1
|
Maurice NJ, Erickson JR, DeJong CS, Mair F, Taber AK, Frutoso M, Islas LV, Vigil ALB, Lawler RL, McElrath MJ, Newell EW, Sullivan LB, Shree R, McCartney SA. Converging cytokine and metabolite networks shape asymmetric T cell fate at the term human maternal-fetal interface. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.10.598377. [PMID: 38915597 PMCID: PMC11195144 DOI: 10.1101/2024.06.10.598377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Placentation presents immune conflict between mother and fetus, yet in normal pregnancy maternal immunity against infection is maintained without expense to fetal tolerance. This is believed to result from adaptations at the maternal-fetal interface (MFI) which affect T cell programming, but the identities (i.e., memory subsets and antigenic specificities) of T cells and the signals that mediate T cell fates and functions at the MFI remain poorly understood. We found intact recruitment programs as well as pro-inflammatory cytokine networks that can act on maternal T cells in an antigen-independent manner. These inflammatory signals elicit T cell expression of co-stimulatory receptors necessary for tissue retention, which can be engaged by local macrophages. Although pro-inflammatory molecules elicit T cell effector functions, we show that additional cytokine (TGF-β1) and metabolite (kynurenine) networks may converge to tune T cell function to those of sentinels. Together, we demonstrate an additional facet of fetal tolerance, wherein T cells are broadly recruited and restrained in an antigen-independent, cytokine/metabolite-dependent manner. These mechanisms provide insight into antigen-nonspecific T cell regulation, especially in tissue microenvironments where they are enriched.
Collapse
Affiliation(s)
- Nicholas J Maurice
- Molecular and Cellular Biology Graduate Program, University of Washington, Seattle, WA
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Jami R Erickson
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Caitlin S DeJong
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Florian Mair
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Alexis K Taber
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Marie Frutoso
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Laura V Islas
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA
| | | | - Richard L Lawler
- Immune Monitoring Core, Fred Hutchinson Cancer Center, Seattle, WA
| | - M Juliana McElrath
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA
- Department of Medicine, University of Washington, Seattle, WA
| | - Evan W Newell
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Lucas B Sullivan
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Raj Shree
- Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, University of Washington, Seattle, WA
| | - Stephen A McCartney
- Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, University of Washington, Seattle, WA
| |
Collapse
|
2
|
Rakebrandt N, Yassini N, Kolz A, Schorer M, Lambert K, Goljat E, Estrada Brull A, Rauld C, Balazs Z, Krauthammer M, Carballido JM, Peters A, Joller N. Innate acting memory Th1 cells modulate heterologous diseases. Proc Natl Acad Sci U S A 2024; 121:e2312837121. [PMID: 38838013 PMCID: PMC11181110 DOI: 10.1073/pnas.2312837121] [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: 07/26/2023] [Accepted: 05/08/2024] [Indexed: 06/07/2024] Open
Abstract
Through immune memory, infections have a lasting effect on the host. While memory cells enable accelerated and enhanced responses upon rechallenge with the same pathogen, their impact on susceptibility to unrelated diseases is unclear. We identify a subset of memory T helper 1 (Th1) cells termed innate acting memory T (TIA) cells that originate from a viral infection and produce IFN-γ with innate kinetics upon heterologous challenge in vivo. Activation of memory TIA cells is induced in response to IL-12 in combination with IL-18 or IL-33 but is TCR independent. Rapid IFN-γ production by memory TIA cells is protective in subsequent heterologous challenge with the bacterial pathogen Legionella pneumophila. In contrast, antigen-independent reactivation of CD4+ memory TIA cells accelerates disease onset in an autoimmune model of multiple sclerosis. Our findings demonstrate that memory Th1 cells can acquire additional TCR-independent functionality to mount rapid, innate-like responses that modulate susceptibility to heterologous challenges.
Collapse
Affiliation(s)
- Nikolas Rakebrandt
- Institute of Experimental Immunology, University of Zurich, 8057Zurich, Switzerland
| | - Nima Yassini
- Institute of Experimental Immunology, University of Zurich, 8057Zurich, Switzerland
- Department of Quantitative Biomedicine, University of Zurich, 8057Zurich, Switzerland
| | - Anna Kolz
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig-Maximilians-Universität München, 82152Planegg, Germany
| | - Michelle Schorer
- Institute of Experimental Immunology, University of Zurich, 8057Zurich, Switzerland
| | - Katharina Lambert
- Institute of Experimental Immunology, University of Zurich, 8057Zurich, Switzerland
| | - Eva Goljat
- Department of Quantitative Biomedicine, University of Zurich, 8057Zurich, Switzerland
| | - Anna Estrada Brull
- Department of Quantitative Biomedicine, University of Zurich, 8057Zurich, Switzerland
| | - Celine Rauld
- Novartis Biomedical Research, 4002Basel, Switzerland
| | - Zsolt Balazs
- Department of Quantitative Biomedicine, University of Zurich, 8057Zurich, Switzerland
| | - Michael Krauthammer
- Department of Quantitative Biomedicine, University of Zurich, 8057Zurich, Switzerland
| | | | - Anneli Peters
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig-Maximilians-Universität München, 82152Planegg, Germany
- Biomedical Center, Faculty of Medicine, Ludwig-Maximilians-Universität München, 82152Planegg, Germany
| | - Nicole Joller
- Institute of Experimental Immunology, University of Zurich, 8057Zurich, Switzerland
- Department of Quantitative Biomedicine, University of Zurich, 8057Zurich, Switzerland
| |
Collapse
|
3
|
Mambetsariev N, Torres Acosta MA, Liu Q, Flores CPR, Joudi AM, Helmin KA, Gurkan JK, Steinert EM, Morales-Nebreda L, Singer BD. FOXP3+ Regulatory T Cells Require TBET to Regulate Activated CD8+ T Cells During Recovery from Influenza Infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.30.596295. [PMID: 38853959 PMCID: PMC11160713 DOI: 10.1101/2024.05.30.596295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
FOXP3+ regulatory T (Treg) cells are necessary to coordinate resolution of lung inflammation and a return to homeostasis after respiratory viral infections, but the specific molecular requirements for these functions and the cell types governed by Treg cells remain unclear. This question holds significance as clinical trials of Treg cell transfer therapy for respiratory viral infection are being planned and executed. Here, we report causal experiments in mice determining that Treg cells are necessary to control the numbers of activated CD8+ T cells during recovery from influenza infection. Using a genetic strategy paired with adoptive transfer techniques, we determined that Treg cells require the transcription factor TBET to regulate these potentially pro-inflammatory CD8+ T cells. Surprisingly, we found that Treg cells are dispensable for the generation of CD8+ lung tissue resident-memory T (Trm) cells yet similarly influence the transcriptional programming of CD8+ Trm and activated T cells. Our study highlights the role of Treg cells in regulating the CD8+ T cell response during recovery from influenza infection.
Collapse
Affiliation(s)
- Nurbek Mambetsariev
- Division of Pulmonary and Critical Care Medicine, Northwestern
University Feinberg School of Medicine, Chicago, IL 60611 USA
- Division of Allergy and Immunology, Northwestern University
Feinberg School of Medicine, Chicago, IL 60611 USA
| | - Manuel A. Torres Acosta
- Division of Pulmonary and Critical Care Medicine, Northwestern
University Feinberg School of Medicine, Chicago, IL 60611 USA
- Medical Scientist Training Program, Northwestern University
Feinberg School of Medicine, Chicago, IL 60611 USA
- Driskill Graduate Program, Northwestern University Feinberg
School of Medicine, Chicago, IL 60611 USA
| | - Qianli Liu
- Division of Pulmonary and Critical Care Medicine, Northwestern
University Feinberg School of Medicine, Chicago, IL 60611 USA
- Driskill Graduate Program, Northwestern University Feinberg
School of Medicine, Chicago, IL 60611 USA
| | - Carla P. Reyes Flores
- Division of Pulmonary and Critical Care Medicine, Northwestern
University Feinberg School of Medicine, Chicago, IL 60611 USA
- Driskill Graduate Program, Northwestern University Feinberg
School of Medicine, Chicago, IL 60611 USA
| | - Anthony M. Joudi
- Division of Pulmonary and Critical Care Medicine, Northwestern
University Feinberg School of Medicine, Chicago, IL 60611 USA
| | - Kathryn A. Helmin
- Division of Pulmonary and Critical Care Medicine, Northwestern
University Feinberg School of Medicine, Chicago, IL 60611 USA
| | - Jonathan K. Gurkan
- Division of Pulmonary and Critical Care Medicine, Northwestern
University Feinberg School of Medicine, Chicago, IL 60611 USA
- Medical Scientist Training Program, Northwestern University
Feinberg School of Medicine, Chicago, IL 60611 USA
- Driskill Graduate Program, Northwestern University Feinberg
School of Medicine, Chicago, IL 60611 USA
| | - Elizabeth M. Steinert
- Division of Pulmonary and Critical Care Medicine, Northwestern
University Feinberg School of Medicine, Chicago, IL 60611 USA
| | - Luisa Morales-Nebreda
- Division of Pulmonary and Critical Care Medicine, Northwestern
University Feinberg School of Medicine, Chicago, IL 60611 USA
| | - Benjamin D. Singer
- Division of Pulmonary and Critical Care Medicine, Northwestern
University Feinberg School of Medicine, Chicago, IL 60611 USA
- Department of Biochemistry and Molecular Genetics, Northwestern
University Feinberg School of Medicine, Chicago, IL 60611 USA
- Simpson Querrey Institute for Epigenetics, Northwestern
University Feinberg School of Medicine, Chicago, IL 60611 USA
- Simpson Querrey Lung Institute for Translational Science (SQ
LIFTS), Northwestern University Feinberg School of Medicine, Chicago, IL 60611 USA
| |
Collapse
|
4
|
Marchesini Tovar G, Gallen C, Bergsbaken T. CD8+ Tissue-Resident Memory T Cells: Versatile Guardians of the Tissue. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:361-368. [PMID: 38227907 PMCID: PMC10794029 DOI: 10.4049/jimmunol.2300399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 09/07/2023] [Indexed: 01/18/2024]
Abstract
Tissue-resident memory T (Trm) cells are a subset of T cells maintained throughout life within nonlymphoid tissues without significant contribution from circulating memory T cells. CD8+ Trm cells contribute to both tissue surveillance and direct elimination of pathogens through a variety of mechanisms. Reactivation of these Trm cells during infection drives systematic changes within the tissue, including altering the state of the epithelium, activating local immune cells, and contributing to the permissiveness of the tissue for circulating immune cell entry. Trm cells can be further classified by their functional outputs, which can be either subset- or tissue-specific, and include proliferation, tissue egress, and modulation of tissue physiology. These functional outputs of Trm cells are linked to the heterogeneity and plasticity of this population, and uncovering the unique responses of different Trm cell subsets and their role in immunity will allow us to modulate Trm cell responses for optimal control of disease.
Collapse
Affiliation(s)
- Giuseppina Marchesini Tovar
- Center for Immunity and Inflammation, Department of Pathology, Immunology, and Laboratory Medicine, Rutgers New Jersey Medical School, Newark, NJ
| | - Corey Gallen
- Center for Immunity and Inflammation, Department of Pathology, Immunology, and Laboratory Medicine, Rutgers New Jersey Medical School, Newark, NJ
| | - Tessa Bergsbaken
- Center for Immunity and Inflammation, Department of Pathology, Immunology, and Laboratory Medicine, Rutgers New Jersey Medical School, Newark, NJ
| |
Collapse
|
5
|
Zhou J, Uddback I, Kohlmeier JE, Christensen JP, Thomsen AR. Vaccine induced memory CD8 + T cells efficiently prevent viral transmission from the respiratory tract. Front Immunol 2023; 14:1322536. [PMID: 38164135 PMCID: PMC10757911 DOI: 10.3389/fimmu.2023.1322536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 11/15/2023] [Indexed: 01/03/2024] Open
Abstract
Introduction Mucosal immunization eliciting local T-cell memory has been suggested for improved protection against respiratory infections caused by viral variants evading pre-existing antibodies. However, it remains unclear whether T-cell targeted vaccines suffice for prevention of viral transmission and to which extent local immunity is important in this context. Methods To study the impact of T-cell vaccination on the course of viral respiratory infection and in particular the capacity to inhibit viral transmission, we used a mouse model involving natural murine parainfluenza infection with a luciferase encoding virus and an adenovirus based nucleoprotein targeting vaccine. Results and discussion Prior intranasal immunization inducing strong mucosal CD8+ T cell immunity provided an almost immediate shut-down of the incipient infection and completely inhibited contact based viral spreading. If this first line of defense did not operate, as in parentally immunized mice, recirculating T cells participated in accelerated viral control that reduced the intensity of inter-individual transmission. These observations underscore the importance of pursuing the development of mucosal T-cell inducing vaccines for optimal protection of the individual and inhibition of inter-individual transmission (herd immunity), while at the same time explain why induction of a strong systemic T-cell response may still impact viral transmission.
