Rehm
- Department of Translational Tumorimmunology, Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | - Anthea Wirges
- Department of Translational Tumorimmunology, Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | - Dana Hoser
- Institute of Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Cornelius Fischer
- Scientific Infrastructure Department, Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | - Stefanie Herda
- Department of Translational Tumorimmunology, Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | - Kerstin Gerlach
- Department of Translational Tumorimmunology, Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | - Sascha Sauer
- Scientific Infrastructure Department, Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | - Gerald Willimsky
- Institute of Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Uta E Höpken
- Department of Microenvironmental Regulation of Autoimmunity and Cancer, Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| |
Collapse
|
21
|
Knop L, Spanier J, Larsen PK, Witte A, Bank U, Dunay IR, Kalinke U, Schüler T. IFNAR signaling in fibroblastic reticular cells can modulate CD8 + memory fate decision. Eur J Immunol 2022; 52:895-906. [PMID: 35365883 DOI: 10.1002/eji.202149760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 02/27/2022] [Accepted: 03/31/2022] [Indexed: 11/12/2022]
Abstract
CD8+ memory T cells (TM ) are crucial for the long-term protection from infections and cancer. Multiple cell types and cytokines are involved in the regulation of CD8+ T cell responses and subsequent TM formation. Besides their direct antiviral effects, type I interferons (IFN-α/β) modulate CD8+ T cell immunity via their action on several immune cell subsets. However, it is largely unclear how non-immune cells are involved in this multicellular network modulating CD8+ TM formation. Fibroblastic reticular cells (FRCs), form the three-dimensional scaffold of secondary lymphoid organs, express the IFN-α/β receptor (IFNAR) and modulate adaptive immune responses. However, it is unclear whether and how early IFNAR signals in lymph node (LN) FRCs affect CD8+ TM differentiation. Using peptide vaccination and viral infection, we studied CD8+ TM differentiation in mice with a FRC-specific IFNAR deletion (FRCΔIFNAR ). We show here that the differentiation of CD8+ TCR-transgenic T cells into central memory cells (TCM ) is enhanced in peptide-vaccinated FRCΔIFNAR mice. Conversely, vesicular stomatitis virus (VSV) infection of FRCΔIFNAR mice is associated with impaired TCM formation and the accumulation of VSV-specific double-positive (dp) CD127lo KLRG-1hi effector memory T cells. In summary, we provide evidence for a context-dependent contribution of FRC-specific IFNAR signaling to CD8+ TM differentiation. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Laura Knop
- Institute of Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University, Magdeburg, 39120, Germany
| | - Julia Spanier
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, Hannover, 30625, Germany
| | - Pia-Katharina Larsen
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, Hannover, 30625, Germany
| | - Amelie Witte
- Institute of Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University, Magdeburg, 39120, Germany
| | - Ute Bank
- Institute of Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University, Magdeburg, 39120, Germany
| | - Ildiko R Dunay
- Institute of Inflammation and Neurodegeneration, Medical Faculty, Otto-von-Guericke University, Magdeburg, 39120, Germany
| | - Ulrich Kalinke
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, Hannover, 30625, Germany
| | - Thomas Schüler
- Institute of Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University, Magdeburg, 39120, Germany
| |
Collapse
|
22
|
Schilling HL, Glehr G, Kapinsky M, Ahrens N, Riquelme P, Cordero L, Bitterer F, Schlitt HJ, Geissler EK, Haferkamp S, Hutchinson JA, Kronenberg K. Development of a Flow Cytometry Assay to Predict Immune Checkpoint Blockade-Related Complications. Front Immunol 2021; 12:765644. [PMID: 34868015 PMCID: PMC8637156 DOI: 10.3389/fimmu.2021.765644] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 10/27/2021] [Indexed: 12/17/2022] Open
Abstract
Treatment of advanced melanoma with combined immune checkpoint inhibitor (ICI) therapy is complicated in up to 50% of cases by immune-related adverse events (irAE) that commonly include hepatitis, colitis and skin reactions. We previously reported that pre-therapy expansion of cytomegalovirus (CMV)-reactive CD4+ effector memory T cells (TEM) predicts ICI-related hepatitis in a subset of patients with Stage IV melanoma given αPD-1 and αCTLA-4. Here, we develop and validate a 10-color flow cytometry panel for reliably quantifying CD4+ TEM cells and other biomarkers of irAE risk in peripheral blood samples. Compared to previous methods, our new panel performs equally well in measuring CD4+ TEM cells (agreement = 98%) and is superior in resolving CD4+ CD197+ CD45RA- central memory T cells (TCM) from CD4+ CD197+ CD45RA+ naive T cells (Tnaive). It also enables us to precisely quantify CD14+ monocytes (CV = 6.6%). Our new “monocyte and T cell” (MoT) assay predicts immune-related hepatitis with a positive predictive value (PPV) of 83% and negative predictive value (NPV) of 80%. Our essential improvements open the possibility of sharing our predictive methods with other clinical centers. Furthermore, condensing measurements of monocyte and memory T cell subsets into a single assay simplifies our workflows and facilitates computational analyses.
Collapse
Affiliation(s)
| | - Gunther Glehr
- Institute of Functional Genomics and Statistical Bioinformatics, University of Regensburg, Regensburg, Germany
| | | | - Norbert Ahrens
- Medizinisches Versorgungszentrum (MVZ) for Laboratory Medicine Raubling, amedes Labor, Raubling, Germany.,Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Regensburg, Germany
| | - Paloma Riquelme
- Department of Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Laura Cordero
- Department of Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Florian Bitterer
- Department of Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Hans J Schlitt
- Department of Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Edward K Geissler
- Department of Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Sebastian Haferkamp
- Department of Dermatology, University Hospital Regensburg, Regensburg, Germany
| | - James A Hutchinson
- Department of Surgery, University Hospital Regensburg, Regensburg, Germany
| | | |
Collapse
|
23
|
Kalia V, Yuzefpolskiy Y, Vegaraju A, Xiao H, Baumann F, Jatav S, Church C, Prlic M, Jha A, Nghiem P, Riddell S, Sarkar S. Metabolic regulation by PD-1 signaling promotes long-lived quiescent CD8 T cell memory in mice. Sci Transl Med 2021; 13:eaba6006. [PMID: 34644150 DOI: 10.1126/scitranslmed.aba6006] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Vandana Kalia
- Division of Hematology and Oncology, Department of Pediatrics, University of Washington School of Medicine, Seattle, WA 98195, USA.,Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Yevgeniy Yuzefpolskiy
- Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Adithya Vegaraju
- Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Hanxi Xiao
- Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Florian Baumann
- QIAGEN Sciences LLC, 19300 Germantown Rd, Germantown, MD 20874, USA
| | | | - Candice Church
- Dermatology Division, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Martin Prlic
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA.,Department of Pathology, University of Washington School of Medicine, Seattle, WA 98195, USA.,Department of Global Health, University of Washington School of Medicine, Seattle, WA 98195, USA
| | | | - Paul Nghiem
- Dermatology Division, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA.,Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Stanley Riddell
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA.,Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Surojit Sarkar
- Division of Hematology and Oncology, Department of Pediatrics, University of Washington School of Medicine, Seattle, WA 98195, USA.,Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute, Seattle, WA 98101, USA.,Department of Pathology, University of Washington School of Medicine, Seattle, WA 98195, USA
| |
Collapse
|
24
|
Ruterbusch M, Pruner KB, Shehata L, Pepper M. In Vivo CD4 + T Cell Differentiation and Function: Revisiting the Th1/Th2 Paradigm. Annu Rev Immunol 2021; 38:705-725. [PMID: 32340571 DOI: 10.1146/annurev-immunol-103019-085803] [Citation(s) in RCA: 272] [Impact Index Per Article: 90.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The discovery of CD4+ T cell subset-defining master transcription factors and framing of the Th1/Th2 paradigm ignited the CD4+ T cell field. Advances in in vivo experimental systems, however, have revealed that more complex lineage-defining transcriptional networks direct CD4+ T cell differentiation in the lymphoid organs and tissues. This review focuses on the layers of fate decisions that inform CD4+ T cell differentiation in vivo. Cytokine production by antigen-presenting cells and other innate cells influences the CD4+ T cell effector program [e.g., T helper type 1 (Th1), Th2, Th17]. Signals downstream of the T cell receptor influence whether individual clones bearing hallmarks of this effector program become T follicular helper cells, supporting development of B cells expressing specific antibody isotypes, or T effector cells, which activate microbicidal innate cells in tissues. These bifurcated, parallel axes allow CD4+ T cells to augment their particular effector program and prevent disease.
Collapse
Affiliation(s)
- Mikel Ruterbusch
- Department of Immunology, University of Washington School of Medicine, Seattle, Washington 98109, USA; ,
| | - Kurt B Pruner
- Department of Immunology, University of Washington School of Medicine, Seattle, Washington 98109, USA; ,
| | - Laila Shehata
- Department of Immunology, University of Washington School of Medicine, Seattle, Washington 98109, USA; ,
| | - Marion Pepper
- Department of Immunology, University of Washington School of Medicine, Seattle, Washington 98109, USA; ,
| |
Collapse
|
25
|
Mold JE, Modolo L, Hård J, Zamboni M, Larsson AJM, Stenudd M, Eriksson CJ, Durif G, Ståhl PL, Borgström E, Picelli S, Reinius B, Sandberg R, Réu P, Talavera-Lopez C, Andersson B, Blom K, Sandberg JK, Picard F, Michaëlsson J, Frisén J. Divergent clonal differentiation trajectories establish CD8 + memory T cell heterogeneity during acute viral infections in humans. Cell Rep 2021; 35:109174. [PMID: 34038736 DOI: 10.1016/j.celrep.2021.109174] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 02/15/2021] [Accepted: 05/04/2021] [Indexed: 02/08/2023] Open
Abstract
The CD8+ T cell response to an antigen is composed of many T cell clones with unique T cell receptors, together forming a heterogeneous repertoire of effector and memory cells. How individual T cell clones contribute to this heterogeneity throughout immune responses remains largely unknown. In this study, we longitudinally track human CD8+ T cell clones expanding in response to yellow fever virus (YFV) vaccination at the single-cell level. We observed a drop in clonal diversity in blood from the acute to memory phase, suggesting that clonal selection shapes the circulating memory repertoire. Clones in the memory phase display biased differentiation trajectories along a gradient from stem cell to terminally differentiated effector memory fates. In secondary responses, YFV- and influenza-specific CD8+ T cell clones are poised to recapitulate skewed differentiation trajectories. Collectively, we show that the sum of distinct clonal phenotypes results in the multifaceted human T cell response to acute viral infections.
Collapse
Affiliation(s)
- Jeff E Mold
- Department of Cell and Molecular Biology, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Laurent Modolo
- LBBE, UMR CNRS 5558, Université Lyon 1, Villeurbanne, France LBMC UMR 5239 CNRS/ENS Lyon, Lyon, France
| | - Joanna Hård
- Department of Cell and Molecular Biology, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Margherita Zamboni
- Department of Cell and Molecular Biology, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Anton J M Larsson
- Department of Cell and Molecular Biology, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Moa Stenudd
- Department of Cell and Molecular Biology, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Carl-Johan Eriksson
- Department of Cell and Molecular Biology, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Ghislain Durif
- LBBE, UMR CNRS 5558, Université Lyon 1, Villeurbanne, France LBMC UMR 5239 CNRS/ENS Lyon, Lyon, France
| | - Patrik L Ståhl
- Department of Cell and Molecular Biology, Karolinska Institutet, 171 77 Stockholm, Sweden; Science for Life Laboratory, Department of Gene Technology, KTH Royal Institute of Technology, 106 91 Stockholm, Sweden
| | - Erik Borgström
- Science for Life Laboratory, Department of Gene Technology, KTH Royal Institute of Technology, 106 91 Stockholm, Sweden
| | - Simone Picelli
- Department of Cell and Molecular Biology, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Björn Reinius
- Department of Cell and Molecular Biology, Karolinska Institutet, 171 77 Stockholm, Sweden; Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Rickard Sandberg
- Department of Cell and Molecular Biology, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Pedro Réu
- Department of Cell and Molecular Biology, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Carlos Talavera-Lopez
- Department of Cell and Molecular Biology, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Björn Andersson
- Department of Cell and Molecular Biology, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Kim Blom
- Center for Infectious Medicine, Department of Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, 141 86 Stockholm, Sweden
| | - Johan K Sandberg
- Center for Infectious Medicine, Department of Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, 141 86 Stockholm, Sweden
| | - Franck Picard
- LBBE, UMR CNRS 5558, Université Lyon 1, Villeurbanne, France LBMC UMR 5239 CNRS/ENS Lyon, Lyon, France
| | - Jakob Michaëlsson
- Center for Infectious Medicine, Department of Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, 141 86 Stockholm, Sweden.
| | - Jonas Frisén
- Department of Cell and Molecular Biology, Karolinska Institutet, 171 77 Stockholm, Sweden.
| |
Collapse
|
26
|
Chung HK, McDonald B, Kaech SM. The architectural design of CD8+ T cell responses in acute and chronic infection: Parallel structures with divergent fates. J Exp Med 2021; 218:e20201730. [PMID: 33755719 PMCID: PMC7992501 DOI: 10.1084/jem.20201730] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 01/12/2021] [Accepted: 01/13/2021] [Indexed: 12/19/2022] Open
Abstract
In response to infection, T cells adopt a range of differentiation states, creating numerous heterogeneous subsets that exhibit different phenotypes, functions, and migration patterns. This T cell heterogeneity is a universal feature of T cell immunity, needed to effectively control pathogens in a context-dependent manner and generate long-lived immunity to those pathogens. Here, we review new insights into differentiation state dynamics and population heterogeneity of CD8+ T cells in acute and chronic viral infections and cancer and highlight the parallels and distinctions between acute and chronic antigen stimulation settings. We focus on transcriptional and epigenetic networks that modulate the plasticity and terminal differentiation of antigen-specific CD8+ T cells and generate functionally diverse T cell subsets with different roles to combat infection and cancer.
Collapse
Affiliation(s)
- H. Kay Chung
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA
| | - Bryan McDonald
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA
- Biomedical Sciences Graduate Program, University of California San Diego, La Jolla, CA
| | - Susan M. Kaech
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA
| |
Collapse
|
27
|
Pinkard H, Baghdassarian H, Mujal A, Roberts E, Hu KH, Friedman DH, Malenica I, Shagam T, Fries A, Corbin K, Krummel MF, Waller L. Learned adaptive multiphoton illumination microscopy for large-scale immune response imaging. Nat Commun 2021; 12:1916. [PMID: 33772022 PMCID: PMC7997974 DOI: 10.1038/s41467-021-22246-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Accepted: 03/04/2021] [Indexed: 11/09/2022] Open
Abstract
Multiphoton microscopy is a powerful technique for deep in vivo imaging in scattering samples. However, it requires precise, sample-dependent increases in excitation power with depth in order to generate contrast in scattering tissue, while minimizing photobleaching and phototoxicity. We show here how adaptive imaging can optimize illumination power at each point in a 3D volume as a function of the sample's shape, without the need for specialized fluorescent labeling. Our method relies on training a physics-based machine learning model using cells with identical fluorescent labels imaged in situ. We use this technique for in vivo imaging of immune responses in mouse lymph nodes following vaccination. We achieve visualization of physiologically realistic numbers of antigen-specific T cells (~2 orders of magnitude lower than previous studies), and demonstrate changes in the global organization and motility of dendritic cell networks during the early stages of the immune response. We provide a step-by-step tutorial for implementing this technique using exclusively open-source hardware and software.
Collapse
Affiliation(s)
- Henry Pinkard
- Department of Electrical Engineering and Computer Sciences, University of California, Berkeley, CA, USA.
- Computational Biology Graduate Group, University of California, Berkeley, CA, USA.
- Berkeley Institute for Data Science, Berkeley, CA, USA.
- University of California San Francisco Bakar Computational Health Sciences Institute, San Francisco, CA, USA.
| | - Hratch Baghdassarian
- Department of Pathology, University of California, San Francisco, San Francisco, CA, USA
| | - Adriana Mujal
- Department of Pathology, University of California, San Francisco, San Francisco, CA, USA
| | - Ed Roberts
- Department of Pathology, University of California, San Francisco, San Francisco, CA, USA
| | - Kenneth H Hu
- Department of Pathology, University of California, San Francisco, San Francisco, CA, USA
| | | | - Ivana Malenica
- Berkeley Institute for Data Science, Berkeley, CA, USA
- Division of Biostatistics, University of California, Berkeley, CA, USA
| | - Taylor Shagam
- Department of Pathology, University of California, San Francisco, San Francisco, CA, USA
| | - Adam Fries
- Department of Pathology, University of California, San Francisco, San Francisco, CA, USA
| | - Kaitlin Corbin
- Department of Pathology, University of California, San Francisco, San Francisco, CA, USA
| | - Matthew F Krummel
- Department of Pathology, University of California, San Francisco, San Francisco, CA, USA
| | - Laura Waller
- Computational Biology Graduate Group, University of California, Berkeley, CA, USA
- Berkeley Institute for Data Science, Berkeley, CA, USA
| |
Collapse
|
28
|
Hutchinson JA, Kronenberg K, Riquelme P, Wenzel JJ, Glehr G, Schilling HL, Zeman F, Evert K, Schmiedel M, Mickler M, Drexler K, Bitterer F, Cordero L, Beyer L, Bach C, Koestler J, Burkhardt R, Schlitt HJ, Hellwig D, Werner JM, Spang R, Schmidt B, Geissler EK, Haferkamp S. Virus-specific memory T cell responses unmasked by immune checkpoint blockade cause hepatitis. Nat Commun 2021; 12:1439. [PMID: 33664251 PMCID: PMC7933278 DOI: 10.1038/s41467-021-21572-y] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 01/29/2021] [Indexed: 02/08/2023] Open
Abstract
Treatment of advanced melanoma with combined PD-1/CTLA-4 blockade commonly causes serious immune-mediated complications. Here, we identify a subset of patients predisposed to immune checkpoint blockade-related hepatitis who are distinguished by chronic expansion of effector memory CD4+ T cells (TEM cells). Pre-therapy CD4+ TEM cell expansion occurs primarily during autumn or winter in patients with metastatic disease and high cytomegalovirus (CMV)-specific serum antibody titres. These clinical features implicate metastasis-dependent, compartmentalised CMV reactivation as the cause of CD4+ TEM expansion. Pre-therapy CD4+ TEM expansion predicts hepatitis in CMV-seropositive patients, opening possibilities for avoidance or prevention. 3 of 4 patients with pre-treatment CD4+ TEM expansion who received αPD-1 monotherapy instead of αPD-1/αCTLA-4 therapy remained hepatitis-free. 4 of 4 patients with baseline CD4+ TEM expansion given prophylactic valganciclovir and αPD-1/αCTLA-4 therapy remained hepatitis-free. Our findings exemplify how pathogen exposure can shape clinical reactions after cancer therapy and how this insight leads to therapeutic innovations.