Collapse
Affiliation(s)
- Jinglin Zhou
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Ida Uddback
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Jacob E. Kohlmeier
- Department of Microbiology and Immunology, Emory University, Atlanta, GA, United States
| | | | - Allan Randrup Thomsen
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
6
|
Liu C, Liu T, Hu Y, Zeng X, Alimu X, Song S, Lu S, Song Y, Wang P. G Protein-Coupled Receptor 56 Characterizes CTLs and Reflects the Progression of Lung Cancer Patients. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:683-692. [PMID: 37378668 DOI: 10.4049/jimmunol.2101048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 06/02/2023] [Indexed: 06/29/2023]
Abstract
CTLs play important roles in host immune responses to tumors. CD4 CTLs are characterized by their ability to secrete cytotoxic effector molecules, such as granzyme B and perforin, and kill target cells in a MHC class II-restricted manner. However, the cell surface markers of CD4 CTLs remain unknown, which hinders their separation and research on their function. In this study, we performed a bioinformatics analysis and experimental validation that revealed that G protein-coupled receptor 56 (GPR56) is a cell surface marker that can be used to characterize CD4 CTLs. We found that GPR56 and granzyme B were coexpressed in extremely high levels in human peripheral blood T cells, and that anti-GPR56 stimulation significantly upregulated the expression of granzyme B in both CD4+GPR56+ and CD8+GPR56+ T cells. These findings suggest that GPR56 expression and the GPR56 signaling pathway could contribute directly to the toxic function of either CD4+ or CD8+ T cells. We also used GPR56 as a biomarker to investigate the clinical significance of CD4 CTLs. GPR56+ T cell levels were increased in patients with lung cancer, and GPR56 expression was significantly correlated with lung cancer progression. A further analysis revealed an increase in exhausted cell states in lung cancer patients because of upregulation of programmed cell death protein 1 expression in GPR56+ T cells. The findings of this study suggest that GPR56 characterizes the cytotoxic states of either CD4+ or CD8+ T cells.
Collapse
Affiliation(s)
- Chen Liu
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, China
| | - Tianci Liu
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, China
| | - Yuzhe Hu
- Department of Immunology, NHC Key Laboratory of Medical Immunology (Peking University), School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- Peking University Center for Human Disease Genomics, Peking University Health Science Center, Beijing, China
| | - Xingyue Zeng
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, China
| | - Xiayidan Alimu
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, China
| | - Shi Song
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, China
| | - Songsong Lu
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, China
| | - Ying Song
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, China
| | - Pingzhang Wang
- Department of Immunology, NHC Key Laboratory of Medical Immunology (Peking University), School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- Peking University Center for Human Disease Genomics, Peking University Health Science Center, Beijing, China
| |
Collapse
|
7
|
Hallisey VM, Schwab SR. Get me out of here: Sphingosine 1-phosphate signaling and T cell exit from tissues during an immune response. Immunol Rev 2023; 317:8-19. [PMID: 37212181 DOI: 10.1111/imr.13219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 05/03/2023] [Indexed: 05/23/2023]
Abstract
During an immune response, the duration of T cell residence in lymphoid and non-lymphoid tissues likely affects T cell activation, differentiation, and memory development. The factors that govern T cell transit through inflamed tissues remain incompletely understood, but one important determinant of T cell exit from tissues is sphingosine 1-phosphate (S1P) signaling. In homeostasis, S1P levels are high in blood and lymph compared to lymphoid organs, and lymphocytes follow S1P gradients out of tissues into circulation using varying combinations of five G-protein coupled S1P receptors. During an immune response, both the shape of S1P gradients and the expression of S1P receptors are dynamically regulated. Here we review what is known, and key questions that remain unanswered, about how S1P signaling is regulated in inflammation and in turn how S1P shapes immune responses.
Collapse
Affiliation(s)
- Victoria M Hallisey
- Department of Cell Biology, New York University Grossman School of Medicine, New York, New York, USA
| | - Susan R Schwab
- Department of Cell Biology, New York University Grossman School of Medicine, New York, New York, USA
| |
Collapse
|
8
|
Damei I, Trickovic T, Mami-Chouaib F, Corgnac S. Tumor-resident memory T cells as a biomarker of the response to cancer immunotherapy. Front Immunol 2023; 14:1205984. [PMID: 37545498 PMCID: PMC10399960 DOI: 10.3389/fimmu.2023.1205984] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 07/03/2023] [Indexed: 08/08/2023] Open
Abstract
Tumor-infiltrating lymphocytes (TIL) often include a substantial subset of CD8+ tissue-resident memory T (TRM) cells enriched in tumor-specific T cells. These TRM cells play a major role in antitumor immune response. They are identified on the basis of their expression of the CD103 (αE(CD103)β7) and/or CD49a (α1(CD49a)β1) integrins, and the C-type lectin CD69, which are involved in tissue residency. TRM cells express several T-cell inhibitory receptors on their surface but they nevertheless react strongly to malignant cells, exerting a strong cytotoxic function, particularly in the context of blocking interactions of PD-1 with PD-L1 on target cells. These TRM cells form stable conjugates with autologous tumor cells and interact with dendritic cells and other T cells within the tumor microenvironment to orchestrate an optimal in situ T-cell response. There is growing evidence to indicate that TGF-β is essential for the formation and maintenance of TRM cells in the tumor, through the induction of CD103 expression on activated CD8+ T cells, and for the regulation of TRM effector functions through bidirectional integrin signaling. CD8+ TRM cells were initially described as a prognostic marker for survival in patients with various types of cancer, including ovarian, lung and breast cancers and melanoma. More recently, these tumor-resident CD8+ T cells have been shown to be a potent predictive biomarker of the response of cancer patients to immunotherapies, including therapeutic cancer vaccines and immune checkpoint blockade. In this review, we will highlight the major characteristics of tumor TRM cell populations and the possibilities for their exploitation in the design of more effective immunotherapy strategies for cancer.
Collapse
|
9
|
Jung IY, Noguera-Ortega E, Bartoszek R, Collins SM, Williams E, Davis M, Jadlowsky JK, Plesa G, Siegel DL, Chew A, Levine BL, Berger SL, Moon EK, Albelda SM, Fraietta JA. Tissue-resident memory CAR T cells with stem-like characteristics display enhanced efficacy against solid and liquid tumors. Cell Rep Med 2023; 4:101053. [PMID: 37224816 PMCID: PMC10313923 DOI: 10.1016/j.xcrm.2023.101053] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 02/21/2023] [Accepted: 04/27/2023] [Indexed: 05/26/2023]
Abstract
Chimeric antigen receptor (CAR) T cells demonstrate remarkable success in treating hematological malignancies, but their effectiveness in non-hematopoietic cancers remains limited. This study proposes enhancing CAR T cell function and localization in solid tumors by modifying the epigenome governing tissue-residency adaptation and early memory differentiation. We identify that a key factor in human tissue-resident memory CAR T cell (CAR-TRM) formation is activation in the presence of the pleotropic cytokine, transforming growth factor β (TGF-β), which enforces a core program of both "stemness" and sustained tissue residency by mediating chromatin remodeling and concurrent transcriptional changes. This approach leads to a practical and clinically actionable in vitro production method for engineering peripheral blood T cells into a large number of "stem-like" CAR-TRM cells resistant to tumor-associated dysfunction, possessing an enhanced ability to accumulate in situ and rapidly eliminate cancer cells for more effective immunotherapy.
Collapse
Affiliation(s)
- In-Young Jung
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Estela Noguera-Ortega
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Robert Bartoszek
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sierra M Collins
- Parker Institute for Cancer Immunotherapy, University of Pennsylvania, Philadelphia, PA 19104, USA; Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Erik Williams
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Megan Davis
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Julie K Jadlowsky
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Gabriela Plesa
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Donald L Siegel
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Anne Chew
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Bruce L Levine
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Shelley L Berger
- Parker Institute for Cancer Immunotherapy, University of Pennsylvania, Philadelphia, PA 19104, USA; Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Edmund K Moon
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Steven M Albelda
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Joseph A Fraietta
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
10
|
Qiu Z, Khairallah C, Chu TH, Imperato JN, Lei X, Romanov G, Atakilit A, Puddington L, Sheridan BS. Retinoic acid signaling during priming licenses intestinal CD103+ CD8 TRM cell differentiation. J Exp Med 2023; 220:e20210923. [PMID: 36809399 PMCID: PMC9960115 DOI: 10.1084/jem.20210923] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 12/02/2022] [Accepted: 02/01/2023] [Indexed: 02/23/2023] Open
Abstract
CD8 tissue-resident memory T (TRM) cells provide frontline protection at barrier tissues; however, mechanisms regulating TRM cell development are not completely understood. Priming dictates the migration of effector T cells to the tissue, while factors in the tissue induce in situ TRM cell differentiation. Whether priming also regulates in situ TRM cell differentiation uncoupled from migration is unclear. Here, we demonstrate that T cell priming in the mesenteric lymph nodes (MLN) regulates CD103+ TRM cell differentiation in the intestine. In contrast, T cells primed in the spleen were impaired in the ability to differentiate into CD103+ TRM cells after entry into the intestine. MLN priming initiated a CD103+ TRM cell gene signature and licensed rapid CD103+ TRM cell differentiation in response to factors in the intestine. Licensing was regulated by retinoic acid signaling and primarily driven by factors other than CCR9 expression and CCR9-mediated gut homing. Thus, the MLN is specialized to promote intestinal CD103+ CD8 TRM cell development by licensing in situ differentiation.
Collapse
Affiliation(s)
- Zhijuan Qiu
- Department of Microbiology and Immunology, Center for Infectious Diseases, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Camille Khairallah
- Department of Microbiology and Immunology, Center for Infectious Diseases, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Timothy H. Chu
- Department of Microbiology and Immunology, Center for Infectious Diseases, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Jessica N. Imperato
- Department of Microbiology and Immunology, Center for Infectious Diseases, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Xinyuan Lei
- Department of Microbiology and Immunology, Center for Infectious Diseases, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Galina Romanov
- Department of Microbiology and Immunology, Center for Infectious Diseases, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Amha Atakilit
- Lung Biology Center, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Lynn Puddington
- Department of Immunology, University of Connecticut Health, Farmington, CT, USA
| | - Brian S. Sheridan
- Department of Microbiology and Immunology, Center for Infectious Diseases, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
| |
Collapse
|
11
|
Unique properties of tissue-resident memory T cells in the lungs: implications for COVID-19 and other respiratory diseases. Nat Rev Immunol 2022; 23:329-335. [PMID: 36494455 PMCID: PMC9735123 DOI: 10.1038/s41577-022-00815-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/22/2022] [Indexed: 12/13/2022]
Abstract
Tissue-resident memory T (TRM) cells were originally identified as a tissue-sequestered population of memory T cells that show lifelong persistence in non-lymphoid organs. That definition has slowly evolved with the documentation of TRM cells having variable terms of tissue residency combined with a capacity to return to the wider circulation. Nonetheless, reductionist experiments have identified an archetypical population of TRM cells showing intrinsic permanent residency in a wide range of non-lymphoid organs, with one notable exception: the lungs. Despite the fact that memory T cells generated during a respiratory infection are maintained in the circulation, local TRM cell numbers in the lung decline concomitantly with a decay in T cell-mediated protection. This Perspective describes the mechanisms that underpin long-term T cell lodgement in non-lymphoid tissues and explains why residency is transient for select TRM cell subsets. In doing so, it highlights the unusual nature of memory T cell egress from the lungs and speculates on the broader disease implications of this process, especially during infection with SARS-CoV-2.
Collapse
|
12
|
Künzli M, O’Flanagan SD, LaRue M, Talukder P, Dileepan T, Stolley JM, Soerens AG, Quarnstrom CF, Wijeyesinghe S, Ye Y, McPartlan JS, Mitchell JS, Mandl CW, Vile R, Jenkins MK, Ahmed R, Vezys V, Chahal JS, Masopust D. Route of self-amplifying mRNA vaccination modulates the establishment of pulmonary resident memory CD8 and CD4 T cells. Sci Immunol 2022; 7:eadd3075. [PMID: 36459542 PMCID: PMC9832918 DOI: 10.1126/sciimmunol.add3075] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Respiratory tract resident memory T cells (TRM), typically generated by local vaccination or infection, can accelerate control of pulmonary infections that evade neutralizing antibody. It is unknown whether mRNA vaccination establishes respiratory TRM. We generated a self-amplifying mRNA vaccine encoding the influenza A virus nucleoprotein that is encapsulated in modified dendron-based nanoparticles. Here, we report how routes of immunization in mice, including contralateral versus ipsilateral intramuscular boosts, or intravenous and intranasal routes, influenced influenza-specific cell-mediated and humoral immunity. Parabiotic surgeries revealed that intramuscular immunization was sufficient to establish CD8 TRM in the lung and draining lymph nodes. Contralateral, compared with ipsilateral, intramuscular boosting broadened the distribution of lymph node TRM and T follicular helper cells but slightly diminished resulting levels of serum antibody. Intranasal mRNA delivery established modest circulating CD8 and CD4 T cell memory but augmented distribution to the respiratory mucosa. Combining intramuscular immunizations with an intranasal mRNA boost achieved high levels of both circulating T cell memory and lung TRM. Thus, routes of mRNA vaccination influence humoral and cell-mediated immunity, and intramuscular prime-boosting establishes lung TRM that can be further expanded by an additional intranasal immunization.