Collapse
Affiliation(s)
- James A. Hutchinson
- grid.411941.80000 0000 9194 7179Department of Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Katharina Kronenberg
- grid.411941.80000 0000 9194 7179Department of Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Paloma Riquelme
- grid.411941.80000 0000 9194 7179Department of Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Jürgen J. Wenzel
- grid.411941.80000 0000 9194 7179Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg, Regensburg, Germany
| | - Gunther Glehr
- grid.7727.50000 0001 2190 5763Institute of Functional Genomics and Statistical Bioinformatics, University of Regensburg, Regensburg, Germany
| | - Hannah-Lou Schilling
- grid.411941.80000 0000 9194 7179Department of Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Florian Zeman
- grid.411941.80000 0000 9194 7179Center for Clinical Studies, University Hospital Regensburg, Regensburg, Germany
| | - Katja Evert
- grid.411941.80000 0000 9194 7179Institute of Pathology, University Hospital Regensburg, Regensburg, Germany
| | - Martin Schmiedel
- grid.411941.80000 0000 9194 7179Department of Nuclear Medicine, University Hospital Regensburg, Regensburg, Germany
| | - Marion Mickler
- grid.411941.80000 0000 9194 7179Department of Dermatology, University Hospital Regensburg, Regensburg, Germany
| | - Konstantin Drexler
- grid.411941.80000 0000 9194 7179Department of Dermatology, University Hospital Regensburg, Regensburg, Germany
| | - Florian Bitterer
- grid.411941.80000 0000 9194 7179Department of Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Laura Cordero
- grid.411941.80000 0000 9194 7179Department of Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Lukas Beyer
- grid.411941.80000 0000 9194 7179Institute of Radiology, University Hospital Regensburg, Regensburg, Germany
| | - Christian Bach
- grid.411668.c0000 0000 9935 6525Department of Medicine V, University Hospital Erlangen, Erlangen, Germany
| | - Josef Koestler
- grid.411941.80000 0000 9194 7179Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg, Regensburg, Germany
| | - Ralph Burkhardt
- grid.411941.80000 0000 9194 7179Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Regensburg, Germany
| | - Hans J. Schlitt
- grid.411941.80000 0000 9194 7179Department of Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Dirk Hellwig
- grid.411941.80000 0000 9194 7179Department of Nuclear Medicine, University Hospital Regensburg, Regensburg, Germany
| | - Jens M. Werner
- grid.411941.80000 0000 9194 7179Department of Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Rainer Spang
- grid.7727.50000 0001 2190 5763Institute of Functional Genomics and Statistical Bioinformatics, University of Regensburg, Regensburg, Germany
| | - Barbara Schmidt
- grid.411941.80000 0000 9194 7179Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg, Regensburg, Germany
| | - Edward K. Geissler
- grid.411941.80000 0000 9194 7179Department of Surgery, University Hospital Regensburg, Regensburg, Germany ,Personalised Tumour Therapy, Fraunhofer Institute for Experimental Medicine and Toxicology, Regensburg, Germany
| | - Sebastian Haferkamp
- grid.411941.80000 0000 9194 7179Department of Dermatology, University Hospital Regensburg, Regensburg, Germany
| |
Collapse
|
29
|
Chen Z, Arai E, Khan O, Zhang Z, Ngiow SF, He Y, Huang H, Manne S, Cao Z, Baxter AE, Cai Z, Freilich E, Ali MA, Giles JR, Wu JE, Greenplate AR, Hakeem MA, Chen Q, Kurachi M, Nzingha K, Ekshyyan V, Mathew D, Wen Z, Speck NA, Battle A, Berger SL, Wherry EJ, Shi J. In vivo CD8 + T cell CRISPR screening reveals control by Fli1 in infection and cancer. Cell 2021; 184:1262-1280.e22. [PMID: 33636129 PMCID: PMC8054351 DOI: 10.1016/j.cell.2021.02.019] [Citation(s) in RCA: 117] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 10/26/2020] [Accepted: 02/05/2021] [Indexed: 12/21/2022]
Abstract
Improving effector activity of antigen-specific T cells is a major goal in cancer immunotherapy. Despite the identification of several effector T cell (TEFF)-driving transcription factors (TFs), the transcriptional coordination of TEFF biology remains poorly understood. We developed an in vivo T cell CRISPR screening platform and identified a key mechanism restraining TEFF biology through the ETS family TF, Fli1. Genetic deletion of Fli1 enhanced TEFF responses without compromising memory or exhaustion precursors. Fli1 restrained TEFF lineage differentiation by binding to cis-regulatory elements of effector-associated genes. Loss of Fli1 increased chromatin accessibility at ETS:RUNX motifs, allowing more efficient Runx3-driven TEFF biology. CD8+ T cells lacking Fli1 provided substantially better protection against multiple infections and tumors. These data indicate that Fli1 safeguards the developing CD8+ T cell transcriptional landscape from excessive ETS:RUNX-driven TEFF cell differentiation. Moreover, genetic deletion of Fli1 improves TEFF differentiation and protective immunity in infections and cancer.
Collapse
Affiliation(s)
- Zeyu Chen
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Parker Institute for Cancer Immunotherapy at University of Pennsylvania, Philadelphia, PA, USA
| | - Eri Arai
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Omar Khan
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Zhen Zhang
- Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Shin Foong Ngiow
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Parker Institute for Cancer Immunotherapy at University of Pennsylvania, Philadelphia, PA, USA
| | - Yuan He
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Hua Huang
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sasikanth Manne
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Zhendong Cao
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Amy E Baxter
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Zhangying Cai
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Elizabeth Freilich
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mohammed A Ali
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Josephine R Giles
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jennifer E Wu
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Allison R Greenplate
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mohamed A Hakeem
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Qingzhou Chen
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Makoto Kurachi
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kito Nzingha
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Viktoriya Ekshyyan
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Divij Mathew
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Zhuoyu Wen
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Nancy A Speck
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Alexis Battle
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA; Department of Computer Science, Johns Hopkins University, Baltimore, MD, USA
| | - Shelley L Berger
- Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - E John Wherry
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Parker Institute for Cancer Immunotherapy at University of Pennsylvania, Philadelphia, PA, USA.
| | - Junwei Shi
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
30
|
Kim YC, Hsueh HT, Kim N, Rodriguez J, Leo KT, Rao D, West NE, Hanes J, Suk JS. Strategy to enhance dendritic cell-mediated DNA vaccination in the lung. ADVANCED THERAPEUTICS 2021; 4:2000228. [PMID: 33709020 PMCID: PMC7941873 DOI: 10.1002/adtp.202000228] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
We here introduce a new paradigm to promote pulmonary DNA vaccination. Specifically, we demonstrate that nanoparticles designed to rapidly penetrate airway mucus (mucus-penetrating particle or MPP) enhance the delivery of inhaled model DNA vaccine (i.e. ovalbumin-expressing plasmids) to pulmonary dendritic cells (DC), leading to robust and durable local and trans-mucosal immunity. In contrast, mucus-impermeable particles were poorly taken up by pulmonary DC following inhalation, despite their superior ability to mediate DC uptake in vitro compared to MPP. In addition to the enhanced immunity achieved in mucosal surfaces, inhaled MPP unexpectedly provided significantly greater systemic immune responses compared to gold-standard approaches applied in the clinic for systemic vaccination, including intradermal injection and intramuscular electroporation. We also showed here that inhaled MPP significantly enhanced the survival of an orthotopic mouse model of aggressive lung cancer compared to the gold-standard approaches. Importantly, we discovered that MPP-mediated pulmonary DNA vaccination induced memory T-cell immunity, particularly the ready-to-act effector memory-biased phenotype, both locally and systemically. The findings here underscore the importance of breaching the airway mucus barrier to facilitate DNA vaccine uptake by pulmonary DC and thus to initiate full-blown immune responses.
Collapse
Affiliation(s)
- Yoo C. Kim
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21231
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD 21231
| | - Henry T. Hsueh
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21231
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218
| | - Namho Kim
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21231
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218
| | - Jason Rodriguez
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21231
| | - Kirby T. Leo
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21231
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21218
| | - Divya Rao
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21231
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218
| | - Natalie E. West
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Justin Hanes
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21231
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD 21231
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21218
| | - Jung Soo Suk
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21231
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD 21231
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218
| |
Collapse
|
31
|
Johnnidis JB, Muroyama Y, Ngiow SF, Chen Z, Manne S, Cai Z, Song S, Platt JM, Schenkel JM, Abdel-Hakeem M, Beltra JC, Greenplate AR, Ali MAA, Nzingha K, Giles JR, Harly C, Attanasio J, Pauken KE, Bengsch B, Paley MA, Tomov VT, Kurachi M, Vignali DAA, Sharpe AH, Reiner SL, Bhandoola A, Johnson FB, Wherry EJ. Inhibitory signaling sustains a distinct early memory CD8 + T cell precursor that is resistant to DNA damage. Sci Immunol 2021; 6:6/55/eabe3702. [PMID: 33452106 DOI: 10.1126/sciimmunol.abe3702] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 12/10/2020] [Indexed: 12/16/2022]
Abstract
The developmental origins of memory T cells remain incompletely understood. During the expansion phase of acute viral infection, we identified a distinct subset of virus-specific CD8+ T cells that possessed distinct characteristics including expression of CD62L, T cell factor 1 (TCF-1), and Eomesodermin; relative quiescence; expression of activation markers; and features of limited effector differentiation. These cells were a quantitatively minor subpopulation of the TCF-1+ pool and exhibited self-renewal, heightened DNA damage surveillance activity, and preferential long-term recall capacity. Despite features of memory and somewhat restrained proliferation during the expansion phase, this subset displayed evidence of stronger TCR signaling than other responding CD8+ T cells, coupled with elevated expression of multiple inhibitory receptors including programmed cell death 1 (PD-1), lymphocyte activating gene 3 (LAG-3), cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), CD5, and CD160. Genetic ablation of PD-1 and LAG-3 compromised the formation of this CD62Lhi TCF-1+ subset and subsequent CD8+ T cell memory. Although central memory phenotype CD8+ T cells were formed in the absence of these cells, subsequent memory CD8+ T cell recall responses were compromised. Together, these results identify an important link between genome integrity maintenance and CD8+ T cell memory. Moreover, the data indicate a role for inhibitory receptors in preserving key memory CD8+ T cell precursors during initial activation and differentiation. Identification of this rare subpopulation within the memory CD8+ T cell precursor pool may help reconcile models of the developmental origin of long-term CD8+ T cell memory.
Collapse
Affiliation(s)
- Jonathan B Johnnidis
- Department of Biology, University of Pennsylvania, Philadelphia, PA 19104, USA.,Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA.,Institute for Immunology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yuki Muroyama
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA.,Institute for Immunology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Shin Foong Ngiow
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA.,Institute for Immunology, University of Pennsylvania, Philadelphia, PA 19104, USA.,Parker Institute for Cancer Immunotherapy, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Zeyu Chen
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA.,Institute for Immunology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sasikanth Manne
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA.,Institute for Immunology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Zhangying Cai
- Division of Biology and Biomedical Sciences, Washington University, St. Louis, MO 63110, USA
| | - Shufei Song
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jesse M Platt
- Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA.,Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Jason M Schenkel
- Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115, USA.,David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Mohamed Abdel-Hakeem
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA.,Institute for Immunology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jean-Christophe Beltra
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA.,Institute for Immunology, University of Pennsylvania, Philadelphia, PA 19104, USA.,Parker Institute for Cancer Immunotherapy, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Allison R Greenplate
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA.,Institute for Immunology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Mohammed-Alkhatim A Ali
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA.,Institute for Immunology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kito Nzingha
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA.,Institute for Immunology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Josephine R Giles
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA.,Institute for Immunology, University of Pennsylvania, Philadelphia, PA 19104, USA.,Parker Institute for Cancer Immunotherapy, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Christelle Harly
- T-Cell Biology and Development Unit, Laboratory of Genome Integrity, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA.,Université de Nantes, INSERM, CNRS, CRCINA, Nantes, France.,LabEx IGO 'Immunotherapy, Graft, Oncology', Nantes, France
| | - John Attanasio
- Institute for Immunology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kristen E Pauken
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA.,Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Bertram Bengsch
- Department of Medicine II, Gastroenterology, Hepatology, Endocrinology, and Infectious Diseases, University Medical Center Freiburg, Germany.,Signaling Research Centers BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Michael A Paley
- Department of Medicine, Washington University in Saint Louis School of Medicine, St. Louis, MO 63110, USA
| | - Vesselin T Tomov
- Institute for Immunology, University of Pennsylvania, Philadelphia, PA 19104, USA.,Department of Medicine, Division of Gastroenterology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Makoto Kurachi
- Department of Molecular Genetics, Graduate School of Medicine, Kanazawa University, Kanazawa, Japan
| | - Dario A A Vignali
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA.,Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh PA 15232, USA.,Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA
| | - Arlene H Sharpe
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA.,Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Steven L Reiner
- Department of Microbiology and Immunology and Department of Pediatrics, Columbia University, New York, NY 10032, USA
| | - Avinash Bhandoola
- T-Cell Biology and Development Unit, Laboratory of Genome Integrity, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - F Bradley Johnson
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - E John Wherry
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA. .,Institute for Immunology, University of Pennsylvania, Philadelphia, PA 19104, USA.,Parker Institute for Cancer Immunotherapy, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
32
|
Rostamian H, Fallah-Mehrjardi K, Khakpoor-Koosheh M, Pawelek JM, Hadjati J, Brown CE, Mirzaei HR. A metabolic switch to memory CAR T cells: Implications for cancer treatment. Cancer Lett 2020; 500:107-118. [PMID: 33290868 DOI: 10.1016/j.canlet.2020.12.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 12/01/2020] [Accepted: 12/02/2020] [Indexed: 12/27/2022]
Abstract
Therapeutic efficacy of chimeric antigen receptor (CAR) T cells is associated with their expansion, persistence and effector function. Although CAR T cell therapy has shown remarkable therapeutic effects in hematological malignancies, its therapeutic efficacy has been limited in some types of cancers - in particular, solid tumors - partially due to the cells' inability to persist and the acquisition of T cell dysfunction within a harsh immunosuppressive tumor microenvironment. Therefore, it would be expected that generation of CAR T cells with intrinsic properties for functional longevity, such as the cells with early-memory phenotypes, could beneficially enhance antitumor immunity. Furthermore, because the metabolic pathways of CAR T cells help determine cellular differentiation and lifespan, therapies targeting such pathways like glycolysis and oxidative phosphorylation, can alter CAR T cell fate and durability within tumors. Here we discuss how reprogramming of CAR T cell metabolism and metabolic switch to memory CAR T cells influences their antitumor activity. We also offer potential strategies for targeting these metabolic circuits in the setting of adoptive CAR T cell therapy, aiming to better unleash the potential of adoptive CAR T cell therapy in the clinic.
Collapse
Affiliation(s)
- Hosein Rostamian
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Keyvan Fallah-Mehrjardi
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Khakpoor-Koosheh
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - John M Pawelek
- Department of Dermatology and the Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA
| | - Jamshid Hadjati
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Christine E Brown
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope Medical Center, Duarte, CA, 91010, USA; Department of Immuno-Oncology, City of Hope Beckman Research Institute, Duarte, CA, 91010, USA.
| | - Hamid R Mirzaei
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
33
|
Puiffe ML, Dupont A, Sako N, Gatineau J, Cohen JL, Mestivier D, Lebon A, Prévost-Blondel A, Castellano F, Molinier-Frenkel V. IL4I1 Accelerates the Expansion of Effector CD8 + T Cells at the Expense of Memory Precursors by Increasing the Threshold of T-Cell Activation. Front Immunol 2020; 11:600012. [PMID: 33343572 PMCID: PMC7746639 DOI: 10.3389/fimmu.2020.600012] [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: 08/28/2020] [Accepted: 11/03/2020] [Indexed: 12/25/2022] Open
Abstract
IL4I1 is an immunoregulatory enzyme that inhibits CD8 T-cell proliferation in vitro and in the tumoral context. Here, we dissected the effect of IL4I1 on CD8 T-cell priming by studying the differentiation of a transgenic CD8 T-cell clone and the endogenous repertoire in a mouse model of acute lymphocytic choriomeningitis virus (LCMV) infection. Unexpectedly, we show that IL4I1 accelerates the expansion of functional effector CD8 T cells during the first several days after infection and increases the average affinity of the elicited repertoire, supporting more efficient LCMV clearance in WT mice than IL4I1-deficient mice. Conversely, IL4I1 restrains the differentiation of CD8 T-cells into long-lived memory precursors and favors the memory response to the most immunodominant peptides. IL4I1 expression does not affect the phenotype or antigen-presenting functions of dendritic cells (DCs), but directly reduces the stability of T-DC immune synapses in vitro, thus dampening T-cell activation. Overall, our results support a model in which IL4I1 increases the threshold of T-cell activation, indirectly promoting the priming of high-affinity clones while limiting memory T-cell differentiation.
Collapse
Affiliation(s)
- Marie-Line Puiffe
- Virus-Immunity-Cancer Department, Institut Mondor de Recherche Biomédicale (IMRB), INSERM U955, Université Paris-Est Créteil, Créteil, France
| | - Aurélie Dupont
- Virus-Immunity-Cancer Department, Institut Mondor de Recherche Biomédicale (IMRB), INSERM U955, Université Paris-Est Créteil, Créteil, France
| | - Nouhoum Sako
- Virus-Immunity-Cancer Department, Institut Mondor de Recherche Biomédicale (IMRB), INSERM U955, Université Paris-Est Créteil, Créteil, France
| | - Jérôme Gatineau
- Virus-Immunity-Cancer Department, Institut Mondor de Recherche Biomédicale (IMRB), INSERM U955, Université Paris-Est Créteil, Créteil, France
| | - José L Cohen
- Virus-Immunity-Cancer Department, Institut Mondor de Recherche Biomédicale (IMRB), INSERM U955, Université Paris-Est Créteil, Créteil, France
| | - Denis Mestivier
- Bioinformatics Core Laboratory, Institut Mondor de Recherche Biomédicale (IMRB), INSERM U955, Université Paris-Est Créteil, Créteil, France
| | - Agnès Lebon
- INSERM U1016, CNRS UMR8104, Institut Cochin, Université de Paris, Paris, France
| | | | - Flavia Castellano
- Virus-Immunity-Cancer Department, Institut Mondor de Recherche Biomédicale (IMRB), INSERM U955, Université Paris-Est Créteil, Créteil, France.,Pathobiology Department, Groupe Hospitalo-Universitaire Chenevier-Mondor, AP-HP, Créteil, France
| | - Valérie Molinier-Frenkel
- Virus-Immunity-Cancer Department, Institut Mondor de Recherche Biomédicale (IMRB), INSERM U955, Université Paris-Est Créteil, Créteil, France.,Pathobiology Department, Groupe Hospitalo-Universitaire Chenevier-Mondor, AP-HP, Créteil, France
| |
Collapse
|
34
|
Parga-Vidal L, van Gisbergen KPJM. Area under Immunosurveillance: Dedicated Roles of Memory CD8 T-Cell Subsets. Cold Spring Harb Perspect Biol 2020; 12:cshperspect.a037796. [PMID: 32839203 DOI: 10.1101/cshperspect.a037796] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Immunological memory, defined as the ability to respond in an enhanced manner upon secondary encounter with the same pathogen, can provide substantial protection against infectious disease. The improved protection is mediated in part by different populations of memory CD8 T cells that are retained after primary infection. Memory cells persist in the absence of pathogen-derived antigens and enable secondary CD8 T-cell responses with accelerated kinetics and of larger magnitude after reencounter with the same pathogen. At least three subsets of memory T cells have been defined that are referred to as central memory CD8 T cells (Tcm), effector memory CD8 T cells (Tem), and tissue-resident memory CD8 T cells (Trm). Tcm and Tem are circulating memory T cells that mediate bodywide immune surveillance in search of invading pathogens. In contrast, Trm permanently reside in peripheral barrier tissues, where they form a stationary defensive line of sentinels that alert the immune system upon pathogen reencounter. The characterization of these different subsets has been instrumental in our understanding of the strategies that memory T cells employ to counter invading pathogens. It is clear that memory T cells not only have a numerical advantage over naive T cells resulting in improved protection in secondary responses, but also acquire distinct sets of competencies that assist in pathogen clearance. Nevertheless, inherent challenges are associated with the allocation of memory T cells to a limited number of subsets. The classification of memory T cells into Tcm, Tem, and Trm may not take into account the full extent of the heterogeneity that is observed in the memory population. Therefore, in this review, we will revisit the current classification of memory subsets, elaborate on functional and migratory properties attributed to Tcm, Tem, and Trm, and discuss how potential heterogeneity within these populations arises and persists.