Collapse
Affiliation(s)
- Marco Künzli
- Center for Immunology, Department of Microbiology and Immunology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Stephen D. O’Flanagan
- Center for Immunology, Department of Microbiology and Immunology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Madeleine LaRue
- Center for Immunology, Department of Microbiology and Immunology, University of Minnesota, Minneapolis, Minnesota, USA
| | | | - Thamotharampillai Dileepan
- Center for Immunology, Department of Microbiology and Immunology, University of Minnesota, Minneapolis, Minnesota, USA
| | - J. Michael Stolley
- Center for Immunology, Department of Microbiology and Immunology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Andrew G. Soerens
- Center for Immunology, Department of Microbiology and Immunology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Clare F. Quarnstrom
- Center for Immunology, Department of Microbiology and Immunology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Sathi Wijeyesinghe
- Center for Immunology, Department of Microbiology and Immunology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Yanqi Ye
- Emory Vaccine Center and Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia, USA
| | | | - Jason S. Mitchell
- Center for Immunology, Department of Microbiology and Immunology, University of Minnesota, Minneapolis, Minnesota, USA
| | | | - Richard Vile
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Marc K. Jenkins
- Center for Immunology, Department of Microbiology and Immunology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Rafi Ahmed
- Emory Vaccine Center and Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Vaiva Vezys
- Center for Immunology, Department of Microbiology and Immunology, University of Minnesota, Minneapolis, Minnesota, USA
| | | | - David Masopust
- Center for Immunology, Department of Microbiology and Immunology, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
13
|
Ma C, Zhang N. Lymphoid tissue residency: A key to understand Tcf-1 +PD-1 + T cells. Front Immunol 2022; 13:1074698. [PMID: 36569850 PMCID: PMC9767944 DOI: 10.3389/fimmu.2022.1074698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 11/21/2022] [Indexed: 12/12/2022] Open
Abstract
During chronic antigen exposure, a subset of exhausted CD8+ T cells differentiate into stem cell-like or progenitor-like T cells expressing both transcription factor Tcf-1 (T cell factor-1) and co-inhibitory receptor PD-1. These Tcf-1+ stem-like or progenitor exhausted T cells represent the key target for immunotherapies. Deeper understanding of the biology of Tcf-1+PD-1+ CD8+ T cells will lead to rational design of future immunotherapies. Here, we summarize recent findings about the migratory and resident behavior of Tcf-1+ T cells. Specifically, we will focus on TGF-β-dependent lymphoid tissue residency program of Tcf-1+ T cells, which may represent a key to understanding the differentiation and maintenance of Tcf-1+ stem-like CD8+ T cells during persistent antigen stimulation.
Collapse
Affiliation(s)
- Chaoyu Ma
- Department of Microbiology, Immunology and Molecular Genetics, Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Nu Zhang
- Department of Microbiology, Immunology and Molecular Genetics, Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| |
Collapse
|
14
|
Hassert M, Harty JT. Tissue resident memory T cells- A new benchmark for the induction of vaccine-induced mucosal immunity. Front Immunol 2022; 13:1039194. [PMID: 36275668 PMCID: PMC9581298 DOI: 10.3389/fimmu.2022.1039194] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 09/21/2022] [Indexed: 11/13/2022] Open
Abstract
Historically, the gold-standard benchmark for vaccine immunogenicity has been the induction of neutralizing antibodies detectable in the serum of peripheral blood. However, in recent years there has been a new appreciation for the mucosa as an important site for vaccine induced immunity. As a point of first contact, the mucosal tissue represents a major site of immune based detection and restriction of pathogen entry and dissemination. Tissue resident memory T cells (Trm) are one of the critical cell types involved in this early detection and restriction of mucosal pathogens. Following tissue-specific infection or vaccination, Trm lodge themselves within tissues and can perform rapid sensing and alarm functions to control local re-infections, in an effort that has been defined as important for restriction of a number of respiratory pathogens including influenza and respiratory syncytial virus. Despite this characterized importance, only minor attention has been paid to the importance of Trm as a benchmark for vaccine immunogenicity. The purpose of this review is to highlight the functions of Trm with particular emphasis on respiratory infections, and to suggest the inclusion of Trm elicitation as a benchmark for vaccine immunogenicity in animal models, and where possible, human samples.
Collapse
|
15
|
Suarez-Ramirez JE, Cauley LS, Chandiran K. CTLs Get SMAD When Pathogens Tell Them Where to Go. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 209:1025-1032. [PMID: 36130123 PMCID: PMC9512391 DOI: 10.4049/jimmunol.2200345] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 07/18/2022] [Indexed: 01/04/2023]
Abstract
Vaccines protect against infections by eliciting both Ab and T cell responses. Because the immunity wanes as protective epitopes get modified by accruing mutations, developing strategies for immunization against new variants is a major priority for vaccine development. CTLs eliminate cells that support viral replication and provide protection against new variants by targeting epitopes from internal viral proteins. This form of protection has received limited attention during vaccine development, partly because reliable methods for directing pathogen-specific memory CD8 T cells to vulnerable tissues are currently unavailable. In this review we examine how recent studies expand our knowledge of mechanisms that contribute to the functional diversity of CTLs as they respond to infection. We discuss the role of TGF-β and the SMAD signaling cascade during genetic programming of pathogen-specific CTLs and the pathways that promote formation of a newly identified subset of terminally differentiated memory CD8 T cells that localize in the vasculature.
Collapse
|
16
|
Zhang M, Li N, He Y, Shi T, Jie Z. Pulmonary resident memory T cells in respiratory virus infection and their inspiration on therapeutic strategies. Front Immunol 2022; 13:943331. [PMID: 36032142 PMCID: PMC9412965 DOI: 10.3389/fimmu.2022.943331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 07/26/2022] [Indexed: 11/25/2022] Open
Abstract
The immune system generates memory cells on infection with a virus for the first time. These memory cells play an essential role in protection against reinfection. Tissue-resident memory T (TRM) cells can be generated in situ once attacked by pathogens. TRM cells dominate the defense mechanism during early stages of reinfection and have gradually become one of the most popular focuses in recent years. Here, we mainly reviewed the development and regulation of various TRM cell signaling pathways in the respiratory tract. Moreover, we explored the protective roles of TRM cells in immune response against various respiratory viruses, such as Respiratory Syncytial Virus (RSV) and influenza. The complex roles of TRM cells against SARS-CoV-2 infection are also discussed. Current evidence supports the therapeutic strategies targeting TRM cells, providing more possibilities for treatment. Rational utilization of TRM cells for therapeutics is vital for defense against respiratory viruses.
Collapse
Affiliation(s)
- Meng Zhang
- Department of Pulmonary and Critical Care Medicine, Shanghai Fifth People’s Hospital, Fudan University, Shanghai, China
| | - Na Li
- Department of Pulmonary and Critical Care Medicine, Shanghai Fifth People’s Hospital, Fudan University, Shanghai, China
| | - Yanchao He
- Department of Pulmonary and Critical Care Medicine, Shanghai Fifth People’s Hospital, Fudan University, Shanghai, China
| | - Tianyun Shi
- Department of Pulmonary and Critical Care Medicine, Shanghai Fifth People’s Hospital, Fudan University, Shanghai, China
| | - Zhijun Jie
- Department of Pulmonary and Critical Care Medicine, Shanghai Fifth People’s Hospital, Fudan University, Shanghai, China
- Center of Community-Based Health Research, Fudan University, Shanghai, China
- *Correspondence: Zhijun Jie,
| |
Collapse
|
17
|
La Manna MP, Shekarkar Azgomi M, Tamburini B, Badami GD, Mohammadnezhad L, Dieli F, Caccamo N. Phenotypic and Immunometabolic Aspects on Stem Cell Memory and Resident Memory CD8+ T Cells. Front Immunol 2022; 13:884148. [PMID: 35784300 PMCID: PMC9247337 DOI: 10.3389/fimmu.2022.884148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 05/19/2022] [Indexed: 11/13/2022] Open
Abstract
The immune system, smartly and surprisingly, saves the exposure of a particular pathogen in its memory and reacts to the pathogen very rapidly, preventing serious diseases.Immunologists have long been fascinated by understanding the ability to recall and respond faster and more vigorously to a pathogen, known as “memory”.T-cell populations can be better described by using more sophisticated techniques to define phenotype, transcriptional and epigenetic signatures and metabolic pathways (single-cell resolution), which uncovered the heterogeneity of the memory T-compartment. Phenotype, effector functions, maintenance, and metabolic pathways help identify these different subsets. Here, we examine recent developments in the characterization of the heterogeneity of the memory T cell compartment. In particular, we focus on the emerging role of CD8+ TRM and TSCM cells, providing evidence on how their immunometabolism or modulation can play a vital role in their generation and maintenance in chronic conditions such as infections or autoimmune diseases.
Collapse
Affiliation(s)
- Marco Pio La Manna
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR) Azienda Ospedaliera Universitaria Policlinico (A.O.U.P.) Paolo Giaccone, University of Palermo, Palermo, Italy
- Department of Biomedicine, Neurosciences and Advanced Diagnostic (Bi.N.D.), University of Palermo, Palermo, Italy
| | - Mojtaba Shekarkar Azgomi
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR) Azienda Ospedaliera Universitaria Policlinico (A.O.U.P.) Paolo Giaccone, University of Palermo, Palermo, Italy
- Department of Biomedicine, Neurosciences and Advanced Diagnostic (Bi.N.D.), University of Palermo, Palermo, Italy
| | - Bartolo Tamburini
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR) Azienda Ospedaliera Universitaria Policlinico (A.O.U.P.) Paolo Giaccone, University of Palermo, Palermo, Italy
- Department of Biomedicine, Neurosciences and Advanced Diagnostic (Bi.N.D.), University of Palermo, Palermo, Italy
| | - Giusto Davide Badami
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR) Azienda Ospedaliera Universitaria Policlinico (A.O.U.P.) Paolo Giaccone, University of Palermo, Palermo, Italy
- Department of Biomedicine, Neurosciences and Advanced Diagnostic (Bi.N.D.), University of Palermo, Palermo, Italy
| | - Leila Mohammadnezhad
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR) Azienda Ospedaliera Universitaria Policlinico (A.O.U.P.) Paolo Giaccone, University of Palermo, Palermo, Italy
- Department of Biomedicine, Neurosciences and Advanced Diagnostic (Bi.N.D.), University of Palermo, Palermo, Italy
| | - Francesco Dieli
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR) Azienda Ospedaliera Universitaria Policlinico (A.O.U.P.) Paolo Giaccone, University of Palermo, Palermo, Italy
- Department of Biomedicine, Neurosciences and Advanced Diagnostic (Bi.N.D.), University of Palermo, Palermo, Italy
| | - Nadia Caccamo
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR) Azienda Ospedaliera Universitaria Policlinico (A.O.U.P.) Paolo Giaccone, University of Palermo, Palermo, Italy
- Department of Biomedicine, Neurosciences and Advanced Diagnostic (Bi.N.D.), University of Palermo, Palermo, Italy
- *Correspondence: Nadia Caccamo,
| |
Collapse
|
18
|
Hirai T, Yoshioka Y. Considerations of CD8+ T Cells for Optimized Vaccine Strategies Against Respiratory Viruses. Front Immunol 2022; 13:918611. [PMID: 35774782 PMCID: PMC9237416 DOI: 10.3389/fimmu.2022.918611] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 05/20/2022] [Indexed: 11/13/2022] Open
Abstract
The primary goal of vaccines that protect against respiratory viruses appears to be the induction of neutralizing antibodies for a long period. Although this goal need not be changed, recent severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants have drawn strong attention to another arm of acquired immunity, CD8+ T cells, which are also called killer T cells. Recent evidence accumulated during the coronavirus disease 2019 (COVID-19) pandemic has revealed that even variants of SARS-CoV-2 that escaped from neutralizing-antibodies that were induced by either infection or vaccination could not escape from CD8+ T cell-mediated immunity. In addition, although traditional vaccine platforms, such as inactivated virus and subunit vaccines, are less efficient in inducing CD8+ T cells, newly introduced platforms for SARS-CoV-2, namely, mRNA and adenoviral vector vaccines, can induce strong CD8+ T cell-mediated immunity in addition to inducing neutralizing antibodies. However, CD8+ T cells function locally and need to be at the site of infection to control it. To fully utilize the protective performance of CD8+ T cells, it would be insufficient to induce only memory cells circulating in blood, using injectable vaccines; mucosal immunization could be required to set up CD8+ T cells for the optimal protection. CD8+ T cells might also contribute to the pathology of the infection, change their function with age and respond differently to booster vaccines in comparison with antibodies. Herein, we overview cutting-edge ideas on CD8+ T cell-mediated immunity that can enable the rational design of vaccines for respiratory viruses.