Collapse
Affiliation(s)
- Loreto Parga-Vidal
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, 1066CX Amsterdam, The Netherlands
| | - Klaas P J M van Gisbergen
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, 1066CX Amsterdam, The Netherlands
| |
Collapse
|
35
|
Mondino A, Manzo T. To Remember or to Forget: The Role of Good and Bad Memories in Adoptive T Cell Therapy for Tumors. Front Immunol 2020; 11:1915. [PMID: 32973794 PMCID: PMC7481451 DOI: 10.3389/fimmu.2020.01915] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 07/16/2020] [Indexed: 12/17/2022] Open
Abstract
The generation of immunological memory is a hallmark of adaptive immunity by which the immune system "remembers" a previous encounter with an antigen expressed by pathogens, tumors, or normal tissues; and, upon secondary encounters, mounts faster and more effective recall responses. The establishment of T cell memory is influenced by both cell-intrinsic and cell-extrinsic factors, including genetic, epigenetic and environmental triggers. Our current knowledge of the mechanisms involved in memory T cell differentiation has instructed new opportunities to engineer T cells with enhanced anti-tumor activity. The development of adoptive T cell therapy has emerged as a powerful approach to cure a subset of patients with advanced cancers. Efficacy of this approach often requires long-term persistence of transferred T cell products, which can vary according to their origin and manufacturing conditions. Host preconditioning and post-transfer supporting strategies have shown to promote their engraftment and survival by limiting the competition with a hostile tumor microenvironment and between pre-existing immune cell subsets. Although in the general view pre-existing memory can confer a selective advantage to adoptive T cell therapy, here we propose that also "bad memories"-in the form of antigen-experienced T cell subsets-co-evolve with consequences on newly transferred lymphocytes. In this review, we will first provide an overview of selected features of memory T cell subsets and, then, discuss their putative implications for adoptive T cell therapy.
Collapse
Affiliation(s)
- Anna Mondino
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Teresa Manzo
- Department of Experimental Oncology, IRCCS European Institute of Oncology, Milan, Italy
| |
Collapse
|
36
|
Bottois H, Ngollo M, Hammoudi N, Courau T, Bonnereau J, Chardiny V, Grand C, Gergaud B, Allez M, Le Bourhis L. KLRG1 and CD103 Expressions Define Distinct Intestinal Tissue-Resident Memory CD8 T Cell Subsets Modulated in Crohn's Disease. Front Immunol 2020; 11:896. [PMID: 32477365 PMCID: PMC7235448 DOI: 10.3389/fimmu.2020.00896] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 04/17/2020] [Indexed: 12/14/2022] Open
Abstract
Intestinal tissue-resident memory CD8 T cells (Trm) are non-recirculating effector cells ideally positioned to detect and react to microbial infections in the gut mucosa. There is an emerging understanding of Trm cell differentiation and functions, but their implication in inflammatory bowel diseases, such as Crohn's disease (CD), is still unknown. Here, we describe CD8 cells in the human intestine expressing KLRG1 or CD103, two receptors of E-cadherin. While CD103 CD8 T cells are present in high numbers in the mucosa of CD patients and controls, KLRG1 CD8 T cells are increased in inflammatory conditions. Mucosal CD103 CD8 T cells are more responsive to TCR restimulation, but KLRG1 CD8 T cells show increased cytotoxic and proliferative potential. CD103 CD8 T cells originate mostly from KLRG1 negative cells after TCR triggering and TGFβ stimulation. Interestingly, mucosal CD103 CD8 T cells from CD patients display major changes in their transcriptomic landscape compared to controls. They express Th17 related genes including CCL20, IL22, and IL26, which could contribute to the pathogenesis of CD. Overall, these findings suggest that CD103 CD8 T cells in CD induce a tissue-wide alert increasing innate immune responses and recruitment of effector cells such as KLRG1 CD8 T cells.
Collapse
Affiliation(s)
- Hugo Bottois
- Université de Paris, INSERM U1160, EMiLy, Institut de Recherche Saint-Louis, Paris, France
| | - Marjolaine Ngollo
- Université de Paris, INSERM U1160, EMiLy, Institut de Recherche Saint-Louis, Paris, France
| | - Nassim Hammoudi
- Université de Paris, INSERM U1160, EMiLy, Institut de Recherche Saint-Louis, Paris, France.,Gastroenterology Department, Hopital Saint Louis, AP-HP, Paris, France
| | - Tristan Courau
- Université de Paris, INSERM U1160, EMiLy, Institut de Recherche Saint-Louis, Paris, France
| | - Julie Bonnereau
- Université de Paris, INSERM U1160, EMiLy, Institut de Recherche Saint-Louis, Paris, France
| | - Victor Chardiny
- Université de Paris, INSERM U1160, EMiLy, Institut de Recherche Saint-Louis, Paris, France
| | - Céline Grand
- Université de Paris, INSERM U1160, EMiLy, Institut de Recherche Saint-Louis, Paris, France
| | - Brice Gergaud
- Université de Paris, INSERM U1160, EMiLy, Institut de Recherche Saint-Louis, Paris, France
| | - Matthieu Allez
- Université de Paris, INSERM U1160, EMiLy, Institut de Recherche Saint-Louis, Paris, France.,Gastroenterology Department, Hopital Saint Louis, AP-HP, Paris, France
| | - Lionel Le Bourhis
- Université de Paris, INSERM U1160, EMiLy, Institut de Recherche Saint-Louis, Paris, France
| |
Collapse
|
37
|
Kim YC, Hsueh HT, Kim N, Rodriguez J, Leo KT, Rao D, West NE, Hanes J, Suk JS. Strategy to Enhance Dendritic Cell‐Mediated DNA Vaccination in the Lung. ADVANCED THERAPEUTICS 2020. [DOI: 10.1002/adtp.202000013] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Yoo C. Kim
- Center for Nanomedicine at the Wilmer Eye Institute Johns Hopkins University School of Medicine Baltimore MD 21231 USA
- Department of Ophthalmology Johns Hopkins University School of Medicine Baltimore MD 21231 USA
| | - Henry T. Hsueh
- Center for Nanomedicine at the Wilmer Eye Institute Johns Hopkins University School of Medicine Baltimore MD 21231 USA
- Department of Chemical & Biomolecular Engineering Johns Hopkins University Baltimore MD 21218 USA
| | - Namho Kim
- Center for Nanomedicine at the Wilmer Eye Institute Johns Hopkins University School of Medicine Baltimore MD 21231 USA
- Department of Chemical & Biomolecular Engineering Johns Hopkins University Baltimore MD 21218 USA
| | - Jason Rodriguez
- Center for Nanomedicine at the Wilmer Eye Institute Johns Hopkins University School of Medicine Baltimore MD 21231 USA
| | - Kirby T. Leo
- Center for Nanomedicine at the Wilmer Eye Institute Johns Hopkins University School of Medicine Baltimore MD 21231 USA
- Department of Biomedical Engineering Johns Hopkins University Baltimore MD 21218 USA
| | - Divya Rao
- Center for Nanomedicine at the Wilmer Eye Institute Johns Hopkins University School of Medicine Baltimore MD 21231 USA
- Department of Chemical & Biomolecular Engineering Johns Hopkins University Baltimore MD 21218 USA
| | - Natalie E. West
- Division of Pulmonary and Critical Care Medicine Department of Medicine Johns Hopkins University School of Medicine Baltimore MD 21287 USA
| | - Justin Hanes
- Center for Nanomedicine at the Wilmer Eye Institute Johns Hopkins University School of Medicine Baltimore MD 21231 USA
- Department of Ophthalmology Johns Hopkins University School of Medicine Baltimore MD 21231 USA
- Department of Chemical & Biomolecular Engineering Johns Hopkins University Baltimore MD 21218 USA
- Department of Biomedical Engineering Johns Hopkins University Baltimore MD 21218 USA
| | - Jung Soo Suk
- Center for Nanomedicine at the Wilmer Eye Institute Johns Hopkins University School of Medicine Baltimore MD 21231 USA
- Department of Ophthalmology Johns Hopkins University School of Medicine Baltimore MD 21231 USA
- Department of Chemical & Biomolecular Engineering Johns Hopkins University Baltimore MD 21218 USA
| |
Collapse
|
38
|
Davenport B, Eberlein J, Nguyen TT, Victorino F, Jhun K, Abuirqeba H, van der Heide V, Heeger P, Homann D. Aging boosts antiviral CD8+T cell memory through improved engagement of diversified recall response determinants. PLoS Pathog 2019; 15:e1008144. [PMID: 31697793 PMCID: PMC6863560 DOI: 10.1371/journal.ppat.1008144] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 11/19/2019] [Accepted: 10/10/2019] [Indexed: 12/13/2022] Open
Abstract
The determinants of protective CD8+ memory T cell (CD8+TM) immunity remain incompletely defined and may in fact constitute an evolving agency as aging CD8+TM progressively acquire enhanced rather than impaired recall capacities. Here, we show that old as compared to young antiviral CD8+TM more effectively harness disparate molecular processes (cytokine signaling, trafficking, effector functions, and co-stimulation/inhibition) that in concert confer greater secondary reactivity. The relative reliance on these pathways is contingent on the nature of the secondary challenge (greater for chronic than acute viral infections) and over time, aging CD8+TM re-establish a dependence on the same accessory signals required for effective priming of naïve CD8+T cells in the first place. Thus, our findings reveal a temporal regulation of complementary recall response determinants that is consistent with the recently proposed "rebound model" according to which aging CD8+TM properties are gradually aligned with those of naïve CD8+T cells; our identification of a broadly diversified collection of immunomodulatory targets may further provide a foundation for the potential therapeutic "tuning" of CD8+TM immunity.
Collapse
Affiliation(s)
- Bennett Davenport
- Department of Anesthesiology & Barbara Davis Center for Childhood Diabetes, University of Colorado Denver, Aurora, Colorado, United States of America
- Integrated Department of Immunology, University of Colorado Denver and National Jewish Health, Denver, Colorado, United States of America
- Diabetes, Obesity & Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Jens Eberlein
- Department of Anesthesiology & Barbara Davis Center for Childhood Diabetes, University of Colorado Denver, Aurora, Colorado, United States of America
| | - Tom T. Nguyen
- Department of Anesthesiology & Barbara Davis Center for Childhood Diabetes, University of Colorado Denver, Aurora, Colorado, United States of America
| | - Francisco Victorino
- Department of Anesthesiology & Barbara Davis Center for Childhood Diabetes, University of Colorado Denver, Aurora, Colorado, United States of America
- Integrated Department of Immunology, University of Colorado Denver and National Jewish Health, Denver, Colorado, United States of America
| | - Kevin Jhun
- Diabetes, Obesity & Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Haedar Abuirqeba
- Diabetes, Obesity & Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Verena van der Heide
- Diabetes, Obesity & Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Peter Heeger
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Dirk Homann
- Department of Anesthesiology & Barbara Davis Center for Childhood Diabetes, University of Colorado Denver, Aurora, Colorado, United States of America
- Integrated Department of Immunology, University of Colorado Denver and National Jewish Health, Denver, Colorado, United States of America
- Diabetes, Obesity & Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
39
|
Prier JE, Li J, Gearing LJ, Olshansky M, Sng XYX, Hertzog PJ, Turner SJ. Early T-BET Expression Ensures an Appropriate CD8 + Lineage-Specific Transcriptional Landscape after Influenza A Virus Infection. THE JOURNAL OF IMMUNOLOGY 2019; 203:1044-1054. [PMID: 31227580 DOI: 10.4049/jimmunol.1801431] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 05/31/2019] [Indexed: 01/12/2023]
Abstract
Virus infection triggers large-scale changes in the phenotype and function of naive CD8+ T cells, resulting in the generation of effector and memory T cells that are then critical for immune clearance. The T-BOX family of transcription factors (TFs) are known to play a key role in T cell differentiation, with mice deficient for the TF T-BET (encoded by Tbx21) unable to generate optimal virus-specific effector responses. Although the importance of T-BET in directing optimal virus-specific T cell responses is accepted, the precise timing and molecular mechanism of action remains unclear. Using a mouse model of influenza A virus infection, we demonstrate that although T-BET is not required for early CD8+ T cell activation and cellular division, it is essential for early acquisition of virus-specific CD8+ T cell function and sustained differentiation and expansion. Whole transcriptome analysis at this early time point showed that Tbx21 deficiency resulted in global dysregulation in early programming events with inappropriate lineage-specific signatures apparent with alterations in the potential TF binding landscape. Assessment of histone posttranslational modifications within the Ifng locus demonstrated that Tbx21 -/- CD8+ T cells were unable to activate "poised" enhancer elements compared with wild-type CD8+ T cells, correlating with diminished Ifng transcription. In all, these data support a model whereby T-BET serves to promote appropriate chromatin remodeling at specific gene loci that underpins appropriate CD8+ T cell lineage-specific commitment and differentiation.
Collapse
Affiliation(s)
- Julia E Prier
- Department of Microbiology and Immunology, the Doherty Institute at the University of Melbourne, Parkville, Victoria 3010, Australia
| | - Jasmine Li
- Department of Microbiology and Immunology, the Doherty Institute at the University of Melbourne, Parkville, Victoria 3010, Australia.,Department of Microbiology, Biomedical Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Linden J Gearing
- Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia; and
| | - Moshe Olshansky
- Department of Microbiology, Biomedical Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Xavier Y X Sng
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria 3800, Australia
| | - Paul J Hertzog
- Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia; and
| | - Stephen J Turner
- Department of Microbiology and Immunology, the Doherty Institute at the University of Melbourne, Parkville, Victoria 3010, Australia; .,Department of Microbiology, Biomedical Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
| |
Collapse
|
40
|
Breed ER, Watanabe M, Hogquist KA. Measuring Thymic Clonal Deletion at the Population Level. THE JOURNAL OF IMMUNOLOGY 2019; 202:3226-3233. [PMID: 31010850 DOI: 10.4049/jimmunol.1900191] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 03/25/2019] [Indexed: 12/18/2022]
Abstract
Clonal deletion of T cells specific for self-antigens in the thymus has been widely studied, primarily by approaches that focus on a single receptor (using TCR transgenes) or a single specificity (using peptide-MHC tetramers). However, less is known about clonal deletion at the population level. In this article, we report an assay that measures cleaved caspase 3 to define clonal deletion at the population level. This assay distinguishes clonal deletion from apoptotic events caused by neglect and approximates the anatomic site of deletion using CCR7. This approach showed that 78% of clonal deletion events occur in the cortex in mice. Medullary deletion events were detected at both the semimature and mature stages, although mature events were associated with failed regulatory T cell induction. Using this assay, we showed that bone marrow-derived APC drive approximately half of deletion events at both stages. We also found that both cortical and medullary deletion rely heavily on CD28 costimulation. These findings demonstrate a useful strategy for studying clonal deletion within the polyclonal repertoire.
Collapse
Affiliation(s)
- Elise R Breed
- Department of Laboratory Medicine and Pathology, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455; and
| | - Masashi Watanabe
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Kristin A Hogquist
- Department of Laboratory Medicine and Pathology, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455; and
| |
Collapse
|
41
|
Hashimoto M, Im SJ, Araki K, Ahmed R. Cytokine-Mediated Regulation of CD8 T-Cell Responses During Acute and Chronic Viral Infection. Cold Spring Harb Perspect Biol 2019; 11:a028464. [PMID: 29101105 PMCID: PMC6314063 DOI: 10.1101/cshperspect.a028464] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The common γ-chain cytokines, interleukin (IL)-2, IL-7, and IL-15, regulate critical aspects of antiviral CD8 T-cell responses. During acute infections, IL-2 controls expansion and differentiation of antiviral CD8 T cells, whereas IL-7 and IL-15 are key cytokines to maintain memory CD8 T cells long term in an antigen-independent manner. On the other hand, during chronic infections, in which T-cell exhaustion is established, precise roles of these cytokines in regulation of antiviral CD8 T-cell responses are not well defined. Nonetheless, administration of IL-2, IL-7, or IL-15 can increase function of exhausted CD8 T cells, and thus can be an attractive therapeutic approach. A new subset of stem-cell-like CD8 T cells, which provides a proliferative burst after programmed cell death (PD)-1 therapy, has been recently described during chronic viral infection. Further understanding of cytokine-mediated regulation of this CD8 T-cell subset will improve cytokine therapies to treat chronic infections and cancer in combination with immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Masao Hashimoto
- Emory Vaccine Center and Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia 30322
| | - Se Jin Im
- Emory Vaccine Center and Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia 30322
| | - Koichi Araki
- Emory Vaccine Center and Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia 30322
| | - Rafi Ahmed
- Emory Vaccine Center and Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia 30322
| |
Collapse
|
42
|
Davenport B, Eberlein J, van der Heide V, Jhun K, Nguyen TT, Victorino F, Trotta A, Chipuk J, Yi Z, Zhang W, Clambey ET, Scott DK, Homann D. Aging of Antiviral CD8 + Memory T Cells Fosters Increased Survival, Metabolic Adaptations, and Lymphoid Tissue Homing. THE JOURNAL OF IMMUNOLOGY 2018; 202:460-475. [PMID: 30552164 DOI: 10.4049/jimmunol.1801277] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 11/04/2018] [Indexed: 02/07/2023]
Abstract
Aging of established antiviral T cell memory can foster a series of progressive adaptations that paradoxically improve rather than compromise protective CD8+ T cell immunity. We now provide evidence that this gradual evolution, the pace of which is contingent on the precise context of the primary response, also impinges on the molecular mechanisms that regulate CD8+ memory T cell (TM) homeostasis. Over time, CD8+ TM generated in the wake of an acute infection with the natural murine pathogen lymphocytic choriomeningitis virus become more resistant to apoptosis and acquire enhanced cytokine responsiveness without adjusting their homeostatic proliferation rates; concurrent metabolic adaptations promote increased CD8+ TM quiescence and fitness but also impart the reacquisition of a partial effector-like metabolic profile; and a gradual redistribution of aging CD8+ TM from blood and nonlymphoid tissues to lymphatic organs results in CD8+ TM accumulations in bone marrow, splenic white pulp, and, particularly, lymph nodes. Altogether, these data demonstrate how temporal alterations of fundamental homeostatic determinants converge to render aged CD8+ TM poised for greater recall responses.