Collapse
Affiliation(s)
- Toshiro Hirai
- Vaccine Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Japan
- Vaccine Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
- Laboratory of Nano-design for Innovative Drug Development, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Japan
- *Correspondence: Toshiro Hirai,
| | - Yasuo Yoshioka
- Vaccine Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Japan
- Vaccine Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
- Laboratory of Nano-design for Innovative Drug Development, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Japan
- The Research Foundation for Microbial Diseases of Osaka University, Suita, Japan
| |
Collapse
|
19
|
Abdeljaoued S, Arfa S, Kroemer M, Ben Khelil M, Vienot A, Heyd B, Loyon R, Doussot A, Borg C. Tissue-resident memory T cells in gastrointestinal cancer immunology and immunotherapy: ready for prime time? J Immunother Cancer 2022; 10:jitc-2021-003472. [PMID: 35470231 PMCID: PMC9039405 DOI: 10.1136/jitc-2021-003472] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/28/2022] [Indexed: 12/12/2022] Open
Abstract
Tissue-resident memory T (TRM) cells have emerged as immune sentinels that patrol the tissue microenvironment and orchestrate localized antitumor immunity in various solid cancers. Recent studies have revealed that TRM cells are key players in cancer immunosurveillance, and their involvement has been linked to favorable responses to immunotherapy as well as general better clinical outcome in cancer patients. In this review, we provide an overview of the major advances and recent findings regarding TRM cells phenotype, transcriptional and epigenetic regulation in cancer with a special focus on gastrointestinal tumors. Finally, we highlight the exciting clinical implication of TRM cells in these types of tumors.
Collapse
Affiliation(s)
- Syrine Abdeljaoued
- RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, University of Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Besançon, France .,Clinical Investigational Center, CIC-1431, Besançon, France
| | - Sara Arfa
- RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, University of Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Besançon, France.,Department of Digestive and Oncologic Surgery, Liver Transplantation Unit, University Hospital of Besançon, Besançon, France
| | - Marie Kroemer
- RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, University of Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Besançon, France.,Clinical Investigational Center, CIC-1431, Besançon, France.,Department of Pharmacy, University Hospital of Besançon, Besançon, France
| | - Myriam Ben Khelil
- RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, University of Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Besançon, France
| | - Angélique Vienot
- RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, University of Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Besançon, France.,Department of Medical Oncology, University Hospital of Besançon, Besançon, France
| | - Bruno Heyd
- Department of Digestive and Oncologic Surgery, Liver Transplantation Unit, University Hospital of Besançon, Besançon, France
| | - Romain Loyon
- RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, University of Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Besançon, France
| | - Alexandre Doussot
- Department of Digestive and Oncologic Surgery, Liver Transplantation Unit, University Hospital of Besançon, Besançon, France
| | - Christophe Borg
- RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, University of Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Besançon, France.,Clinical Investigational Center, CIC-1431, Besançon, France.,Department of Medical Oncology, University Hospital of Besançon, Besançon, France
| |
Collapse
|
20
|
Tan HX, Juno JA, Esterbauer R, Kelly HG, Wragg KM, Konstandopoulos P, Alcantara S, Alvarado C, Jones R, Starkey G, Wang BZ, Yoshino O, Tiang T, Grayson ML, Opdam H, D'Costa R, Vago A, Mackay LK, Gordon CL, Masopust D, Groom JR, Kent SJ, Wheatley AK. Lung-resident memory B cells established after pulmonary influenza infection display distinct transcriptional and phenotypic profiles. Sci Immunol 2022; 7:eabf5314. [PMID: 35089815 DOI: 10.1126/sciimmunol.abf5314] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Recent studies have established that memory B cells, largely thought to be circulatory in the blood, can take up long-term residency in inflamed tissues, analogous to widely described tissue-resident T cells. The dynamics of recruitment and retention of memory B cells to tissues and their immunological purpose remains unclear. Here, we characterized tissue-resident memory B cells (BRM) that are stably maintained in the lungs of mice after pulmonary influenza infection. Influenza-specific BRM were localized within inducible bronchus-associated lymphoid tissues (iBALTs) and displayed transcriptional signatures distinct from classical memory B cells in the blood or spleen while showing partial overlap with memory B cells in lung-draining lymph nodes. We identified lung-resident markers, including elevated expression of CXCR3, CCR6, and CD69, on hemagglutinin (HA)- and nucleoprotein (NP)-specific lung BRM. We found that CCR6 facilitates increased recruitment and/or retention of BRM in lungs and differentiation into antibody-secreting cells upon recall. Although expression of CXCR3 and CCR6 was comparable in total and influenza-specific memory B cells isolated across tissues of human donors, CD69 expression was higher in memory B cells from lung and draining lymph nodes of human organ donors relative to splenic and PBMC-derived populations, indicating that mechanisms underpinning BRM localization may be evolutionarily conserved. Last, we demonstrate that human memory B cells in lungs are transcriptionally distinct to populations in lung-draining lymph nodes or PBMCs. These data suggest that BRM may constitute a discrete component of B cell immunity, positioned at the lung mucosa for rapid humoral response against respiratory viral infections.
Collapse
Affiliation(s)
- Hyon-Xhi Tan
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| | - Jennifer A Juno
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| | - Robyn Esterbauer
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| | - Hannah G Kelly
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia.,ARC Centre for Excellence in Convergent Bio-Nano Science and Technology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Kathleen M Wragg
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| | - Penny Konstandopoulos
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| | - Sheilajen Alcantara
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia.,ARC Centre for Excellence in Convergent Bio-Nano Science and Technology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Carolina Alvarado
- Division of Immunology, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3050, Australia
| | - Robert Jones
- Department of Surgery, Austin Health, Heidelberg, Victoria 3084, Australia
| | - Graham Starkey
- Department of Surgery, Austin Health, Heidelberg, Victoria 3084, Australia
| | - Boa Zhong Wang
- Department of Surgery, Austin Health, Heidelberg, Victoria 3084, Australia
| | - Osamu Yoshino
- Department of Surgery, Austin Health, Heidelberg, Victoria 3084, Australia
| | - Thomas Tiang
- Department of Surgery, Austin Health, Heidelberg, Victoria 3084, Australia
| | - M Lindsay Grayson
- Department of Infectious Diseases, Austin Health, Heidelberg, Victoria 3084, Australia
| | - Helen Opdam
- DonateLife, The Australian Organ and Tissue Authority, Canberra, Australian Capital Territory 2601, Australia.,Department of Intensive Care, Austin Health, Heidelberg, Victoria 3084, Australia
| | - Rohit D'Costa
- DonateLife Victoria, Carlton, Victoria 3053, Australia.,Intensive Care Unit, The Royal Melbourne Hospital, Parkville, Victoria 3050, Australia
| | - Angela Vago
- Department of Surgery, Austin Health, Heidelberg, Victoria 3084, Australia
| | | | - Laura K Mackay
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| | - Claire L Gordon
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia.,Department of Infectious Diseases, Austin Health, Heidelberg, Victoria 3084, Australia
| | - David Masopust
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN, USA.,Center for Immunology, University of Minnesota, Minneapolis, MN, USA
| | - Joanna R Groom
- Division of Immunology, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3050, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Stephen J Kent
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia.,ARC Centre for Excellence in Convergent Bio-Nano Science and Technology, University of Melbourne, Parkville, Victoria 3010, Australia.,Melbourne Sexual Health Centre and Department of Infectious Diseases, Alfred Hospital and Central Clinical School, Monash University, Melbourne, Victoria 3004, Australia
| | - Adam K Wheatley
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia.,ARC Centre for Excellence in Convergent Bio-Nano Science and Technology, University of Melbourne, Parkville, Victoria 3010, Australia
| |
Collapse
|
21
|
Zheng MZM, Wakim LM. Tissue resident memory T cells in the respiratory tract. Mucosal Immunol 2022; 15:379-388. [PMID: 34671115 PMCID: PMC8526531 DOI: 10.1038/s41385-021-00461-z] [Citation(s) in RCA: 86] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 09/27/2021] [Accepted: 10/01/2021] [Indexed: 02/04/2023]
Abstract
Owing to their capacity to rapidly spread across the population, airborne pathogens represent a significant risk to global health. Indeed, several of the past major global pandemics have been instigated by respiratory pathogens. A greater understanding of the immune cells tasked with protecting the airways from infection will allow for the development of strategies that curb the spread and impact of these airborne diseases. A specific subset of memory T-cell resident in both the upper and lower respiratory tract, termed tissue-resident memory (Trm), have been shown to play an instrumental role in local immune responses against a wide breadth of both viral and bacterial infections. In this review, we discuss factors that influence respiratory tract Trm development, longevity, and immune surveillance and explore vaccination regimes that harness these cells, such approaches represent exciting new strategies that may be utilized to tackle the next global pandemic.
Collapse
Affiliation(s)
- Ming Z. M. Zheng
- grid.1008.90000 0001 2179 088XDepartment of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000 Australia
| | - Linda M. Wakim
- grid.1008.90000 0001 2179 088XDepartment of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000 Australia
| |
Collapse
|
22
|
Park SL, Mackay LK. Decoding Tissue-Residency: Programming and Potential of Frontline Memory T Cells. Cold Spring Harb Perspect Biol 2021; 13:a037960. [PMID: 33753406 PMCID: PMC8485744 DOI: 10.1101/cshperspect.a037960] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Memory T-cell responses are partitioned between the blood, secondary lymphoid organs, and nonlymphoid tissues. Tissue-resident memory T (Trm) cells are a population of immune cells that remain permanently in tissues without recirculating in blood. These nonrecirculating cells serve as a principal node in the anamnestic response to invading pathogens and developing malignancies. Here, we contemplate how T-cell tissue residency is defined and shapes protective immunity in the steady state and in the context of disease. We review the properties and heterogeneity of Trm cells, highlight the critical roles these cells play in maintaining tissue homeostasis and eliciting immune pathology, and explore how they might be exploited to treat disease.
Collapse
Affiliation(s)
- Simone L Park
- Department of Microbiology & Immunology at The Peter Doherty Institute for Infection & Immunity, The University of Melbourne, Melbourne, Victoria 3000, Australia
| | - Laura K Mackay
- Department of Microbiology & Immunology at The Peter Doherty Institute for Infection & Immunity, The University of Melbourne, Melbourne, Victoria 3000, Australia
| |
Collapse
|
23
|
Son YM, Sun J. Co-Ordination of Mucosal B Cell and CD8 T Cell Memory by Tissue-Resident CD4 Helper T Cells. Cells 2021; 10:cells10092355. [PMID: 34572004 PMCID: PMC8471972 DOI: 10.3390/cells10092355] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 09/02/2021] [Accepted: 09/03/2021] [Indexed: 12/25/2022] Open
Abstract
Adaptive cellular immunity plays a major role in clearing microbial invasion of mucosal tissues in mammals. Following the clearance of primary pathogens, memory lymphocytes are established both systemically and locally at pathogen entry sites. Recently, resident memory CD8 T and B cells (TRM and BRM respectively), which are parked mainly in non-lymphoid mucosal tissues, were characterized and demonstrated to be essential for protection against secondary microbial invasion. Here we reviewed the current understanding of the cellular and molecular cues regulating CD8 TRM and BRM development, maintenance and function. We focused particularly on elucidating the role of a novel tissue-resident helper T (TRH) cell population in assisting TRM and BRM responses in the respiratory mucosa following viral infection. Finally, we argue that the promotion of TRH responses by future mucosal vaccines would be key to the development of successful universal influenza or coronavirus vaccines, providing long-lasting immunity against a broad spectrum of viral strains.
Collapse
Affiliation(s)
- Young Min Son
- Division of Pulmonary and Critical Medicine, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA;
- Department of Immunology, Mayo Clinic, Rochester, MN 55905, USA
| | - Jie Sun
- Division of Pulmonary and Critical Medicine, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA;
- Department of Immunology, Mayo Clinic, Rochester, MN 55905, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
- Carter Immunology Center, University of Virginia, Charlottesville, VA 22908, USA
- Division of Infectious Disease and International Health, Department of Medicine, University of Virginia, Charlottesville, VA 22908, USA
- Correspondence: or
| |
Collapse
|
24
|
Farsakoglu Y, McDonald B, Kaech SM. Motility Matters: How CD8 + T-Cell Trafficking Influences Effector and Memory Cell Differentiation. Cold Spring Harb Perspect Biol 2021; 13:cshperspect.a038075. [PMID: 34001529 PMCID: PMC8327832 DOI: 10.1101/cshperspect.a038075] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Immunological memory is a hallmark of adaptive immunity that confers long-lasting protection from reinfections. Memory CD8+ T cells provide protection by actively scanning for their cognate antigen and migrating into inflamed tissues. Trafficking patterns of CD8+ T cells are also a major determinant of cell fate outcomes during differentiation into effector and memory cell states. CD8+ T-cell trafficking must therefore be dynamically and tightly regulated to ensure that CD8+ T cells arrive at the correct locations and differentiate to acquire appropriate effector functions. This review aims to discuss the importance of CD8+ T-cell trafficking patterns in regulating effector and memory differentiation, maintenance, and reactivation.
Collapse
Affiliation(s)
- Yagmur Farsakoglu
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, California 92037, USA
| | - Bryan McDonald
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, California 92037, USA.,Biomedical Sciences Graduate Program, University of California San Diego, La Jolla, California 92093, USA
| | - Susan M Kaech
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, California 92037, USA
| |
Collapse
|
25
|
Abstract
Elicitation of lung tissue-resident memory CD8 T cells (TRMs) is a goal of T cell-based vaccines against respiratory viral pathogens, such as influenza A virus (IAV). C-C chemokine receptor type 2 (CCR2)-dependent monocyte trafficking plays an essential role in the establishment of CD8 TRMs in lungs of IAV-infected mice. Here, we used a combination adjuvant-based subunit vaccine strategy that evokes multifaceted (TC1/TC17/TH1/TH17) IAV nucleoprotein-specific lung TRMs to determine whether CCR2 and monocyte infiltration are essential for vaccine-induced TRM development and protective immunity to IAV in lungs. Following intranasal vaccination, neutrophils, monocytes, conventional dendritic cells (DCs), and monocyte-derived dendritic cells internalized and processed vaccine antigen in lungs. We found that basic leucine zipper ATF-like transcription factor 3 (BATF3)-dependent DCs were essential for eliciting T cell responses, but CCR2 deficiency enhanced the differentiation of CD127hi, KLRG-1lo, OX40+ve CD62L+ve, and mucosally imprinted CD69+ve CD103+ve effector and memory CD8 T cells in lungs and airways of vaccinated mice. Mechanistically, increased development of lung TRMs induced by CCR2 deficiency was linked to dampened expression of T-bet but not altered TCF-1 levels or T cell receptor signaling in CD8 T cells. T1/T17 functional programming, parenchymal localization of CD8/CD4 effector and memory T cells, recall T cell responses, and protective immunity to a lethal IAV infection were unaffected in CCR2-deficient mice. Taken together, we identified a negative regulatory role for CCR2 and monocyte trafficking in mucosal imprinting and differentiation of vaccine-induced TRMs. Mechanistic insights from this study may aid the development of T-cell-based vaccines against respiratory viral pathogens, including IAV and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). IMPORTANCE While antibody-based immunity to influenza A virus (IAV) is type and subtype specific, lung- and airway-resident memory T cells that recognize conserved epitopes in the internal viral proteins are known to provide heterosubtypic immunity. Hence, broadly protective IAV vaccines need to elicit robust T cell memory in the respiratory tract. We have developed a combination adjuvant-based IAV nucleoprotein vaccine that elicits strong CD4 and CD8 T cell memory in lungs and protects against H1N1 and H5N1 strains of IAV. In this study, we examined the mechanisms that control vaccine-induced protective memory T cells in the respiratory tract. We found that trafficking of monocytes into lungs might limit the development of antiviral lung-resident memory T cells following intranasal vaccination. These findings suggest that strategies that limit monocyte infiltration can potentiate vaccine-induced frontline T-cell immunity to respiratory viruses, such as IAV and SARS-CoV-2.
Collapse
|
26
|
Pritzl CJ, Daniels MA, Teixeiro E. Interplay of Inflammatory, Antigen and Tissue-Derived Signals in the Development of Resident CD8 Memory T Cells. Front Immunol 2021; 12:636240. [PMID: 34234771 PMCID: PMC8255970 DOI: 10.3389/fimmu.2021.636240] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 04/29/2021] [Indexed: 12/21/2022] Open
Abstract
CD8 positive, tissue resident memory T cells (TRM) are a specialized subset of CD8 memory T cells that surveil tissues and provide critical first-line protection against tumors and pathogen re-infection. Recently, much effort has been dedicated to understanding the function, phenotype and development of TRM. A myriad of signals is involved in the development and maintenance of resident memory T cells in tissue. Much of the initial research focused on the roles tissue-derived signals play in the development of TRM, including TGFß and IL-33 which are critical for the upregulation of CD69 and CD103. However, more recent data suggest further roles for antigenic and pro-inflammatory cytokines. This review will focus on the interplay of pro-inflammatory, tissue and antigenic signals in the establishment of resident memory T cells.
Collapse
Affiliation(s)
| | | | - Emma Teixeiro
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO, United States
| |
Collapse
|
27
|
Marceaux C, Weeden CE, Gordon CL, Asselin-Labat ML. Holding our breath: the promise of tissue-resident memory T cells in lung cancer. Transl Lung Cancer Res 2021; 10:2819-2829. [PMID: 34295680 PMCID: PMC8264348 DOI: 10.21037/tlcr-20-819] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 08/10/2020] [Indexed: 12/12/2022]
Abstract
T cell memory is critical in controlling infection and plays an important role in anti-tumor responses in solid cancers. While effector memory and central memory T cells circulate and patrol non-lymphoid and lymphoid organs respectively, tissue resident memory T cells (TRM) permanently reside in tissues and provide local protective immune responses. In a number of solid tumors, tumor-specific T cell memory responses likely play an important role in keeping tumors in check, limiting cancer cell dissemination and reducing risk of relapse. In non-small cell lung cancer (NSCLC), a subset of tumor infiltrating lymphocytes (TILs) display phenotypic and functional characteristics associated with lung TRM (TRM-like TILs), including the expression of tissue-specific homing molecules and immune exhaustion markers. High infiltration of TRM-like TILs correlates with better survival outcomes for lung cancer patients, indicating that TRM-like TILs may contribute to anti-tumor responses. However, a number of TRM-like TILs do not display tumor specificity and the exact role of TRM-like TILs in mediating anti-tumor response in lung cancer is unclear. Here we review the characteristics of TRM-like TILs in lung cancer, the role these cells play in mediating anti-tumor immunity and the therapeutic implications of TRM-like TILs in the use and development of immunotherapy for lung cancer.
Collapse
Affiliation(s)
- Claire Marceaux
- Personalised Oncology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, Australia
| | - Clare E Weeden
- Personalised Oncology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, Australia
| | - Claire L Gordon
- Department of Microbiology and Immunology, The Peter Doherty Institute, The University of Melbourne, Parkville, Australia.,Department of Infectious Diseases, Austin Health, Heidelberg, Australia
| | - Marie-Liesse Asselin-Labat
- Personalised Oncology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, Australia.,Cancer Early Detection and Advanced Research Center, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| |
Collapse
|
28
|
Nelson MA, Ngamcherdtrakul W, Luoh SW, Yantasee W. Prognostic and therapeutic role of tumor-infiltrating lymphocyte subtypes in breast cancer. Cancer Metastasis Rev 2021; 40:519-536. [PMID: 33963482 PMCID: PMC8424653 DOI: 10.1007/s10555-021-09968-0] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 04/21/2021] [Indexed: 02/06/2023]
Abstract
Increased levels of total tumor-infiltrating lymphocytes (TILs) are generally associated with good prognosis in several breast cancer subtypes. Subtypes of TILs impact both tumor cells and immune cells in a variety of different ways, leading to either a pro-tumor or antitumor effect. Tumor-infiltrating CD8+ T cells and natural killer (NK) cells perform as effector cells against tumor cells and are associated with better clinical outcome. Immunotherapy approaches that improve the antitumor activity and proliferation of CD8+ T and NK cells include PD-1/PD-L1 blockade, CAR T cell therapy, or ex vivo-stimulated NK cells. A subset of CD8+ T cells, tissue-resident memory T cells, has also recently been associated with good prognosis in breast cancer patients, and has potential to serve as a predictive biomarker and therapeutic target. Tumor-infiltrating B cells also secrete apoptosis-inducing IgG antibodies and can act as antigen-presenting cells to prime CD4+ and CD8+ T cells. On the other hand, regulatory T and regulatory B cells modulate the immune response from CD8+ T cells and NK cells by secreting immunosuppressive cytokines and inhibiting maturation of antigen-presenting cells (APCs). These regulatory cells are typically associated with poor prognosis, therefore rendering suppression of their regulatory function a key immunotherapeutic strategy.
Collapse
Affiliation(s)
| | | | - Shiuh-Wen Luoh
- VA Portland Health Care System, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| | - Wassana Yantasee
- PDX Pharmaceuticals, Inc., Portland, OR, USA.
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR, USA.
| |
Collapse
|
29
|
Pauken KE, Godec J, Odorizzi PM, Brown KE, Yates KB, Ngiow SF, Burke KP, Maleri S, Grande SM, Francisco LM, Ali MA, Imam S, Freeman GJ, Haining WN, Wherry EJ, Sharpe AH. The PD-1 Pathway Regulates Development and Function of Memory CD8 + T Cells following Respiratory Viral Infection. Cell Rep 2021; 31:107827. [PMID: 32610128 DOI: 10.1016/j.celrep.2020.107827] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 07/05/2019] [Accepted: 06/05/2020] [Indexed: 12/11/2022] Open
Abstract
The PD-1 pathway regulates dysfunctional T cells in chronic infection and cancer, but the role of this pathway during acute infection remains less clear. Here, we demonstrate that PD-1 signals are needed for optimal memory. Mice deficient in the PD-1 pathway exhibit impaired CD8+ T cell memory following acute influenza infection, including reduced virus-specific CD8+ T cell numbers and compromised recall responses. PD-1 blockade during priming leads to similar differences early post-infection but without the defect in memory formation, suggesting that timing and/or duration of PD-1 blockade could be tailored to modulate host responses. Our studies reveal a role for PD-1 as an integrator of CD8+ T cell signals that promotes CD8+ T cell memory formation and suggest PD-1 continues to fine-tune CD8+ T cells after they migrate into non-lymphoid tissues. These findings have important implications for PD-1-based immunotherapy, in which PD-1 inhibition may influence memory responses in patients.
Collapse
Affiliation(s)
- Kristen E Pauken
- Department of Immunology, Blavatnik Institute, Harvard Medical School, and Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Jernej Godec
- Department of Immunology, Blavatnik Institute, Harvard Medical School, and Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA; Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Pamela M Odorizzi
- Institute for Immunology and Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Keturah E Brown
- Department of Immunology, Blavatnik Institute, Harvard Medical School, and Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Kathleen B Yates
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Shin Foong Ngiow
- Institute for Immunology and Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Kelly P Burke
- Department of Immunology, Blavatnik Institute, Harvard Medical School, and Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA; Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA 02115, USA
| | - Seth Maleri
- Department of Immunology, Blavatnik Institute, Harvard Medical School, and Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Shannon M Grande
- Department of Immunology, Blavatnik Institute, Harvard Medical School, and Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Loise M Francisco
- Department of Immunology, Blavatnik Institute, Harvard Medical School, and Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Mohammed-Alkhatim Ali
- Institute for Immunology and Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Sabrina Imam
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Gordon J Freeman
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA 02115, USA
| | - W Nicholas Haining
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Division of Hematology/Oncology, Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - E John Wherry
- Institute for Immunology and Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA.
| | - Arlene H Sharpe
- Department of Immunology, Blavatnik Institute, Harvard Medical School, and Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115, USA.
| |
Collapse
|
30
|
Qiu Z, Chu TH, Sheridan BS. TGF-β: Many Paths to CD103 + CD8 T Cell Residency. Cells 2021; 10:cells10050989. [PMID: 33922441 PMCID: PMC8145941 DOI: 10.3390/cells10050989] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 04/19/2021] [Accepted: 04/21/2021] [Indexed: 12/24/2022] Open
Abstract
CD8 tissue-resident memory T (TRM) cells primarily reside in nonlymphoid tissues without recirculating and provide front-line protective immunity against infections and cancers. CD8 TRM cells can be generally divided into CD69+ CD103− TRM cells (referred to as CD103− TRM cells) and CD69+ CD103+ TRM cells (referred to as CD103+ TRM cells). TGF-β plays a critical role in the development and maintenance of CD103+ CD8 TRM cells. In this review, we summarize the current understanding of tissue-specific activation of TGF-β mediated by integrins and how it contributes to CD103+ CD8 TRM cell development and maintenance. Furthermore, we discuss the underlying mechanisms utilized by TGF-β to regulate the development and maintenance of CD103+ CD8 TRM cells. Overall, this review highlights the importance of TGF-β in regulating this unique subset of memory CD8 T cells that may shed light on improving vaccine design to target this population.