Collapse
Affiliation(s)
- Bennett Davenport
- Department of Anesthesiology, University of Colorado Denver, Aurora, CO 80045.,Barbara Davis Center for Childhood Diabetes, University of Colorado Denver, Aurora, CO 80045.,Integrated Department of Immunology, University of Colorado Denver and National Jewish Health, Denver, CO 80045.,Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029.,Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Jens Eberlein
- Department of Anesthesiology, University of Colorado Denver, Aurora, CO 80045.,Barbara Davis Center for Childhood Diabetes, University of Colorado Denver, Aurora, CO 80045
| | - Verena van der Heide
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029.,Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Kevin Jhun
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029.,Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Tom T Nguyen
- Department of Anesthesiology, University of Colorado Denver, Aurora, CO 80045.,Barbara Davis Center for Childhood Diabetes, University of Colorado Denver, Aurora, CO 80045
| | - Francisco Victorino
- Department of Anesthesiology, University of Colorado Denver, Aurora, CO 80045.,Barbara Davis Center for Childhood Diabetes, University of Colorado Denver, Aurora, CO 80045.,Integrated Department of Immunology, University of Colorado Denver and National Jewish Health, Denver, CO 80045
| | - Andrew Trotta
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029; and
| | - Jerry Chipuk
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029; and
| | - Zhengzi Yi
- Bioinformatics Laboratory, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Weijia Zhang
- Bioinformatics Laboratory, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Eric T Clambey
- Department of Anesthesiology, University of Colorado Denver, Aurora, CO 80045.,Integrated Department of Immunology, University of Colorado Denver and National Jewish Health, Denver, CO 80045
| | - Donald K Scott
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Dirk Homann
- Department of Anesthesiology, University of Colorado Denver, Aurora, CO 80045; .,Barbara Davis Center for Childhood Diabetes, University of Colorado Denver, Aurora, CO 80045.,Integrated Department of Immunology, University of Colorado Denver and National Jewish Health, Denver, CO 80045.,Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029.,Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| |
Collapse
|
43
|
Baker RL, Jamison BL, Wiles TA, Lindsay RS, Barbour G, Bradley B, Delong T, Friedman RS, Nakayama M, Haskins K. CD4 T Cells Reactive to Hybrid Insulin Peptides Are Indicators of Disease Activity in the NOD Mouse. Diabetes 2018; 67:1836-1846. [PMID: 29976617 PMCID: PMC6110316 DOI: 10.2337/db18-0200] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 06/20/2018] [Indexed: 12/19/2022]
Abstract
We recently established that hybrid insulin peptides (HIPs), formed in islet β-cells by fusion of insulin C-peptide fragments to peptides of chromogranin A or islet amyloid polypeptide, are ligands for diabetogenic CD4 T-cell clones. The goal of this study was to investigate whether HIP-reactive T cells were indicative of ongoing autoimmunity. MHC class II tetramers were used to investigate the presence, phenotype, and function of HIP-reactive and insulin-reactive T cells in NOD mice. Insulin-reactive T cells encounter their antigen early in disease, but they express FoxP3 and therefore may contribute to immune regulation. In contrast, HIP-reactive T cells are proinflammatory and highly diabetogenic in an adoptive transfer model. Because the frequency of antigen-experienced HIP-reactive T cells increases over progression of disease, they may serve as biomarkers of autoimmune diabetes.
Collapse
MESH Headings
- Animals
- Autoantigens/chemistry
- Autoantigens/genetics
- Autoantigens/metabolism
- Autoimmune Diseases/immunology
- Autoimmune Diseases/metabolism
- Autoimmune Diseases/pathology
- Autoimmune Diseases/physiopathology
- Autoimmunity
- Biomarkers/blood
- C-Peptide/chemistry
- C-Peptide/genetics
- C-Peptide/metabolism
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- CD4-Positive T-Lymphocytes/pathology
- Cells, Cultured
- Chromogranin A/chemistry
- Chromogranin A/genetics
- Chromogranin A/metabolism
- Clone Cells
- Crosses, Genetic
- Diabetes Mellitus, Type 1/immunology
- Diabetes Mellitus, Type 1/metabolism
- Diabetes Mellitus, Type 1/pathology
- Diabetes Mellitus, Type 1/physiopathology
- Disease Progression
- Female
- Islet Amyloid Polypeptide/chemistry
- Islet Amyloid Polypeptide/genetics
- Islet Amyloid Polypeptide/metabolism
- Lymphocyte Activation
- Mice, Inbred NOD
- Mice, Knockout
- Mice, SCID
- Peptide Fragments/chemistry
- Peptide Fragments/genetics
- Peptide Fragments/metabolism
- Recombination, Genetic
- Specific Pathogen-Free Organisms
Collapse
Affiliation(s)
- Rocky L Baker
- Department of Immunology and Microbiology, University of Colorado School of Medicine at Denver, Aurora, CO
| | - Braxton L Jamison
- Department of Immunology and Microbiology, University of Colorado School of Medicine at Denver, Aurora, CO
| | - Timothy A Wiles
- Department of Immunology and Microbiology, University of Colorado School of Medicine at Denver, Aurora, CO
| | - Robin S Lindsay
- Department of Biomedical Research, National Jewish Health, Denver, CO
| | - Gene Barbour
- Department of Immunology and Microbiology, University of Colorado School of Medicine at Denver, Aurora, CO
| | - Brenda Bradley
- Department of Immunology and Microbiology, University of Colorado School of Medicine at Denver, Aurora, CO
| | - Thomas Delong
- Department of Immunology and Microbiology, University of Colorado School of Medicine at Denver, Aurora, CO
| | - Rachel S Friedman
- Department of Biomedical Research, National Jewish Health, Denver, CO
| | - Maki Nakayama
- Barbara Davis Center for Childhood Diabetes, University of Colorado School of Medicine at Denver, Aurora, CO
| | - Kathryn Haskins
- Department of Immunology and Microbiology, University of Colorado School of Medicine at Denver, Aurora, CO
| |
Collapse
|
44
|
Keating R, Morris MY, Yue W, Reynolds CE, Harris TL, Brown SA, Doherty PC, Thomas PG, McGargill MA. Potential killers exposed: tracking endogenous influenza-specific CD8 + T cells. Immunol Cell Biol 2018; 96:1104-1119. [PMID: 29972699 PMCID: PMC6282960 DOI: 10.1111/imcb.12189] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Revised: 07/01/2018] [Accepted: 07/02/2018] [Indexed: 12/31/2022]
Abstract
Current influenza A virus (IAV) vaccines stimulate antibody responses that are directed against variable regions of the virus, and are therefore ineffective against divergent strains. As CD8+ T cells target the highly conserved, internal IAV proteins, they have the potential to increase heterosubtypic immunity. Early T‐cell priming events influence lasting memory, which is required for long‐term protection. However, the early responding, IAV‐specific cells are difficult to monitor because of their low frequencies. Here, we tracked the dissemination of endogenous IAV‐specific CD8+ T cells during the initial phases of the immune response following IAV infection. We exposed a significant population of recently activated, CD25+CD43+ IAV‐specific T cells that were not detected by tetramer staining. By tracking this population, we found that initial T‐cell priming occurred in the mediastinal lymph nodes, which gave rise to the most expansive IAV‐specific CD8+ T‐cell population. Subsequently, IAV‐specific CD8+ T cells dispersed to the bronchoalveolar lavage and blood, followed by spleen and liver, and finally to the lung. These data provide important insight into the priming and tissue dispersion of an endogenous CD8+ T‐cell response. Importantly, the CD25+CD43+ phenotype identifies an inclusive population of early responding CD8+ T cells, which may provide insight into TCR repertoire selection and expansion. A better understanding of this response is critical for designing improved vaccines that target CD8+ T cells.
Collapse
Affiliation(s)
- Rachael Keating
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Melissa Y Morris
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Wen Yue
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Cory E Reynolds
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Tarsha L Harris
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Scott A Brown
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Peter C Doherty
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA.,Department of Microbiology and Immunology, University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Parkville, VIC, 3010, Australia
| | - Paul G Thomas
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Maureen A McGargill
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| |
Collapse
|
45
|
Listeria Monocytogenes: A Model Pathogen Continues to Refine Our Knowledge of the CD8 T Cell Response. Pathogens 2018; 7:pathogens7020055. [PMID: 29914156 PMCID: PMC6027175 DOI: 10.3390/pathogens7020055] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2018] [Revised: 06/13/2018] [Accepted: 06/14/2018] [Indexed: 12/12/2022] Open
Abstract
Listeria monocytogenes (Lm) infection induces robust CD8 T cell responses, which play a critical role in resolving Lm during primary infection and provide protective immunity to re-infections. Comprehensive studies have been conducted to delineate the CD8 T cell response after Lm infection. In this review, the generation of the CD8 T cell response to Lm infection will be discussed. The role of dendritic cell subsets in acquiring and presenting Lm antigens to CD8 T cells and the events that occur during T cell priming and activation will be addressed. CD8 T cell expansion, differentiation and contraction as well as the signals that regulate these processes during Lm infection will be explored. Finally, the formation of memory CD8 T cell subsets in the circulation and in the intestine will be analyzed. Recently, the study of CD8 T cell responses to Lm infection has begun to shift focus from the intravenous infection model to a natural oral infection model as the humanized mouse and murinized Lm have become readily available. Recent findings in the generation of CD8 T cell responses to oral infection using murinized Lm will be explored throughout the review. Finally, CD8 T cell-mediated protective immunity against Lm infection and the use of Lm as a vaccine vector for cancer immunotherapy will be highlighted. Overall, this review will provide detailed knowledge on the biology of CD8 T cell responses after Lm infection that may shed light on improving rational vaccine design.
Collapse
|
46
|
Polonsky M, Rimer J, Kern-Perets A, Zaretsky I, Miller S, Bornstein C, David E, Kopelman NM, Stelzer G, Porat Z, Chain B, Friedman N. Induction of CD4 T cell memory by local cellular collectivity. Science 2018; 360:360/6394/eaaj1853. [DOI: 10.1126/science.aaj1853] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Revised: 12/19/2017] [Accepted: 04/17/2018] [Indexed: 12/11/2022]
Abstract
Cell differentiation is directed by signals driving progenitors into specialized cell types. This process can involve collective decision-making, when differentiating cells determine their lineage choice by interacting with each other. We used live-cell imaging in microwell arrays to study collective processes affecting differentiation of naïve CD4+ T cells into memory precursors. We found that differentiation of precursor memory T cells sharply increases above a threshold number of locally interacting cells. These homotypic interactions involve the cytokines interleukin-2 (IL-2) and IL-6, which affect memory differentiation orthogonal to their effect on proliferation and survival. Mathematical modeling suggests that the differentiation rate is continuously modulated by the instantaneous number of locally interacting cells. This cellular collectivity can prioritize allocation of immune memory to stronger responses.
Collapse
|
47
|
Chiaranunt P, Tometich JT, Ji J, Hand TW. T Cell Proliferation and Colitis Are Initiated by Defined Intestinal Microbes. THE JOURNAL OF IMMUNOLOGY 2018; 201:243-250. [PMID: 29777027 DOI: 10.4049/jimmunol.1800236] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 04/23/2018] [Indexed: 12/22/2022]
Abstract
Inflammatory bowel disease has been associated with the dysregulation of T cells specific to Ags derived from the intestinal microbiota. How microbiota-specific T cells are regulated is not completely clear but is believed to be mediated by a combination of IgA, regulatory T cells, and type 3 innate lymphoid cells. To test the role of these regulatory components on microbiota-specific T cells, we bred CBir1 TCR transgenic (CBir1Tg) mice (specific to flagellin from common intestinal bacteria) onto a lymphopenic Rag1-/- background. Surprisingly, T cells from CBir1Tg mice bred onto a Rag1-/- background could not induce colitis and did not differentiate to become effectors under lymphopenic conditions, despite deficits in immunoregulatory factors, such as IgA, regulatory T cells, and type 3 innate lymphoid cells. In fact, upon transfer of conventional CBir1Tg T cells into lymphopenic mice, the vast majority of proliferating T cells responded to Ags other than CBir1 flagellin, including those found on other bacteria, such as Helicobacter spp. Thus, we discovered a caveat in the CBir1Tg model within our animal facility that illustrates the limitations of using TCR transgenics at mucosal surfaces, where multiple TCR specificities can respond to the plethora of foreign Ags. Our findings also indicate that T cell specificity to the microbiota alone is not sufficient to induce T cell activation and colitis. Instead, other interrelated factors, such as the composition and ecology of the intestinal microbiota and host access to Ag, are paramount in controlling the activation of microbiota-specific T cell clones.
Collapse
Affiliation(s)
- Pailin Chiaranunt
- Richard King Mellon Foundation Institute for Pediatric Research, Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh, Pittsburgh, PA 15224.,Department of Pediatrics, University of Pittsburgh Medical School, Pittsburgh, PA 15224
| | - Justin T Tometich
- Richard King Mellon Foundation Institute for Pediatric Research, Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh, Pittsburgh, PA 15224.,Department of Pediatrics, University of Pittsburgh Medical School, Pittsburgh, PA 15224
| | - Junyi Ji
- Richard King Mellon Foundation Institute for Pediatric Research, Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh, Pittsburgh, PA 15224.,Department of Pediatrics, University of Pittsburgh Medical School, Pittsburgh, PA 15224.,School of Medicine, Tsinghua University, Beijing 100084, China; and
| | - Timothy W Hand
- Richard King Mellon Foundation Institute for Pediatric Research, Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh, Pittsburgh, PA 15224; .,Department of Pediatrics, University of Pittsburgh Medical School, Pittsburgh, PA 15224.,Department of Immunology, University of Pittsburgh Medical School, Pittsburgh, PA 15213
| |
Collapse
|
48
|
Imaging of cytotoxic antiviral immunity while considering the 3R principle of animal research. J Mol Med (Berl) 2018; 96:349-360. [PMID: 29460050 DOI: 10.1007/s00109-018-1628-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 02/01/2018] [Accepted: 02/05/2018] [Indexed: 10/18/2022]
Abstract
Adoptive cell transfer approaches for antigen-specific CD8+ T cells are used widely to study their effector potential during infections or cancer. However, contemporary methodological adaptations regarding transferred cell numbers, advanced imaging, and the 3R principle of animal research have been largely omitted. Here, we introduce an improved cell transfer method that reduces the number of donor animals substantially and fulfills the requirements for intravital imaging under physiological conditions. For this, we analyzed the well-established Friend retrovirus (FV) mouse model. Donor mice that expressed a FV-specific T cell receptor (TCRtg) and the fluorescent protein tdTomato were used as source of antigen-specific CD8+ T cells. Only a few drops of peripheral blood were sufficient to isolate ~ 150,000 naive reporter cells from which 1000 were adoptively transferred into recently FV-infected recipients. The cells became activated and functional and expanded strongly in the spleen and bone marrow within 10 days post infection. Transferred CD8+ T cells participated in the antiviral host response within a natural range and developed an effector phenotype indistinguishable from endogenous effector CD8+ T cells. Additionally, the generated reporter cell frequency allowed single cell visualization and tracking of a physiological antiretroviral CD8+ T cell response by intravital two-photon microscopy. Highly reproducible results were obtained in independent experiments by reusing the same donors repetitively for multiple transfers. Our approach allows a strong reduction of experimental animals required for studies on antigen-specific CD8+ T cell function and should be applicable to other transfer models. KEY MESSAGES TCRtg CD8+ T cells are obtained repetitively from the blood samples of single donors. One thousand transferred TCRtg CD8+ T cells get activated, are functional, and proliferate. Several adoptive cell transfers from the same donor show reproducible results. One thousand transferred cells take part in the FV immune response without modifying it. Use of fluorescent transfer cells allows in vivo imaging and single cell tracking.
Collapse
|
49
|
Acetylation of the Cd8 Locus by KAT6A Determines Memory T Cell Diversity. Cell Rep 2018; 16:3311-3321. [PMID: 27653692 DOI: 10.1016/j.celrep.2016.08.056] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Revised: 07/21/2016] [Accepted: 08/17/2016] [Indexed: 11/20/2022] Open
Abstract
How functionally diverse populations of pathogen-specific killer T cells are generated during an immune response remains unclear. Here, we propose that fine-tuning of CD8αβ co-receptor levels via histone acetylation plays a role in lineage fate. We show that lysine acetyltransferase 6A (KAT6A) is responsible for maintaining permissive Cd8 gene transcription and enabling robust effector responses during infection. KAT6A-deficient CD8(+) T cells downregulated surface CD8 co-receptor expression during clonal expansion, a finding linked to reduced Cd8α transcripts and histone-H3 lysine 9 acetylation of the Cd8 locus. Loss of CD8 expression in KAT6A-deficient T cells correlated with reduced TCR signaling intensity and accelerated contraction of the effector-like memory compartment, whereas the long-lived memory compartment appeared unaffected, a result phenocopied by the removal of the Cd8 E8I enhancer element. These findings suggest a direct role of CD8αβ co-receptor expression and histone acetylation in shaping functional diversity within the cytotoxic T cell pool.
Collapse
|
50
|
Lee LN, Bolinger B, Banki Z, de Lara C, Highton AJ, Colston JM, Hutchings C, Klenerman P. Adenoviral vaccine induction of CD8+ T cell memory inflation: Impact of co-infection and infection order. PLoS Pathog 2017; 13:e1006782. [PMID: 29281733 PMCID: PMC5760110 DOI: 10.1371/journal.ppat.1006782] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 01/09/2018] [Accepted: 11/29/2017] [Indexed: 12/14/2022] Open
Abstract
The efficacies of many new T cell vaccines rely on generating large populations of long-lived pathogen-specific effector memory CD8 T cells. However, it is now increasingly recognized that prior infection history impacts on the host immune response. Additionally, the order in which these infections are acquired could have a major effect. Exploiting the ability to generate large sustained effector memory (i.e. inflationary) T cell populations from murine cytomegalovirus (MCMV) and human Adenovirus-subtype (AdHu5) 5-beta-galactosidase (Ad-lacZ) vector, the impact of new infections on pre-existing memory and the capacity of the host's memory compartment to accommodate multiple inflationary populations from unrelated pathogens was investigated in a murine model. Simultaneous and sequential infections, first with MCMV followed by Ad-lacZ, generated inflationary populations towards both viruses with similar kinetics and magnitude to mono-infected groups. However, in Ad-lacZ immune mice, subsequent acute MCMV infection led to a rapid decline of the pre-existing Ad-LacZ-specific inflating population, associated with bystander activation of Fas-dependent apoptotic pathways. However, responses were maintained long-term and boosting with Ad-lacZ led to rapid re-expansion of the inflating population. These data indicate firstly that multiple specificities of inflating memory cells can be acquired at different times and stably co-exist. Some acute infections may also deplete pre-existing memory populations, thus revealing the importance of the order of infection acquisition. Importantly, immunization with an AdHu5 vector did not alter the size of the pre-existing memory. These phenomena are relevant to the development of adenoviral vectors as novel vaccination strategies for diverse infections and cancers. (241 words).
Collapse
MESH Headings
- Adenovirus Infections, Human/immunology
- Adenovirus Infections, Human/prevention & control
- Adenoviruses, Human/genetics
- Adenoviruses, Human/immunology
- Adenoviruses, Human/pathogenicity
- Animals
- CD8-Positive T-Lymphocytes/immunology
- Coinfection/immunology
- Coinfection/prevention & control
- Epitopes, T-Lymphocyte/genetics
- Epitopes, T-Lymphocyte/immunology
- Herpesviridae Infections/immunology
- Herpesviridae Infections/prevention & control
- Host-Pathogen Interactions/immunology
- Humans
- Immunologic Memory
- Lac Operon
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Knockout
- Models, Immunological
- Muromegalovirus/genetics
- Muromegalovirus/immunology
- Muromegalovirus/pathogenicity
- Receptors, Interleukin-18/deficiency
- Receptors, Interleukin-18/genetics
- Receptors, Interleukin-18/immunology
- Viral Vaccines/genetics
- Viral Vaccines/immunology
Collapse
Affiliation(s)
- Lian N. Lee
- Peter Medawar Building and Translational Gastroenterology Unit, Oxford, United Kingdom
| | - Beatrice Bolinger
- Peter Medawar Building and Translational Gastroenterology Unit, Oxford, United Kingdom
- Schweizerischer Apothekerverband, pharmaSuisse, Bern, Switzerland
| | - Zoltan Banki
- Peter Medawar Building and Translational Gastroenterology Unit, Oxford, United Kingdom
- Division of Virology, Innsbruck Medical University, Innsbruck, Austria
| | - Catherine de Lara
- Peter Medawar Building and Translational Gastroenterology Unit, Oxford, United Kingdom
| | - Andrew J. Highton
- Peter Medawar Building and Translational Gastroenterology Unit, Oxford, United Kingdom
| | - Julia M. Colston
- Peter Medawar Building and Translational Gastroenterology Unit, Oxford, United Kingdom
| | - Claire Hutchings
- Peter Medawar Building and Translational Gastroenterology Unit, Oxford, United Kingdom
| | - Paul Klenerman
- Peter Medawar Building and Translational Gastroenterology Unit, Oxford, United Kingdom
| |
Collapse
|