Collapse
|
31
|
Lee W, Kingstad-Bakke B, Kedl RM, Kawaoka Y, Suresh M. CCR2 Regulates Vaccine-Induced Mucosal T-Cell Memory to Influenza A Virus. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021. [PMID: 33791695 DOI: 10.1101/2021.03.24.436901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Elicitation of lung tissue-resident memory CD8 T cells (T RM s) is a goal of T-cell based vaccines against respiratory viral pathogens such as influenza A virus (IAV). Chemokine receptor 2 (CCR2)-dependent monocyte trafficking plays an essential role in the establishment of CD8 T RM s in lungs of IAV-infected mice. Here, we used a combination adjuvant-based subunit vaccine strategy that evokes multifaceted (T C 1/T C 17/T H 1/T H 17) IAV nucleoprotein-specific lung T RM s, to determine whether CCR2 and monocyte infiltration are essential for vaccine-induced T RM development and protective immunity to IAV in lungs. Following intranasal vaccination, neutrophils, monocytes, conventional dendrtitic cells (DCs) and monocyte-derived DCs internalized and processed vaccine antigen in lungs. We also found that Basic Leucine Zipper ATF-Like Transcription Factor 3 (BATF-3)-dependent DCs were essential for eliciting T cell responses, but CCR2 deficiency enhanced the differentiation of CD127 HI /KLRG-1 LO , OX40 +ve CD62L +ve and mucosally imprinted CD69 +ve CD103 +ve effector and memory CD8 T cells in lungs and airways of vaccinated mice. Mechanistically, increased development of lung T RM s, induced by CCR2 deficiency was linked to dampened expression of T-bet, but not altered TCF-1 levels or T cell receptor signaling in CD8 T cells. T1/T17 functional programming, parenchymal localization of CD8/CD4 effector and memory T cells, recall T cell responses and protective immunity to a lethal IAV infection were unaffected in CCR2-deficient mice. Taken together, we identified a negative regulatory role for CCR2 and monocyte trafficking in mucosal imprinting and differentiation of vaccine-induced T RM s. Mechanistic insights from this study may aid the development of T-cell-based vaccines against respiratory viral pathogens including IAV and SARS-CoV-2. Importance While antibody-based immunity to influenza A virus (IAV) is type and sub-type specific, lung and airway-resident memory T cells that recognize conserved epitopes in the internal viral proteins are known to provide heterosubtypic immunity. Hence, broadly protective IAV vaccines need to elicit robust T-cell memory in the respiratory tract. We have developed a combination adjuvant-based IAV nucleoprotein vaccine that elicits strong CD4 and CD8 T cell memory in lungs and protects against H1N1 and H5N1 strains of IAV. In this study, we examined the mechanisms that control vaccine-induced protective memory T cells in the respiratory tract. We found that trafficking of monocytes into lungs might limit the development of anti-viral lung-resident memory T cells, following intranasal vaccination. These findings suggested that strategies that limit monocyte infiltration can potentiate vaccine-induced frontline T-cell immunity to respiratory viruses such as IAV and SARS-CoV-2.
Collapse
|
32
|
Mora-Buch R, Bromley SK. Discipline in Stages: Regulating CD8 + Resident Memory T Cells. Front Immunol 2021; 11:624199. [PMID: 33815352 PMCID: PMC8017121 DOI: 10.3389/fimmu.2020.624199] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 12/31/2020] [Indexed: 12/13/2022] Open
Abstract
Resident memory CD8+ T (TRM) cells are a lymphocyte lineage distinct from circulating memory CD8+ T cells. TRM lodge within peripheral tissues and secondary lymphoid organs where they provide rapid, local protection from pathogens and control tumor growth. However, dysregulation of CD8+ TRM formation and/or activation may contribute to the pathogenesis of autoimmune diseases. Intrinsic mechanisms, including transcriptional networks and inhibitory checkpoint receptors control TRM differentiation and response. Additionally, extrinsic stimuli such as cytokines, cognate antigen, fatty acids, and damage signals regulate TRM formation, maintenance, and expansion. In this review, we will summarize knowledge of CD8+ TRM generation and highlight mechanisms that regulate the persistence and responses of heterogeneous TRM populations in different tissues and distinct microenvironments.
Collapse
Affiliation(s)
- Rut Mora-Buch
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Shannon K Bromley
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
33
|
Stolley JM, Johnston TS, Soerens AG, Beura LK, Rosato PC, Joag V, Wijeyesinghe SP, Langlois RA, Osum KC, Mitchell JS, Masopust D. Retrograde migration supplies resident memory T cells to lung-draining LN after influenza infection. J Exp Med 2021; 217:151876. [PMID: 32568362 PMCID: PMC7398169 DOI: 10.1084/jem.20192197] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 03/20/2020] [Accepted: 05/12/2020] [Indexed: 01/06/2023] Open
Abstract
Numerous observations indicate that resident memory T cells (TRM) undergo unusually rapid attrition within the lung. Here we demonstrate that contraction of lung CD8+ T cell responses after influenza infection is contemporized with egress of CD69+/CD103+ CD8+ T cells to the draining mediastinal LN via the lymphatic vessels, which we term retrograde migration. Cells within the draining LN retained canonical markers of lung TRM, including CD103 and CD69, lacked Ly6C expression (also a feature of lung TRM), maintained granzyme B expression, and did not equilibrate among immunized parabiotic mice. Investigations of bystander infection or removal of the TCR from established memory cells revealed that the induction of the TRM phenotype was dependent on antigen recognition; however, maintenance was independent. Thus, local lung infection induces CD8+ T cells with a TRM phenotype that nevertheless undergo retrograde migration, yet remain durably committed to the residency program within the draining LN, where they provide longer-lived regional memory while chronicling previous upstream antigen experiences.
Collapse
Affiliation(s)
- J Michael Stolley
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN.,Center for Immunology, University of Minnesota, Minneapolis, MN
| | - Timothy S Johnston
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN.,Center for Immunology, University of Minnesota, Minneapolis, MN
| | - Andrew G Soerens
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN.,Center for Immunology, University of Minnesota, Minneapolis, MN
| | - Lalit K Beura
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN.,Center for Immunology, University of Minnesota, Minneapolis, MN.,Department of Microbiology and Immunology, Brown University, Providence, RI
| | - Pamela C Rosato
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN.,Center for Immunology, University of Minnesota, Minneapolis, MN
| | - Vineet Joag
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN.,Center for Immunology, University of Minnesota, Minneapolis, MN
| | - Sathi P Wijeyesinghe
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN.,Center for Immunology, University of Minnesota, Minneapolis, MN
| | - Ryan A Langlois
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN.,Center for Immunology, University of Minnesota, Minneapolis, MN
| | - Kevin C Osum
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN.,Center for Immunology, University of Minnesota, Minneapolis, MN
| | - Jason S Mitchell
- Center for Immunology, University of Minnesota, Minneapolis, MN.,Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN
| | - David Masopust
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN.,Center for Immunology, University of Minnesota, Minneapolis, MN
| |
Collapse
|
34
|
Fernandez-Ruiz D, de Menezes MN, Holz LE, Ghilas S, Heath WR, Beattie L. Harnessing liver-resident memory T cells for protection against malaria. Expert Rev Vaccines 2021; 20:127-141. [PMID: 33501877 DOI: 10.1080/14760584.2021.1881485] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Tissue-resident memory T cells (TRM cells) are powerful mediators of protracted adaptive immunity to infection in peripheral organs. Harnessing TRM cells through vaccination hence promises unprecedented potential for protection against infection. A paramount example of this is malaria, a major infectious disease for which immunity through traditional vaccination strategies remains challenging. Liver TRM cells appear to be highly protective against malaria, and recent developments in our knowledge of the biology of these cells have defined promising, novel strategies for their induction. AREAS COVERED Here, we describe the path that led to the discovery of TRM cells and discuss the importance of liver TRM cells in immunity against Plasmodium spp. infection; we summarize current knowledge on TRM cell biology and discuss the current state and potential of TRM-based vaccination against malaria. EXPERT OPINION TRM based vaccination has emerged as a promising means to achieve efficient protection against malaria. Recent advances provide a solid basis for continuing the development of this area of research. Deeper understanding of the mechanisms that mediate TRM formation and maintenance and identification of immunogenic and protective target epitopes suitable for human vaccination remain the main challenges for translation of these discoveries.
Collapse
Affiliation(s)
- Daniel Fernandez-Ruiz
- Dept. Of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, the University of Melbourne, Melbourne, Vic, Australia.,Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Melbourne,Vic, Australia
| | - Maria N de Menezes
- Dept. Of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, the University of Melbourne, Melbourne, Vic, Australia
| | - Lauren E Holz
- Dept. Of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, the University of Melbourne, Melbourne, Vic, Australia.,Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Melbourne,Vic, Australia
| | - Sonia Ghilas
- Dept. Of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, the University of Melbourne, Melbourne, Vic, Australia.,Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Melbourne,Vic, Australia
| | - William R Heath
- Dept. Of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, the University of Melbourne, Melbourne, Vic, Australia.,Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Melbourne,Vic, Australia
| | - Lynette Beattie
- Dept. Of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, the University of Melbourne, Melbourne, Vic, Australia.,Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Melbourne,Vic, Australia
| |
Collapse
|
35
|
Pauken KE, Torchia JA, Chaudhri A, Sharpe AH, Freeman GJ. Emerging concepts in PD-1 checkpoint biology. Semin Immunol 2021; 52:101480. [PMID: 34006473 PMCID: PMC8545711 DOI: 10.1016/j.smim.2021.101480] [Citation(s) in RCA: 102] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 05/03/2021] [Indexed: 12/11/2022]
Abstract
The PD-1 pathway is a cornerstone in immune regulation. While the PD-1 pathway has received considerable attention for its role in contributing to the maintenance of T cell exhaustion in chronic infection and cancer, the PD-1 pathway plays diverse roles in regulating host immunity beyond T cell exhaustion. Here, we discuss emerging concepts in the PD-1 pathway, including (1) the impact of PD-1 inhibitors on diverse T cell differentiation states including effector and memory T cell development during acute infection, as well as T cell exhaustion during chronic infection and cancer, (2) the role of PD-1 in regulating Treg cells, NK cells, and ILCs, and (3) the functions of PD-L1/B7-1 and PD-L2/RGMb/neogenin interactions. We then discuss the emerging use of neoadjuvant PD-1 blockade in the treatment of early-stage cancers and how the timing of PD-1 blockade may improve clinical outcomes. The diverse binding partners of PD-1 and its associated ligands, broad expression patterns of the receptors and ligands, differential impact of PD-1 modulation on cells depending on location and state of differentiation, and timing of PD-1 blockade add additional layers of complexity to the PD-1 pathway, and are important considerations for improving the efficacy and safety of PD-1 pathway therapeutics.
Collapse
Affiliation(s)
- 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
| | - James A Torchia
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, 02215, USA
| | - Apoorvi Chaudhri
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, 02215, USA; Department of Melanoma Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA; The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, 77030, 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; Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA; Department of Pathology, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Gordon J Freeman
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, 02215, USA.
| |
Collapse
|
36
|
van de Wall S, Badovinac VP, Harty JT. Influenza-Specific Lung-Resident Memory CD8 + T Cells. Cold Spring Harb Perspect Biol 2021; 13:cshperspect.a037978. [PMID: 33288540 PMCID: PMC7849341 DOI: 10.1101/cshperspect.a037978] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Despite the availability of seasonal vaccines, influenza A (IAV) prevails as a leading cause of respiratory infection worldwide. Current vaccination efforts aim at increasing protection against heterologous and potentially pandemic IAV strains. Lung-resident CD8+ T cells (Trm) generated upon IAV infection are vital for heterosubtypic immunity to IAV reexposure and provide quick and robust responses upon reactivation. Yet, protection wanes with time as lung Trm cell numbers decline, a contrasting feature with Trm cells at other mucosal sites such as the skin. In this review, we discuss current data on lung Trm compared to Trm cells in other tissues. Furthermore, major knowledge gaps in the generation and maintenance of IAV-specific lung Trm are addressed and mechanisms that may contribute to their decline are discussed. Further understanding in the mechanisms that govern effector function versus immunopathology is paramount for future IAV vaccine design in enhancing durability of lung Trm cells.
Collapse
Affiliation(s)
- Stephanie van de Wall
- Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa 52242, USA
| | - Vladimer P. Badovinac
- Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa 52242, USA;,Department of Pathology,, University of Iowa, Iowa City, Iowa 52242, USA;,Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, Iowa 52242, USA
| | - John T. Harty
- Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa 52242, USA;,Department of Pathology,, University of Iowa, Iowa City, Iowa 52242, USA;,Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, Iowa 52242, USA
| |
Collapse
|
37
|
Hirai T, Yang Y, Zenke Y, Li H, Chaudhri VK, De La Cruz Diaz JS, Zhou PY, Nguyen BAT, Bartholin L, Workman CJ, Griggs DW, Vignali DAA, Singh H, Masopust D, Kaplan DH. Competition for Active TGFβ Cytokine Allows for Selective Retention of Antigen-Specific Tissue- Resident Memory T Cells in the Epidermal Niche. Immunity 2021; 54:84-98.e5. [PMID: 33212014 PMCID: PMC7856016 DOI: 10.1016/j.immuni.2020.10.022] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 09/01/2020] [Accepted: 10/28/2020] [Indexed: 01/13/2023]
Abstract
Following antigen-driven expansion in lymph node, transforming growth factor-β (TGFβ) is required for differentiation of skin-recruited CD8+ T cell effectors into epidermal resident memory T (Trm) cells and their epidermal persistence. We found that the source of TGFβ -supporting Trm cells was autocrine. In addition, antigen-specific Trm cells that encountered cognate antigen in the skin, and bystander Trm cells that did not, both displayed long-term persistence in the epidermis under steady-state conditions. However, when the active-TGFβ was limited or when new T cell clones were recruited into the epidermis, antigen-specific Trm cells were more efficiently retained than bystander Trm cells. Genetically enforced TGFβR signaling allowed bystander Trm cells to persist in the epidermis as efficiently as antigen-specific Trm cells in both contexts. Thus, competition between T cells for active TGFβ represents an unappreciated selective pressure that promotes the accumulation and persistence of antigen-specific Trm cells in the epidermal niche.
Collapse
Affiliation(s)
- Toshiro Hirai
- Department of Dermatology, University of Pittsburgh, Pittsburgh, PA 15261, USA; Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15261, USA.
| | - Yi Yang
- Department of Dermatology, University of Pittsburgh, Pittsburgh, PA 15261, USA; Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15261, USA; Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; The Third Xiangya Hospital, Central South University, Changsha, China
| | - Yukari Zenke
- Department of Dermatology, University of Pittsburgh, Pittsburgh, PA 15261, USA; Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15261, USA; Department of Dermatology, St. Luke's International Hospital, Tokyo, Japan
| | - Haiyue Li
- Department of Dermatology, University of Pittsburgh, Pittsburgh, PA 15261, USA; Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15261, USA; School of Medicine, Tsinghua University, No. 1 Tsinghua Yuan, Haidian District, Beijing 100084, China
| | - Virendra K Chaudhri
- Center for Systems Immunology and the Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jacinto S De La Cruz Diaz
- Department of Dermatology, University of Pittsburgh, Pittsburgh, PA 15261, USA; Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Paul Yifan Zhou
- Department of Dermatology, University of Pittsburgh, Pittsburgh, PA 15261, USA; Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Breanna Anh-Thu Nguyen
- Department of Dermatology, University of Pittsburgh, Pittsburgh, PA 15261, USA; Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Laurent Bartholin
- TGF-b & Pancreatic Cancer Lab, Centre de Recherche en Cancérologie de Lyon (CRCL), Centre Léon Bérard, INSERM 1052, CNRS 5286, Université de Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | - Creg J Workman
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15261, USA; Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA
| | - David W Griggs
- Department of Molecular Microbiology and Immunology, Saint Louis University, MO 63104, USA
| | - Dario A A Vignali
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15261, 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
| | - Harinder Singh
- Center for Systems Immunology and the Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| | - David Masopust
- Department of Microbiology and Immunology, Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Daniel H Kaplan
- Department of Dermatology, University of Pittsburgh, Pittsburgh, PA 15261, USA; Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15261, USA.
| |
Collapse
|
38
|
Takamura S. Divergence of Tissue-Memory T Cells: Distribution and Function-Based Classification. Cold Spring Harb Perspect Biol 2020; 12:cshperspect.a037762. [PMID: 32816841 DOI: 10.1101/cshperspect.a037762] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Tissue-resident memory T cells (Trm) comprise the majority of memory cells in nonlymphoid tissues and play a predominant role in immunity at barrier surfaces. A better understanding of Trm cell maintenance and function is essential for the development of vaccines that confer frontline protection. However, it is currently challenging to precisely distinguish Trm cells from other T cells, and this has led to confusion in the literature. Here we highlight gaps in our understanding of tissue memory and discuss recent advances in the classification of Trm cell subsets based on their distribution and functional characteristics.
Collapse
Affiliation(s)
- Shiki Takamura
- Department of Immunology, Kindai University Faculty of Medicine, Osaka-Sayama, Osaka 589-8511, Japan
| |
Collapse
|
39
|
Chu KL, Batista NV, Girard M, Watts TH. Monocyte-Derived Cells in Tissue-Resident Memory T Cell Formation. THE JOURNAL OF IMMUNOLOGY 2020; 204:477-485. [PMID: 31964721 DOI: 10.4049/jimmunol.1901046] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 10/02/2019] [Indexed: 12/15/2022]
Abstract
There is currently much interest in how different dendritic cell and macrophage populations contribute to T cell-mediated immunity. Although conventional dendritic cell subsets have received much attention for their role in T cell priming, there is emerging evidence for a role for monocyte-derived APC (MoAPC) in tissue-resident memory T cell (Trm) formation. Cells of the monocyte/macrophage lineage play a key role in providing chemokines and cytokines for the localization, differentiation, and survival of Trm and Trm precursors. In addition, inflammatory MoAPC are the key providers of TNF superfamily costimulatory signals, a signal we refer to as signal 4 for T cell activation. Recent evidence suggests that signal 4 from MoAPC occurs postpriming and substantially increases Trm formation. Key questions remain, such as the Ag dependence of signal 4 and the specific mechanisms by which MoAPC-Trm interactions affect the long-term maintenance of Trm.
Collapse
Affiliation(s)
- Kuan-Lun Chu
- Department of Immunology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Nathália V Batista
- Department of Immunology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Mélanie Girard
- Department of Immunology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Tania H Watts
- Department of Immunology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| |
Collapse
|
40
|
Garrido-Martin EM, Mellows TWP, Clarke J, Ganesan AP, Wood O, Cazaly A, Seumois G, Chee SJ, Alzetani A, King EV, Hedrick CC, Thomas G, Friedmann PS, Ottensmeier CH, Vijayanand P, Sanchez-Elsner T. M1 hot tumor-associated macrophages boost tissue-resident memory T cells infiltration and survival in human lung cancer. J Immunother Cancer 2020; 8:e000778. [PMID: 32699181 PMCID: PMC7375465 DOI: 10.1136/jitc-2020-000778] [Citation(s) in RCA: 102] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/26/2020] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND The role of tumor-associated macrophages (TAMs) in determining the outcome between the antitumor effects of the adaptive immune system and the tumor's anti-immunity stratagems, is controversial. Macrophages modulate their activities and phenotypes by integration of signals in the tumor microenvironment. Depending on how macrophages are activated, they may adopt so-called M1-like, antitumor or M2-like, protumor profiles. In many solid tumors, a dominance of M2-like macrophages is associated with poor outcomes but in some tumor types, strong M1-like profiles are linked to better outcomes. We aimed to investigate the interrelationship of these TAM populations to establish how they modulate the efficacy of the adaptive immune system in early lung cancer. METHODS Macrophages from matched lung (non-tumor-associated macrophages (NTAMs)) and tumor samples (TAMs) from resected lung cancers were assessed by bulk and single-cell transcriptomic analysis. Protein expression of genes characteristic of M1-like (chemokine (C-X-C motif) ligand 9) or M2-like (matrix metallopeptidase 12) functions was confirmed by confocal microscopy. Immunohistochemistry related the distribution of TAM transcriptomic signatures to density of CD8+ tissue-resident memory T cells (TRM) in tumors and survival data from an independent cohort of 393 patients with lung cancer. RESULTS TAMs have significantly different transcriptomic profiles from NTAMs with >1000 differentially expressed genes. TAMs displayed a strong M2-like signature with no significant variation between patients. However, single-cell RNA-sequencing supported by immuno-stained cells revealed that additionally, in 25% of patients the M2-like TAMs also co-expressed a strong/hot M1-like signature (M1hot). Importantly, there was a strong association between the density of M1hot TAMs and TRM cells in tumors that was in turn linked to better survival. Our data suggest a mechanism by which M1hot TAMs may recruit TRM cells via CXCL9 expression and sustain them by making available more of the essential fatty acids on which TRM depend. CONCLUSIONS We showed that in early lung cancer, expression of M1-like and M2-like gene signatures are not mutually exclusive since the same TAMs can simultaneously display both gene-expression profiles. The presence of M1hot TAMs was associated with a strong TRM tumor-infiltrate and better outcomes. Thus, therapeutic approaches to re-program TAMs to an M1hot phenotype are likely to augment the adaptive antitumor responses.
Collapse
Affiliation(s)
- Eva M Garrido-Martin
- Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Southampton, UK
| | - Toby W P Mellows
- Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Southampton, UK
| | - James Clarke
- La Jolla Institute for Immunology, La Jolla, California, USA
| | | | - Oliver Wood
- Cancer Sciences, University of Southampton Faculty of Medicine, Southampton, UK
| | - Angelica Cazaly
- Cancer Sciences, University of Southampton Faculty of Medicine, Southampton, UK
| | - Gregory Seumois
- La Jolla Institute for Immunology, La Jolla, California, USA
| | - Serena J Chee
- Cancer Sciences, University of Southampton Faculty of Medicine, Southampton, UK
| | - Aiman Alzetani
- Southampton University Hospitals NHS Trust, Southampton, UK
| | - Emma V King
- Cancer Sciences, University of Southampton Faculty of Medicine, Southampton, UK
| | - Catherine C Hedrick
- Inflammatory Biology, La Jolla Institute for Immunology, La Jolla, California, USA
| | - Gareth Thomas
- Cancer Sciences, University of Southampton Faculty of Medicine, Southampton, UK
| | - Peter S Friedmann
- Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Southampton, UK
| | | | - Pandurangan Vijayanand
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, California, USA
| | - Tilman Sanchez-Elsner
- Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Southampton, UK
| |
Collapse
|
41
|
T RM integrins CD103 and CD49a differentially support adherence and motility after resolution of influenza virus infection. Proc Natl Acad Sci U S A 2020; 117:12306-12314. [PMID: 32439709 PMCID: PMC7275699 DOI: 10.1073/pnas.1915681117] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Current influenza vaccination strategies require annual immunizations, with fairly low efficacy rates. One technique to improve protection against a greater breadth of influenza viruses is to elicit broadly cross-reactive cell-mediated immunity and generate a local population of cytotoxic T cells to respond to conserved regions of circulating viruses. However, this approach requires improved understanding of how these cells migrate within and attach to the tissue in order to persist and offer long-term immunity. This study investigates how receptors on the T cell surface impact the cell’s ability to interact with the tissue and provide evidence of which of these receptors are essential for protection. Furthermore, these studies reveal functional in vivo mechanisms of cellular markers used to characterize TRM. Tissue-resident memory CD8 T (TRM) cells are a unique immune memory subset that develops and remains in peripheral tissues at the site of infection, providing future host resistance upon reexposure to that pathogen. In the pulmonary system, TRM are identified through S1P antagonist CD69 and expression of integrins CD103/β7 and CD49a/CD29(β1). Contrary to the established role of CD69 on CD8 T cells, the functions of CD103 and CD49a on this population are not well defined. This study examines the expression patterns and functions of CD103 and CD49a with a specific focus on their impact on T cell motility during influenza virus infection. We show that the TRM cell surface phenotype develops by 2 wk postinfection, with the majority of the population expressing CD49a and a subset that is also positive for CD103. Despite a previously established role in retaining TRM in peripheral tissues, CD49a facilitates locomotion of virus-specific CD8 T cells, both in vitro and in vivo. These results demonstrate that CD49a may contribute to local surveillance mechanisms of the TRM population.
Collapse
|
42
|
Abstract
In mammals, adaptive immunity is mediated by a broadly diverse repertoire of naive B and T lymphocytes that recirculate between secondary lymphoid organs. Initial antigen exposure promotes lymphocyte clonal expansion and differentiation, including the formation of memory cells. Antigen-specific memory cells are maintained at higher frequencies than their naive counterparts and have different functional and homing abilities. Importantly, a subset of memory cells, known as tissue-resident memory cells, is maintained without recirculating in nonlymphoid tissues, often at barrier surfaces, where they can be reactivated by antigen and rapidly perform effector functions that help protect the tissue in which they reside. Although antigen-experienced B cells are abundant at many barrier surfaces, their characterization as tissue-resident memory B (BRM) cells is not well developed. In this study, we describe the characteristics of memory B cells in various locations and discuss their possible contributions to immunity and homeostasis as bona fide BRM cells.
Collapse
Affiliation(s)
- S. Rameeza Allie
- Division of Clinical Immunology and Rheumatology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Troy D. Randall
- Division of Clinical Immunology and Rheumatology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
43
|
Abstract
The signaling lipid sphingosine 1-phosphate (S1P) plays critical roles in an immune response. Drugs targeting S1P signaling have been remarkably successful in treatment of multiple sclerosis, and they have shown promise in clinical trials for colitis and psoriasis. One mechanism of these drugs is to block lymphocyte exit from lymph nodes, where lymphocytes are initially activated, into circulation, from which lymphocytes can reach sites of inflammation. Indeed, S1P can be considered a circulation marker, signaling to immune cells to help them find blood and lymphatic vessels, and to endothelial cells to stabilize the vasculature. That said, S1P plays pleiotropic roles in the immune response, and it will be important to build an integrated view of how S1P shapes inflammation. S1P can function so effectively because its distribution is exquisitely tightly controlled. Here we review how S1P gradients regulate immune cell exit from tissues, with particular attention to key outstanding questions in the field.
Collapse
Affiliation(s)
- Audrey A.L. Baeyens
- Skirball Institute of Biomolecular Medicine, New York University School of Medicine, New York, NY 10016, USA;,
| | - Susan R. Schwab
- Skirball Institute of Biomolecular Medicine, New York University School of Medicine, New York, NY 10016, USA;,
| |
Collapse
|
44
|
T Cell Factor 1 Suppresses CD103+ Lung Tissue-Resident Memory T Cell Development. Cell Rep 2020; 31:107484. [DOI: 10.1016/j.celrep.2020.03.048] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 01/07/2020] [Accepted: 03/13/2020] [Indexed: 01/31/2023] Open
|
45
|
Crepeau RL, Ford ML. Programmed T cell differentiation: Implications for transplantation. Cell Immunol 2020; 351:104099. [PMID: 32247511 DOI: 10.1016/j.cellimm.2020.104099] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 03/27/2020] [Accepted: 03/27/2020] [Indexed: 12/27/2022]
Abstract
While T cells play a critical role in protective immunity against infection, they are also responsible for graft rejection in the setting of transplantation. T cell differentiation is regulated by both intrinsic transcriptional pathways as well as extrinsic factors such as antigen encounter and the cytokine milieu. Herein, we review recent discoveries in the transcriptional regulation of T cell differentiation and their impact on the field of transplantation. Recent studies uncovering context-dependent differentiation programs that differ in the setting of infection or transplantation will also be discussed. Understanding the key transcriptional pathways that underlie T cell responses in transplantation has important clinical implications, including development of novel therapeutic agents to mitigate graft rejection.
Collapse
Affiliation(s)
- Rebecca L Crepeau
- Emory Transplant Center, Department of Surgery, Emory University, 101 Woodruff Circle, Suite 5208, Atlanta, GA 30322, United States
| | - Mandy L Ford
- Emory Transplant Center, Department of Surgery, Emory University, 101 Woodruff Circle, Suite 5208, Atlanta, GA 30322, United States.
| |
Collapse
|
46
|
El-Zaatari M, Bishu S, Zhang M, Grasberger H, Hou G, Haley H, Humphries B, Syu LJ, Dlugosz AA, Luker K, Luker GD, Eaton K, Kamada N, Cascalho M, Kao JY. Aim2-mediated/IFN-β-independent regulation of gastric metaplastic lesions via CD8+ T cells. JCI Insight 2020; 5:94035. [PMID: 32053518 DOI: 10.1172/jci.insight.94035] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 02/12/2020] [Indexed: 01/24/2023] Open
Abstract
Development of gastric cancer is often preceded by chronic inflammation, but the immune cellular mechanisms underlying this process are unclear. Here we demonstrated that an inflammasome molecule, absent in melanoma 2 (Aim2), was upregulated in patients with gastric cancer and in spasmolytic polypeptide-expressing metaplasia of chronically Helicobacter felis-infected stomachs in mice. However, we found that Aim2 was not necessary for inflammasome function during gastritis. In contrast, Aim2 deficiency led to an increase in gastric CD8+ T cell frequency, which exacerbated metaplasia. These gastric CD8+ T cells from Aim2-/- mice were found to have lost their homing receptor expression (sphingosine-1-phosphate receptor 1 [S1PR1] and CD62L), a feature of tissue-resident memory T cells. The process was not mediated by Aim2-dependent regulation of IFN-β or by dendritic cell-intrinsic Aim2. Rather, Aim2 deficiency contributed to an increased production of CXCL16 by B cells, which could suppress S1PR1 and CD62L in CD8+ T cells. This study describes a potentially novel function of Aim2 that regulates CD8+ T cell infiltration and retention within chronically inflamed solid organ tissue. This function operates independent of the inflammasome, IFN-β, or dendritic cells. We provide evidence that B cells can contribute to this mechanism via CXCL16.
Collapse
Affiliation(s)
- Mohamad El-Zaatari
- Division of Gastroenterology and Hepatology, Department of Internal Medicine
| | - Shrinivas Bishu
- Division of Gastroenterology and Hepatology, Department of Internal Medicine
| | - Min Zhang
- Division of Gastroenterology and Hepatology, Department of Internal Medicine
| | - Helmut Grasberger
- Division of Gastroenterology and Hepatology, Department of Internal Medicine
| | - Guoqing Hou
- Division of Gastroenterology and Hepatology, Department of Internal Medicine
| | - Henry Haley
- Department of Radiology, Microbiology and Immunology, and Biomedical Engineering
| | - Brock Humphries
- Department of Radiology, Microbiology and Immunology, and Biomedical Engineering
| | - Li-Jyun Syu
- Department of Dermatology, School of Medicine
| | | | - Kathy Luker
- Department of Radiology, Microbiology and Immunology, and Biomedical Engineering
| | - Gary D Luker
- Department of Radiology, Microbiology and Immunology, and Biomedical Engineering
| | | | - Nobuhiko Kamada
- Division of Gastroenterology and Hepatology, Department of Internal Medicine
| | - Marilia Cascalho
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA
| | - John Y Kao
- Division of Gastroenterology and Hepatology, Department of Internal Medicine
| |
Collapse
|
47
|
Pizzolla A, Wakim LM. Memory T Cell Dynamics in the Lung during Influenza Virus Infection. THE JOURNAL OF IMMUNOLOGY 2019; 202:374-381. [PMID: 30617119 DOI: 10.4049/jimmunol.1800979] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 09/08/2018] [Indexed: 01/06/2023]
Abstract
Influenza A virus is highly contagious, infecting 5-15% of the global population every year. It causes significant morbidity and mortality, particularly among immunocompromised and at-risk individuals. Influenza virus is constantly evolving, undergoing continuous, rapid, and unpredictable mutation, giving rise to novel viruses that can escape the humoral immunity generated by current influenza virus vaccines. Growing evidence indicates that influenza-specific T cells resident along the respiratory tract are highly effective at providing potent and rapid protection against this inhaled pathogen. As these T cells recognize fragments of the virus that are highly conserved and less prone to mutation, they have the potential to provide cross-strain protection against a wide breadth of influenza viruses, including newly emerging strains. In this review, we will discuss how influenza-specific memory T cells in the lung are established and maintained and how we can harness this knowledge to design broadly protective influenza A virus vaccines.
Collapse
Affiliation(s)
- Angela Pizzolla
- Department of Microbiology and Immunology, University of Melbourne, at Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| | - Linda M Wakim
- Department of Microbiology and Immunology, University of Melbourne, at Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| |
Collapse
|
48
|
Knight FC, Gilchuk P, Kumar A, Becker KW, Sevimli S, Jacobson ME, Suryadevara N, Wang-Bishop L, Boyd KL, Crowe JE, Joyce S, Wilson JT. Mucosal Immunization with a pH-Responsive Nanoparticle Vaccine Induces Protective CD8 + Lung-Resident Memory T Cells. ACS NANO 2019; 13:10939-10960. [PMID: 31553872 PMCID: PMC6832804 DOI: 10.1021/acsnano.9b00326] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Tissue-resident memory T cells (TRM) patrol nonlymphoid organs and provide superior protection against pathogens that commonly infect mucosal and barrier tissues, such as the lungs, intestine, liver, and skin. Thus, there is a need for vaccine technologies that can induce a robust, protective TRM response in these tissues. Nanoparticle (NP) vaccines offer important advantages over conventional vaccines; however, there has been minimal investigation into the design of NP-based vaccines for eliciting TRM responses. Here, we describe a pH-responsive polymeric nanoparticle vaccine for generating antigen-specific CD8+ TRM cells in the lungs. With a single intranasal dose, the NP vaccine elicited airway- and lung-resident CD8+ TRM cells and protected against respiratory virus challenge in both sublethal (vaccinia) and lethal (influenza) infection models for up to 9 weeks after immunization. In elucidating the contribution of material properties to the resulting TRM response, we found that the pH-responsive activity of the carrier was important, as a structurally analogous non-pH-responsive control carrier elicited significantly fewer lung-resident CD8+ T cells. We also demonstrated that dual-delivery of protein antigen and nucleic acid adjuvant on the same NP substantially enhanced the magnitude, functionality, and longevity of the antigen-specific CD8+ TRM response in the lungs. Compared to administration of soluble antigen and adjuvant, the NP also mediated retention of vaccine cargo in pulmonary antigen-presenting cells (APCs), enhanced APC activation, and increased production of TRM-related cytokines. Overall, these data suggest a promising vaccine platform technology for rapid generation of protective CD8+ TRM cells in the lungs.
Collapse
Affiliation(s)
- Frances C. Knight
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Pavlo Gilchuk
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN 37212, USA
| | - Amrendra Kumar
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN 37212, USA
| | - Kyle W. Becker
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Sema Sevimli
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Max E. Jacobson
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Naveenchandra Suryadevara
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN 37212, USA
| | - Lihong Wang-Bishop
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Kelli L. Boyd
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - James E. Crowe
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Chemical and Physical Biology Program, Vanderbilt University, Nashville, TN 37235, USA
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37235, USA
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Sebastian Joyce
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN 37212, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - John T. Wilson
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN 37235, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Corresponding Author:
| |
Collapse
|
49
|
Topham DJ, Reilly EC, Emo KL, Sportiello M. Formation and Maintenance of Tissue Resident Memory CD8+ T Cells after Viral Infection. Pathogens 2019; 8:E196. [PMID: 31635290 PMCID: PMC6963622 DOI: 10.3390/pathogens8040196] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 10/15/2019] [Accepted: 10/16/2019] [Indexed: 01/17/2023] Open
Abstract
Tissue resident memory (TRM) CD8 T cells comprise a memory population that forms in peripheral, non-lymphoid tissues after an infection that does not recirculate into the bloodstream or other tissues. TRM cells often recognize conserved peptide epitopes shared among different strains of a pathogen and so offer a protective role upon secondary encounter with the same or related pathogens. Several recent studies have begun to shed light on the intrinsic and extrinsic factors regulating TRM. In addition, work is being done to understand how canonical "markers" of TRM actually affect the function of these cells. Many of these markers regulate the generation or persistence of these TRM cells, an important point of study due to the differences in persistence of TRM between tissues, which may impact future vaccine development to cater towards these important differences. In this review, we will discuss recent advances in TRM biology that may lead to strategies designed to promote this important protective immune subset.
Collapse
Affiliation(s)
- David J Topham
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA.
| | - Emma C Reilly
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA.
| | - Kris Lambert Emo
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA.
| | - Mike Sportiello
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA.
| |
Collapse
|
50
|
CD103 hi T reg cells constrain lung fibrosis induced by CD103 lo tissue-resident pathogenic CD4 T cells. Nat Immunol 2019; 20:1469-1480. [PMID: 31591568 DOI: 10.1038/s41590-019-0494-y] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 08/12/2019] [Indexed: 01/05/2023]
Abstract
Tissue-resident memory T cells (TRM cells) are crucial mediators of adaptive immunity in nonlymphoid tissues. However, the functional heterogeneity and pathogenic roles of CD4+ TRM cells that reside within chronic inflammatory lesions remain unknown. We found that CD69hiCD103lo CD4+ TRM cells produced effector cytokines and promoted the inflammation and fibrotic responses induced by chronic exposure to Aspergillus fumigatus. Simultaneously, immunosuppressive CD69hiCD103hiFoxp3+ CD4+ regulatory T cells were induced and constrained the ability of pathogenic CD103lo TRM cells to cause fibrosis. Thus, lung tissue-resident CD4+ T cells play crucial roles in the pathology of chronic lung inflammation, and CD103 expression defines pathogenic effector and immunosuppressive tissue-resident cell subpopulations in the inflamed lung.
Collapse
|